Inflammatory stress signaling via NF-kB alters accessible cholesterol to upregulate SREBP2 transcriptional activity in endothelial cells

  1. Joseph Wayne M Fowler
  2. Rong Zhang
  3. Bo Tao
  4. Nabil E Boutagy
  5. William C Sessa  Is a corresponding author
  1. Vascular Biology and Therapeutics Program, Department of Pharmacology, Yale University School of Medicine, United States
7 figures, 1 table and 2 additional files

Figures

Figure 1 with 1 supplement
TNFα and NF-κB control SREBP2-dependent gene expression in human endothelial cells.

Primary HUVEC were treated siRNA against non-targeting sequence (siCTRL) or RELA for 48 hr and then incubated with or without 10 ng/mL TNFα for 10 hr. (a) Volcano plot for RNA-seq analysis of differentially expressed genes. Dotted red lines indicate cutoff used for IPA analysis (p<0.05, 1.5<Fold Change (F.C)<−1.5). (b) IPA analysis of most significant canonical pathways and predicted upstream transcriptional regulators for genes that increase at 10 hr TNFα. (c) IPA analysis of most significant canonical pathways and predicted upstream transcriptional regulators for genes that decrease in cells knocked down with RELA siRNA and treated 10 hr TNFα compared to control cells treated with 10 hr TNFα. (d) Representative heatmap of NF-κB and SREBP2 transcriptionally controlled genes from (b) and (c) showing three independent donors.

Figure 1—figure supplement 1
Complete transcriptomic pathway analysis in HUVEC treated with TNFa for 0, 4, and 10 hr and with or with RELA siRNA.

(a) Ingenuity pathway analysis for pathways and upstream transcription regulators using differentially expressed genes (upregulated) in HUVEC after 4 hr TNFα treatment (F.C.>1.5; p<0.05). (b) Metacore metabolic network analysis using upregulated genes from (Figure 1a) (p<0.005). (c) GSEA hallmark analysis using upregulated genes from (Figure 1a). (d) Ingenuity pathway analysis of gene set overlap between significantly upregulated genes in 10 hr TNFα compared to 0 hr TNFα and significantly downregulated genes after 10 hr TNFα and in siRELA compared to siCTRL.

Figure 2 with 1 supplement
TNFα increases SREBP2 cleavage and transcription of canonical sterol-responsive genes.

(a) SREBP2 immunoblot from whole-cell lysates from HUVEC treated with TNFα (10 ng/mL) for indicated time. Data are normalized to respective GAPDH and then to untreated cells (n=3). (b) qRT-PCR analysis of RNA from HUVEC treated with TNFα (10 ng/mL) for indicated time. Data are normalized to respective ACTB and then to untreated cells (n=8). (c) LDLR protein levels of TNFα-treated HUVEC treated with or without native LDL (25 μg/mL). Data are normalized to respective HSP90 levels and then to untreated cells (n=4). (d) Flow cytometry analysis of exogenous DiI-LDL uptake in HUVEC treated with TNFα and with indicated media. 2.5 μg/mL DiI-LDL was incubated for 1 hr at 37 °C before processing for flow cytometry. Uptake was quantified by PE mean fluorescence intensity per cell and normalized to untreated cells in LPDS across two experiments (10,000 events/replicate, n=4). *p<0.05; **p<0.01; ***p<0.001; ***p<0.0001 by one-way ANOVA with Dunnett’s multiple comparisons test (a and b) or two-way ANOVA with Sidak’s multiple comparisons test (c and d).

Figure 2—figure supplement 1
TNFa predominantly activates targets involved in cholesterol biosynthesis, not fatty acid synthesis.

(a) Representative SREBP2 immunoblot from whole-cell lysates from HUVEC treated with TNFα (16 hr) at indicated dose. Cells were incubated with fetal bovine serum (FBS) or lipoprotein depleted serum (LPDS). (b) Heatmap of classical SREBP1-dependent fatty acid synthesis genes from previous RNA-seq analysis. (c) Representative HMGCR immunoblot of HUVEC treated with TNFα (10 ng/mL) for indicated time and media.

