Cell membrane phosphatidylcholine (PC) composition is regulated by lysophosphatidylcholine acyltransferase (LPCAT); changes in membrane PC saturation are implicated in metabolic disorders. Here, we identified LPCAT3 as the major isoform of LPCAT in adipose tissue and created adipocyte-specific Lpcat3–knockout mice to study adipose tissue lipid metabolism. Transcriptome sequencing and plasma adipokine profiling were used to investigate how LPCAT3 regulates adipose tissue insulin signaling. LPCAT3 deficiency reduced polyunsaturated PCs in adipocyte plasma membranes, increasing insulin sensitivity. LPCAT3 deficiency influenced membrane lipid rafts, which activated insulin receptors and AKT in adipose tissue, and attenuated diet-induced insulin resistance. Conversely, higher LPCAT3 activity in adipose tissue from ob/ob, db/db, and high-fat diet–fed mice reduced insulin signaling. Adding polyunsaturated PCs to mature human or mouse adipocytes in vitro worsened insulin signaling. We suggest that targeting LPCAT3 in adipose tissue to manipulate membrane phospholipid saturation is a new strategy to treat insulin resistance.

Article Highlights
  • Phosphatidylcholine (PC) remodeling has an impact on metabolic disorders, including diabetes, insulin resistance, and cardiovascular diseases.

  • We investigated the effect of lysophosphatidylcholine acyltransferase 3 (LPCAT3), the major PC remodeling enzyme in adipose tissue, on insulin sensitivity.

  • We found that blocking adipocyte PC remodeling in mice promotes systemic insulin sensitivity, primarily by reducing the amount of polyunsaturated PCs in adipocyte plasma membranes. This is also true for human adipocytes.

  • Our study suggests that targeting LPCAT3 in adipose tissue to manipulate membrane phospholipid saturation is a new strategy to treat insulin resistance.

Phospholipids are a major class of lipids on cell membranes (1) and in plasma lipoproteins (2). Polyunsaturated fatty acids are usually esterified at the sn-2 position, whereas saturated and monounsaturated fatty acids are usually esterified at the sn-1 position (3,4). Seventy percent of phospholipids are phosphatidylcholines (PCs). PCs are synthesized de novo by the Kennedy pathway (5). The asymmetrical distribution of fatty acids at the sn-1 and sn-2 positions is maintained in part by a deacylation–reacylation process first proposed by Lands 60 years ago (Lands cycle) (3,6). The deacylation step of this cycle is catalyzed by calcium-independent phospholipase A2, which removes saturated or monounsaturated fatty acids from the sn-2 position of PCs. The reacylation step of the cycle is catalyzed by lysophosphatidylcholine acyltransferase (LPCAT), which adds polyunsaturated free fatty acids at the sn-2 position of PCs.

PCs within the membranes of mammalian cells have considerable structural diversity (7). Polyunsaturated PCs have kinks that prevent the molecules from packing together, enhancing membrane fluidity. Alteration in cell membrane PC saturation has been implicated in a variety of metabolic disorders, including diabetes, obesity, and heart disease (8,9). Previous studies have demonstrated that membrane PC composition is regulated by LPCATs (10,11). Thus, LPCAT activities are important for maintaining cell membrane structure.

LPCAT has four isoforms (11), LPCAT1 to LPCAT4. LPCAT3 plays an important role in the function of the liver and small intestine (12,13), because it is the major LPCAT isoform expressed in those organs (14,15). We previously reported that systemic, intestine-specific, or liver-specific deletion of Lpcat3 in mice affects lipidemia, and the phenotypic variations are attributable to a reduced amount of polyunsaturated PCs on the plasma membranes of enterocytes and hepatocytes (14,15). LPCAT3 expression has been directly linked to the biosynthesis of inflammatory lipid mediators in humans (16). Notably, LPCAT3 expression was associated with a protective effect on endoplasmic reticulum stress in response to saturated fatty acids in vitro and in mouse models of metabolic diseases in which LPCAT3 expression was directly associated with a decrease in hepatic inflammation (17).

In this study, we identified LPCAT3 as the major isoform of LPCAT in adipose tissue. Adipose tissue is a metabolically active endocrine organ that plays a critical role in regulating systemic lipid and glucose homeostasis (18) and modulates insulin action by secreting adipokines (19). Although LPCAT3 function is well studied in various metabolic tissues, the effect of LPCAT3 in adipose tissue remains unknown.