RELA DNA-binding is necessary for activation of SREBP2 by inflammatory stress.

(a) Representative immunoblot of SREBP2 and LDLR protein levels in HUVEC treated with TNFα (10 ng/mL), IL1β (10 ng/mL), or LPS (100 ng/mL). (b) Representative immunoblot of SREBP2 and LDLR protein levels in HUVEC treated with actinomycin D (ActD, 10 ng/mL) and with or without TNFα (10 ng/mL). (c) Quantification of SREBP2 precursor (p) and cleaved (c) from (b). Data are normalized to respective HSP90 and then to untreated cells (n=4). (d) SREBP2 and LDLR protein levels in HUVEC treated with IL1β (10 ng/mL) or TNFα (10 ng/mL) and with or without NF-κB inhibitor, BAY11-7082 (5 μM). Data are normalized to respective HSP90 and then to untreated cells (n=3). (e) qRT-PCR analysis of SREBP2-dependent genes, SREBF2, LDLR, HMGCS1, HMGCR, and INSIG1, expression in HUVEC treated with or without TNFα (10 ng/mL) and BAY11-7082 (5 μM). Data are normalized to respective ACTB and then to untreated cells (n=6). (f) SREBP2 and RELA levels in TNFα (10 ng/mL)-treated HUVEC treated with or without siRNA targeting RELA. Data are normalized to respective HSP90 and then to untreated cells (n=4). *p<0.05; **p<0.01; ***p<0.001; ***P<0.0001 by one-way ANOVA (c, d, and f) or two-way ANOVA (e) with Tukey’s multiple comparison’s test.

Figure 4 with 1 supplement
Cytokine-mediated upregulation of SREBP2 cleavage requires proper SCAP shuttling and proteolytic processing in the Golgi.

(a) Schematic of where 25-hyroxycholesterol (25HC), cholesterol, siSCAP, and PF-429242 inhibit SREBP processing throughout the pathway. (b) Representative immunoblot of SREBP2 and LDLR protein levels in HUVEC treated with TNFα (10 ng/mL) and cholesterol (Chol) (25 μg/mL). Data are normalized to respective HSP90 and then to untreated cells. (c) Representative immunoblot of SREBP2 and LDLR protein levels in HUVEC treated with TNFα (10 ng/mL) and increasing concentrations of LDL. Data are normalized to respective HSP90 and then to untreated cells. (d) Representative immunoblot SREBP2 cleavage in HUVEC treated with IL1β (10 ng/mL) or TNFα (10 ng/mL) and SCAP siRNA. Data are normalized to respective HSP90 and then to untreated cells. (e) Representative immunoblot of SREBP2 and LDLR protein levels in HUVEC treated with TNFα (10 ng/mL) and increasing concentrations of 25-hydroxycholesterol (25HC). Data are normalized to respective HSP90 and then to untreated cells. (f) Representative immunoblot of SREBP2 and LDLR protein levels in HUVEC treated with TNFα (10 ng/mL) and PF-429242 (10 μM) for indicated time. Data are normalized to respective HSP90 and then to untreated cells. (g) qRT-PCR analysis of SREBF2, HMGCS1, and SCAP from RNA of HUVECs treated with TNFα (10 ng/mL) and indicated SREBP2 inhibitor. Data are normalized to respective ACTB and then to untreated cells (n=6). *p<0.05; **p<0.01; ***p<0.001; ***p<0.0001 by two-way ANOVA with Sidak’s multiple comparisons test.

Figure 4—figure supplement 1
Immunoblots of SREBP2 processing inhibitors at effective doses.

(a) SREBP2 and LDLR protein levels in HUVEC treated with TNFα (10 ng/mL) and with or without low density lipoprotein (LDL) (250 μg/mL). Data are normalized to respective HSP90 and then to untreated cells (n=4). (b) Representative immunoblot of SREBP2 and LDLR protein levels in HUVEC treated with TNFα (10 ng/mL) and increasing concentrations of fatostatin. (c) SREBP2 and LDLR protein levels in HUVEC treated with TNFα (10 ng/mL) and with or without 25-hydroxycholesterol (25HC) (10 μM). Data are normalized to respective HSP90 and then to untreated cells (n=4). (d) SREBP2 and LDLR protein levels in HUVEC treated with TNFα (10 ng/mL) and with or without PF-429242 (10 μM). Data are normalized to respective HSP90 and then to untreated cells (n=4). (e) Quantification of SREBP2 and LDLR protein levels in HUVEC treated with TNFα (10 ng/mL) and with or without MβCD-cholesterol (Chol) (65 μM). Data are normalized to respective HSP90 and then to untreated cells (n=3). (f) Quantification of SREBP2 protein levels in HUVEC treated with TNFα (10 ng/mL) and with or without siSCAP. Data are normalized to respective HSP90 and then to untreated cells (n=4). *p<0.05; **p<0.01; ***p<0.001; ***p<0.0001 by two-way ANOVA with Sidak’s multiple comparisons test.

Figure 5 with 1 supplement
TNFα decreases accessible cholesterol in cultured HUVEC and mouse lung ECs in vivo.

(a) Quantification of total cholesterol extracted from HUVEC treated with or without TNFα (10 ng/mL) and indicated positive controls, lipoprotein deficient serum (LPDS), fetal bovine serum (FBS), or MβCD-cholesterol. Data were normalized to respective total protein (n=3). (b) Total cholesterol in HUVEC after 4 or 10 hr of TNFα (10 ng/mL) quantified by mass spectrometry (n=3). (c) ALOD4 protein levels in HUVEC treated with TNFα (10 ng/mL). Data are normalized to respective HSP90 and then to untreated cells (n=7). (d) In-cell western blot of ALOD4 protein levels in HUVEC treated with TNFα (10 ng/mL) and PF-429242 (10 μM). Data are normalized to respective total protein and then to untreated cells (n=6). (e) In-cell western blot of ALOD4 protein levels in HUVEC treated with TNFα (10 ng/mL) and PF-429242 (10 μM) for indicated time. Data are normalized to respective total protein and then to untreated cells (n=6). (f) ALOD4 protein levels in TNFα (10 ng/mL)-treated HUVEC treated with or without RELA siRNA. Data are normalized to respective HSP90 and then to untreated cells (n=8). (g) Schematic of protocol to isolate mouse lung endothelial cells and quantify ALOD4 binding by flow cytometry. (h) Representative histogram of ALOD4 binding in Cd31 +lung endothelial cells in mice treated with or without LPS (15 mg/kg) for 6 hr. (i) Quantification of ALOD4 binding across 2 flow cytometry experiments in mice treated with or without LPS (15 mg/kg). Binding was quantified as AlexaFluor647 mean fluorescent intensity per cell (100,000 events/replicate). Data are normalized to nontreated mice (-LPS, n=6;+LPS, n=6). *p<0.05; **p<0.01; ***p<0.001; ***p<0.0001 by one-way ANOVA with Tukey’s multiple comparison’s test (a and d) or Dunnett’s multiple comparisons test (e), unpaired t-test (c and i), or two-way ANOVA with Sidak’s multiple comparisons test (f).

Figure 5—figure supplement 1
Optimization of assays used to quantify accessible cholesterol on cellular plasma membranes.