In this study, we used a lipidomic analysis of white adipose tissue (WAT) from adipocyte-specific Lpcat3 (aLpcat3)–knockout (KO) mice and found that several species of polyunsaturated PCs were significantly reduced. With the guidance of RNA sequencing (RNA-seq) and adipokine profiling, we tested our hypothesis that LPCAT3-mediated PC remodeling affects insulin receptor (IR) phosphorylation by modulating adipocyte plasma membrane lipid organization and thus influencing insulin sensitivity and glucose tolerance. Here, we present evidence that genetic and diet-induced mouse models with high LPCAT3 activity can inhibit insulin signaling in adipose tissue. Specifically, supplementing mature adipocytes from humans or mice with polyunsaturated PCs led to insulin resistance in vitro. Our comprehensive mechanistic study supports our proposal that manipulating membrane phospholipid saturation by regulating LPCAT3 activity is a potential strategy to treat insulin resistance.

Animal Experiments

Lpcat3-Flox mice (14) were crossed with adiponectin-Cre transgenic mice (The Jackson Laboratory) to establish Lpcat3-Flox/adiponectin-Cre mice (Supplementary Fig. 1). All mice used had C57BL/6J genetic backgrounds, and both male and female mice were used in this study. Mice were fed a standard normal chow diet (cat. no. 5053; PicoLab Rodent Diet), a Western diet (cat. no. TD.88137; Envigo) for 8 weeks, or a high-fat/high-calorie (HFHC) diet (cat. no. D12331; Research Diets) for 16 weeks. All animal experiments were approved by the Institutional Animal Care and Use Committee of The State University of New York Downstate Health Sciences University.

Human Adipose Tissue Samples

Fresh human adipose tissue samples were obtained from a donor patient from the New Jersey Organ and Tissue Sharing Network who was screened to be free of diabetes and cancer, was not a cigarette or heavy alcohol user, and was not taking medications known to alter metabolism. Fresh WAT was used to isolate mature adipocytes.

Western Blot

For insulin signaling analysis by Western blot, mice were fasted overnight and then injected intraperitoneally with insulin (5 units/kg body weight; Sigma-Aldrich) or PBS as a positive control 30 min before sacrifice. Adipose tissue was immediately dissected and frozen separately using dry ice. Approximately 100 mg subcutaneous WAT (sWAT) was homogenized and lysed in radioimmunoprecipitation assay buffer with phosphatase inhibitors (Roche) and proteinase inhibitors (Roche). The following primary antibodies are summarized in Supplementary Table 1. The blotting membrane was stained with Ponceau S (Sigma-Aldrich) for total protein normalization.

RNA Extraction, Reverse Transcription, and Quantitative PCR

Total RNA was extracted from cells or tissue using TRIzol (Thermo Fisher Scientific), and cDNA was synthesized with a reverse transcription kit (Applied Biosystems). Quantitative PCR was performed using the SYBR Select Master Mix Kit (Applied Biosystems). Relative gene expression was normalized to 36B4 or Gapdh and calculated using the ΔΔCt method (Supplementary Research Design and Methods). Primer sequences are summarized in Supplementary Table 2.

PC Subspecies Measurement

As previously described, PC subspecies were measured using 200 mg sWAT by liquid chromatography–coupled tandem mass spectrometry (20).

LPCAT Activity Assay

LPCAT3 activity was measured in accordance with a published protocol (20) (Supplementary Research Design and Methods).

Glucose Tolerance Test and Insulin Tolerance Test

Mice were fasted overnight before intraperitoneal injection of glucose (2 g/kg body weight) for the glucose tolerance test or insulin (0.75 units/kg body weight) for the insulin tolerance test. Blood glucose was measured by tail vein bleeding and analyzed using a blood glucose monitoring system at 0, 15, 30, 60, and 120 min after injection.

Insulin Measurement in Plasma

Plasma insulin was quantified by insulin mouse ELISA kit (cat. no. EMINS; Invitrogen) in accordance with the manufacturer’s instructions (Supplementary Research Design and Methods).

Cell Culture and Differentiation

Mouse stromal vascular fractions (SVFs) were isolated from 5-week-old aLpcat3-KO mouse sWAT (21). Mouse SVFs or human white subcutaneous preadipocytes (cat. no. CSC-C4010×; Creative Bioarray) were differentiated into mature adipocytes as previously described (21) (Supplementary Research Design and Methods). Mature human adipocytes were isolated from fresh adipose tissue as previously described (22) (Supplementary Research Design and Methods). Mature adipocytes were cultured in maintenance medium containing DMEM, 0.5 μg/mL insulin, 10% FBS, and 1% penicillin-streptomycin.

Exogenous PC Supplementation In Vitro

For insulin sensitivity analysis, induced mature adipocytes or isolated mature human adipocytes were cultured in insulin-free serum-free DMEM and treated with 0, 10, 50, or 100 μmol/L polyunsaturated PCs including 18:0/20:4 PC (cat. no. 850469; Avanti) or 18:0/18:2 PC (cat. no. 850468; Avanti) for 24 h and then stimulated with 100 nmol/L insulin for 30 min. Cells were washed with cold PBS and prepared for immunoblotting.