(a) Diagram of pipeline for immunoblotting protocol to quantify EC accessible cholesterol (top). Representative immunoblot of HIS (ALOD4) after treatment with cholesterol modifying agents: MβCD-cholesterol (Chol) (25 μg/mL), LDL (100 μg/ml), or MβCD (1%) (bottom). (b) Diagram of pipeline for in-cell Western blotting protocol to quantify EC accessible cholesterol (top). Representative in-cell western blot of secondary alone (α-HIS-647) or HIS (ALOD4) after treatment with cholesterol modifying agents: MβCD-cholesterol (Chol) (25 μg/mL), LDL (100 μg/ml), or MβCD (1%) (bottom). (c) Representative SDS-PAGE gel of purified unconjugated ALOD4 and fluorescent ALOD4-647 stained with Coomassie (left) or recorded with the 700 nm channel on LICOR Biosciences Odyssey CLx platform. (d) Schematic of flow cytometry pipeline to quantify ALOD4 binding in cultured ECs with ALOD4-647. (e) Flow cytometry analysis of bound ALOD4-647 per HUVEC after treatment with positive controls, lipoprotein depleted serum (LPDS), fetal bovine serum (FBS), LDL (100 μg/mL), MβCD-cholesterol (Chol) (25 μg/mL), or MβCD (1%). ALOD4 binding was quantified by mean fluorescence intensity of AlexaFluor647 channel (10,000 events/replicate, n=3). (f) Flow cytometry analysis of ALOD4-647 bound to HUVEC treated with TNFα (10 ng/mL) for 16 hr. ALOD4 binding was quantified by mean fluorescence intensity of AlexaFluor647 channel (10,000 events/replicate, n=3). (g) Circulating TNFα from serum of mice treated with LPS (15 mg/kg) for 2 or 6 hr (n=6). (h) Total cholesterol from serum of mice used in (Figure 5g) (n=6). *p<0.05; **p<0.01; ***p<0.001; ***p<0.0001 by unpaired t-test (f and g) or one-way ANOVA with Tukey’s multiple comparison’s test.

Figure 6 with 3 supplements
STARD10 is necessary for complete TNFα-mediated accessible cholesterol reduction and SREBP2 activation.

(a) Heatmap of genes that regulate lipid homeostasis, significantly increased with TNFα (10 ng/mL) treatment after 4 or 10 hr, and were significantly inhibited by RELA knockdown. (b) STARD10 gene locus from RELA ChIP-seq analysis of human aortic endothelial cells (HAEC) treated with TNFα (2 ng/mL) or IL1β (10 ng/mL) for 4 hr. Data are scaled from 0 (bottom) to 5 (top). Data originated from GSE89970. (c) Immunoblot of STARD10 protein levels in HUVEC treated with RELA or STARD10 (S10) siRNA and with or without TNFα (10 ng/mL). Data are normalized to respective HSP90 levels and then to untreated cells (n=4). (d) qRT-PCR analysis of RNA from HUVEC treated with TNFα (10 ng/mL) and two independent siRNA targeting STARD10 (#1,#2). Data are normalized to respective ACTB and then to untreated cells (n=5). (e) Immunoblot of ALOD4, SREBP2, and LDLR protein levels in HUVEC treated with STARD10 siRNA (siS10) and with or without TNFα (10 ng/mL). Data are normalized to respective HSP90 levels and then to untreated cells (n=3). *p<0.05; **p<0.01; ***p<0.001; ***p<0.0001 by two-way ANOVA with Sidak’s multiple comparisons test (d and e).

Figure 6—figure supplement 1
Complete analysis of common cholesterol transport mechanisms in HUVEC under inflammatory stress.

(a) Schematic of possible mechanisms to deplete plasma membrane accessible cholesterol: (1) efflux, (2) sphingomyelin shielding, (3) esterification, and (4) lysosomal/endosomal accumulation. (b) Total sphingomyelin (SM) and cholesteryl ester (CE) content in HUVEC after 4 or 10 hr of TNFα (10 ng/mL) quantified by mass spectrometry (n=3). (c) Thin layer chromatography of 3H-cholesterol isolated from HUVEC treated with oleic acid (OA) (0.5 mM), Sandoz 58–035 (ACATi) (1 μM), or TNFα (10 ng/mL) for 16 hr. Esterification was quantified as a ratio between cholesteryl ester (CE) and free cholesterol (FC) (n=4). (d) Representative immunoblot of OlyA, ALOD4, SREBP2, and LDLR protein levels in HUVEC treated with TNFα (10 ng/mL) and sphingomyelinase (SMase) (100mU/mL). (e) Schematic of protocol for measurement of cholesterol efflux (top). 3H-cholesterol efflux in HUVEC treated with T0901317 (T090) (5 μM) or TNFα (10 ng/mL) and with indicated acceptors, BSA, HDL, lipoprotein depleted serum (LPDS), or fetal bovine serum (FBS). Efflux was quantified as the ratio of 3H-cholesterol in the media compared to lysates (n=4). (f) Immunoblot of ALOD4, SREBP2, and LDLR protein levels in HUVEC treated with U18666A (U186) (5 μM) or choloroquine (CQ) (10 μM) and with or without TNFα (10 ng/mL). Data are normalized to respective HSP90 and then to untreated cells (n=3). (g) Representative images of Filipin and FITC-ulex eruopaeus agglutinin I (UEAI) stained HUVEC after treatment with TNFα (10 ng/mL) or U18666A (U186) (5 μM). White scale bar = 30 μm. *p<0.05; **p<0.01; ***p<0.001; ***p<0.0001 by one-way ANOVA with Tukey’s multiple comparison’s test (c and e).

Figure 6—figure supplement 2
RELA ChIP-seq analysis of lipid mediator genes.

RELA ChiP-seq analysis of lipid mediator genes of human aortic endothelial cells (HAEC) treated with TNFα (2 ng/mL) or IL1β (10 ng/mL) for 4 hr. Data are scaled from 0 (bottom) to 5 (top). VCAM1 is shown in first panel as a positive control. Data originated from GSE89970.

Figure 6—figure supplement 3
ABCG1 is significantly upregulated by TNFa, but is not responsible for accessible cholesterol depletion or SREBP2 activation.

(a) Representative immunoblot of ABCG1 and ABCA1 protein levels in HUVEC treated with TNFα (10 ng/mL), T0901317 (T090) (5 μM), and/or BAY11-7082 (5 μM). (b) Representative immunoblot of ABCG1, ALOD4, SREBP2, and LDLR protein levels in HUVEC treated with TNFα (10 ng/mL) and two independent siRNA targeting ABCG1 (#1,#2). (c) qRT-PCR analysis of RNA from HUVEC treated with TNFα (10 ng/mL) and two independent siRNA targeting ABCG1 (#1,#2). Data are normalized to respective ACTB and then to untreated cells (n=2). *p<0.05; **p<0.01; ***p<0.001; ***p<0.0001 by two-way ANOVA with Sidak’s multiple comparisons test.

Working model of the relationship between sterol sensing and EC acute inflammatory response.

Pro-inflammatory cytokines, such as TNFα and IL1β, promote NF-κB activation of gene transcription in endothelial cells. NF-κB upregulates factors, such as STARD10, that significantly decrease accessible cholesterol on the plasma membrane. SCAP senses the reduction in accessible cholesterol and shuttles SREBP2 to the Golgi to initiate classical proteolytic processing. Active N-SREBP2 translocates to the nucleus to transcriptionally upregulate canonical cholesterol biosynthetic genes.