Glucose Uptake In Vitro

PC-treated cells were stimulated with 100 nmol/L insulin for 30 min and then cultured with 1 mmol/L 2-deoxyglucose for 20 min. Next, we measured 2-deoxyglucose-6-phosphate using the bioluminescent Glucose Uptake-Glo Assay Kit (cat. no. J1341; Promega) in accordance with the manufacturer’s instructions. Luminescence was recorded with 0.3- to 1-s integration on a luminometer.

Adipokine Profile

Adipokines were measured in 70 μL pooled plasma samples from HFHC diet–fed aLpcat3-KO and wild-type (WT) mice (n = 6; male) using the Mouse Adipokine Array Kit (cat. no. ARY013; R&D Systems) in accordance with the manufacturer’s instructions (Supplementary Research Design and Methods).

Immunofluorescence Staining and Hematoxylin-Eosin Staining

Mouse sWAT was fixed in 4% formalin overnight at 4°C, and 10 μm paraffin sections were prepared. Each slice was deparaffinized and stained for immunofluorescence staining and hematoxylin-eosin staining. Primary antibodies are summarized in Supplementary Table 1. The rabbit monoclonal antibody immunoglobulin G isotype was used as a negative control. To minimize observer error, images were chosen and analyzed in a blind manner (Supplementary Research Design and Methods).

Adipose Tissue Lipid Raft Isolation

Adipose tissue lipid rafts were isolated as previously described (2325) (Supplementary Research Design and Methods).

RNA-Seq Data Processing and Differential Gene Expression Analysis

sWAT isolated from 2-month-old mice (n = 4 per group; male) was sent to Azenta Life Sciences for RNA extraction, library preparation, and standard RNA-seq. The raw data of sequence reads were processed using Trimmomatic (version 0.36) and further mapped to the Mus musculus GRCm38 reference genome (Ensembl [version 89]) using the STAR aligner (version 2.5.2b) (26). Each sample had at least 28,300,000 unique mapped reads. Unique gene hit counts were calculated using featureCounts from the Subread package (version 1.5.2). Gene hit counts were extracted and compared between each group using DESeq2 (version 1.30.1) (27). The Wald test was used to generate P values and log twofold changes. P values were adjusted using the Benjamini-Hochberg method. Genes with an adjusted P value of <0.05 and absolute fold change of >1.5 were considered differentially expressed genes for each comparison. Gene ontology functional enrichment analysis was performed using the PANTHER GO-slim biological process database (https://geneontology.org/) with default parameters (28,29). The dot enrichment plot was plotted by SRplot (https://www.bioinformatics.com.cn/srplot), an online platform for data analysis and visualization (adjusted P < 0.05).

Single-Cell RNA-Seq

The single-cell suspension from the sWAT SVF was sent to the Columbia Genome Center at Columbia University for library construction and further sequencing. The raw single-cell RNA-seq (scRNA-seq) data were processed using Cell Ranger (version 6.1.2; 10× Genomics). Filtered feature-barcode matrix files were obtained from the Cell Ranger pipeline, which contained only barcodes associated with cells. Additional analyses were performed using the R package Seurat (version 4.10). Cells with <300 genes and a total of fewer than three molecules detected within the cell were removed from the data, and gene counts were normalized and scaled using sctransform. We integrated our data from two conditions into a single analysis using an algorithm implemented in Seurat (30). We then applied a graph-based clustering algorithm to group cells into different clusters using the FindClusters function. We used cluster tree analysis (clustree function) to assess relatedness between clusters and annotate the clusters using known markers and data from relevant single-cell studies.

To identify genes that were differentially expressed between clusters, we compared the expression of each gene in each cluster with the same gene in all other clusters using a Wilcoxon rank sum test implemented in the FindAllMarkers function of Seurat. We set the min.pct argument to 0.25 (to include only genes with at least a 25% difference in mean expression) and the logfc.threshold argument to 0.25 (log fold change of at least 0.25 between the clusters).

Statistical Analyses

Each experiment was repeated at least three times. Statistical analyses were performed using GraphPad Prism software (version 8.0). Data between two groups were analyzed by the unpaired two-tailed Student t test. For multiple group comparisons, data were analyzed by one-way ANOVA followed by the Tukey post hoc multiple comparisons test. Data are presented as mean ± SEM. Statistical significance was set at P < 0.05. Standard symbols are presented as follows: P > 0.05, not significant; *P < 0.05; **P < 0.01; ***P < 0.001; and ****P < 0.0001.