Tables

Appendix 1—key resources table
Reagent type (species) or resourceDesignationSource or referenceIdentifiersAdditional information
antibodyAnti-His Tag Dylight 680 (Mouse monoclonal)ThermoFisher ScientificMA1-21315-D680In-cell Western (1:1,000)
antibodyAnti-6x His (Mouse monoclonal)abcamab18184WB (1:1,000)
antibodyAnti-ABCG1 (Rabbit monoclonal)abcamab52617WB (1:1,000)
antibodyAnti-mmCd31-BV605 (Rat monoclonal)BioLegend102427FACS (1:200)
antibodyAnti-GAPDH (Rabbit monoclonal)Cell Signalling2118 SWB (1:2,000)
antibodyAnti-HSP90 (Mouse monoclonal)Santa Cruzsc-13119WB (1:2,000)
antibodyAnti-ICAM1 (Rabbit monoclonal)Cell Signaling4915 SWB (1:1,000)
antibodyAnti-JNK1 (Mouse monoclonal)Cell Signaling3708 SWB (1:1,000)
antibodyAnti-LC3b (Rabbit monoclonal)Cell Signaling2775 SWB (1:1,000)
antibodyAnti-LDLR (Rabbit monoclonal)abcamab52818WB (1:1,000)
antibodyAnti-p-JNK1 (Rabbit monoclonal)Cell Signaling9261 SWB (1:1,000)
antibodyAnti-p-p38 (Rabbit monoclonal)Cell Signaling9211 SWB (1:1,000)
antibodyAnti-p38 (Rabbit monoclonal)Cell Signaling9212 SWB (1:1,000)
antibodyAnti-P65/RELA (Rabbit monoclonal)Cell Signaling8242 SWB (1:1,000)
antibodyAnti-SREBP1a (Mouse monoclonal)Santa Cruzsc-13551WB (1:1,000)
antibodyAnti-SREBP2 (Mouse monoclonal)BD Biosciences557037WB (1:1,000)
antibodyAnti-STARD10 (Rabbit polyclonal)Thermofisher ScientificPA5-36947WB (1:1,000)
antibodyAnti-VCAM1 (Mouse monoclonal)Santa Cruzsc-13160WB (1:1,000)
sequence-based reagentsiRNA: RELAThermofisher Scientifics11914Silencer Select
sequence-based reagentsiRNA: SREBF2Thermofisher Scientifics27Silencer Select
sequence-based reagentsiRNA: HMGCRThermofisher Scientific110740Silencer
sequence-based reagentsiRNA: SCAPThermofisher Scientifics695Silencer Select
sequence-based reagentsiRNA: STARD10 #1Thermofisher Scientifics21244Silencer Select
sequence-based reagentsiRNA: STARD10 #2Thermofisher Scientifics21243Silencer Select
sequence-based reagentsiRNA: ABCG1 #1Thermofisher Scientifics18482Silencer Select
sequence-based reagentsiRNA: ABCG1 #2Thermofisher ScientificS18484Silencer Select
sequence-based reagenthsACTB_FThis PaperqRT-PCR PrimersAGCACTGTGTTGGCGTACAG
sequence-based reagenthsACTB_RThis PaperqRT-PCR PrimersGGACTTCGAGCAAGAGATGG
sequence-based reagenthsLDLR_FThis PaperqRT-PCR PrimersTCTGCAACATGGCTAGAGACT
sequence-based reagenthsLDLR_RThis PaperqRT-PCR PrimersTCCAAGCATTCGTTGGTCCC
sequence-based reagenthsHMGCS1_FThis PaperqRT-PCR PrimersCAAAAAGATCCATGCCCAGT
sequence-based reagenthsHMGCS1_RThis PaperqRT-PCR PrimersAAAGGCTTCCAGGCCACTAT
sequence-based reagenthsHMGCR_FThis PaperqRT-PCR PrimersTGATTGACCTTTCCAGAGCAAG
sequence-based reagenthsINSIG1_FThis PaperqRT-PCR PrimersCTAAAATTGCCATTCCACGAGC
sequence-based reagenthsINSIG1_RThis PaperqRT-PCR PrimersGCACTGCATTAAACGTGTGG
sequence-based reagenthsSREBF2_FThis PaperqRT-PCR PrimersTAAAGGAGAGGCACAGGA
sequence-based reagenthsSREBF2_RThis PaperqRT-PCR PrimersAGGAGAACATGGTGCTGA
sequence-based reagenthsICAM1_FThis PaperqRT-PCR PrimersGTGGTAGCAGCCGCAGTC
sequence-based reagenthsICAM1_RThis PaperqRT-PCR PrimersGGCTTGTGTGTTCGGTTTCA
sequence-based reagenthsCXCL1_FThis PaperqRT-PCR PrimersAGGGAATTCACCCCAAGAAC
sequence-based reagenthsCXCL1_RThis PaperqRT-PCR PrimersTGGATTTGTCACTGTTCAGCA
sequence-based reagenthsSELE_FThis PaperqRT-PCR PrimersACCTCCACGGAAGCTATGACT
sequence-based reagenthsSELE_RThis PaperqRT-PCR PrimersCAGACCCACACATTGTTGACTT
sequence-based reagenthsSCAP_FThis PaperqRT-PCR PrimersCGCAAACAAGGAGAGCCTAC
sequence-based reagenthsSCAP_RThis PaperqRT-PCR PrimersTGTCTCTCAGCACGTGGTTC
sequence-based reagenthsSTARD10_FThis PaperqRT-PCR PrimersGAAAGACTTGGTCCGAGCTG
sequence-based reagenthsSTARD10_RThis PaperqRT-PCR PrimersTTCCACTCGGGGTACTTGAG
chemical compound, drug25-hydroxycholesterolSigma AldrichH1015
chemical compound, drugActinomycin DThermoFisher11805017
chemical compound, drugBAY 117082Sigma AldrichB556-10MG
chemical compound, drugCholesterol, 1,2-3H(N)Perkin ElmerNET139250UC
chemical compound, drugChoroquine (CQ)Sigma AldrichC6628
chemical compound, drugDiI LDLKalen Biomedical770230
chemical compound, drugEGM2LonzaCC-3162
chemical compound, drugFatostatinCayman13562
chemical compound, drugFilipinCayman70440
chemical compound, drugFITC-UEAIThermoFisherL32476
chemical compound, drugLipopolysaccharide from E. coli O111:B4Sigma AldrichL2630
chemical compound, drugLipoprotein Depleted Serum (LPDS)Kalen Biomedical880100
chemical compound, drugMβCDSigma AldrichC4555
chemical compound, drugMβCD-CholesterolSigma AldrichC4951
chemical compound, drugNative LDLKalen Biomedical770200
chemical compound, drugPF-429242Sigma AldrichSML0667
chemical compound, drugrhIL1βRD Systems201-LB-010/CF
chemical compound, drugrhTNFαRD Systems210-TA-020/CF
chemical compound, drugSandoz 58–035Sigma AldrichS9318-25mg
chemical compound, drugSphingomyelinaseSigma AldrichS8633
chemical compound, drugT0901317Sigma AldrichT2320
chemical compound, drugTriacin CRD Systems2472
chemical compound, drugU18666ASigma AldrichU3633
commercial assay or kitCholesterol/Cholesteryl Ester Assay KitAbcamAb65359
software, algorithmPartek FlowPartekhttps://www.partek.com/partek-flow/
software, algorithmIngenuity Pathway AnalssisQiagenhttps://digitalinsights.qiagen.com/products-overview/discovery-insights-portfolio/analysis-and-visualization/qiagen-ipa/

Additional files

Supplementary file 1

RNA-seq normalized counts and lipidomics.

(RNA-seq) HUVEC were treated with TNFα for 0, 4, and 10 hr and with or without siRNA targeting RELA. (Lipidomics). HUVEC were treated with TNFα for 0, 4, and 10 hr. Data represented as molar percentage of lipid

https://cdn.elifesciences.org/articles/79529/elife-79529-supp1-v2.xlsx
MDAR checklist
https://cdn.elifesciences.org/articles/79529/elife-79529-mdarchecklist1-v2.pdf

Download links

A two-part list of links to download the article, or parts of the article, in various formats.

Downloads (link to download the article as PDF)

Open citations (links to open the citations from this article in various online reference manager services)

Cite this article (links to download the citations from this article in formats compatible with various reference manager tools)

  1. Joseph Wayne M Fowler
  2. Rong Zhang
  3. Bo Tao
  4. Nabil E Boutagy
  5. William C Sessa
(2022)
Inflammatory stress signaling via NF-kB alters accessible cholesterol to upregulate SREBP2 transcriptional activity in endothelial cells
eLife 11:e79529.
https://doi.org/10.7554/eLife.79529