Data and Resource Availability

RNA-seq data sets were submitted to the National Center for Biotechnology Information Gene Expression Omnibus database under the accession number GSE235304. All other data sets generated and/or analyzed in this study are available from the corresponding author upon request.

aLpcat3-KO Mice Have Reduced Polyunsaturated PCs in Adipose Tissue

LPCAT3 is the major LPCAT isoform expressed in WAT and brown adipose tissue (BAT) (Fig.1A). We crossed Lpcat3-Flox mice with adiponectin-Cre transgenic mice to generate aLpcat3 KO mice (Supplementary Fig. 1). There were no statistically significant differences in mouse body mass, phospholipid, triglyceride, plasma cholesterol, or apolipoprotein levels between WT control mice and aLpcat3-KO mice when mice were fed a normal chow diet (Supplementary Fig. 2AE). Expression levels of several genes involved in sWAT de novo lipogenesis, fatty acid oxidation, and lipolysis processes were unchanged between the two groups (Supplementary Fig. 2F). We measured total LPCAT activity in sWAT, epididymal WAT, and BAT from male aLpcat3-KO and WT mice. LPCAT activity was reduced by 65–70% in these adipose tissues compared with controls, whereas no changes were observed in the liver or skeletal muscle (Fig.1B and C). Similar results were obtained in female mice (data not shown). To evaluate the effect of LPCAT3 deficiency on PCs in adipose tissue, we measured PC subspecies using liquid chromatography–coupled tandem mass spectrometry. The concentrations of multiple sn-2 polyunsaturated PCs were significantly lower in aLpcat3-KO male mice compared with WT mice, including 16:0/20:4, 18:0/18:2, 18:0/20:3, 18:0/20:4, and 18:1/20:4 PC (Fig.1D). The concentrations of two major lysoPCs (16:0 and 18:0) were unchanged in aLpcat3-KO male mice compared with WT mice, whereas the concentrations of two minor lysoPCs (16:1 and 18:1) were greater in aLpcat3-KO male mice compared with WT mice (Fig.1E). The ratio of lysoPC to PC was elevated when LPCAT3 was absent (Fig.1F), suggesting that LPCAT3 can influence adipose tissue phospholipid saturation.

Figure 1

Characterization of aLpcat3-KO mice. A: Relative mRNA levels of LPCAT isoforms 1 to 4 in sWAT, epididymal WAT (eWAT), and BAT in aLpcat3-KO and WT mice. B: Total LPCAT activity in sWAT, eWAT, BAT, liver, and skeletal muscle from male aLpcat3-KO and WT mice. C: Measurement of LPCAT activity from panel B. D: Measurement of PC subspecies in aLpcat3-KO and WT mice. Lipids were extracted from sWAT from aLpcat3-KO and WT mice fed a normal chow diet. E: Quantification of lysoPC subspecies in aLpcat3-KO and WT mice. F: Ratio of lysoPC to PC in aLpcat3-KO and WT mice from data in panels D and E. Data are presented as mean ± SEM (n = 4 per group; male). Statistical significance was calculated by two-tailed Student t test. ns, not significant (P > 0.05). *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.

Figure 1

Characterization of aLpcat3-KO mice. A: Relative mRNA levels of LPCAT isoforms 1 to 4 in sWAT, epididymal WAT (eWAT), and BAT in aLpcat3-KO and WT mice. B: Total LPCAT activity in sWAT, eWAT, BAT, liver, and skeletal muscle from male aLpcat3-KO and WT mice. C: Measurement of LPCAT activity from panel B. D: Measurement of PC subspecies in aLpcat3-KO and WT mice. Lipids were extracted from sWAT from aLpcat3-KO and WT mice fed a normal chow diet. E: Quantification of lysoPC subspecies in aLpcat3-KO and WT mice. F: Ratio of lysoPC to PC in aLpcat3-KO and WT mice from data in panels D and E. Data are presented as mean ± SEM (n = 4 per group; male). Statistical significance was calculated by two-tailed Student t test. ns, not significant (P > 0.05). *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.

Close modal

Characterization of Adipocyte LPCAT3-Deficient Adipose Tissue

To investigate the role of LPCAT3 in adipose tissue, we performed RNA-seq of sWAT from mice fed a normal chow diet to assess the overall gene expression differences between aLpcat3-KO and WT mice. The absence of LPCAT3 in adipose tissue altered the expression of 594 genes (adjusted P < 0.05 and ∣FC∣ > 1.5); 414 were upregulated, and 180 were downregulated (Fig.2A). An overview of the top 30 up- and downregulated genes is given in Fig.2B. Notably, PANTHER GO-slim analysis revealed that lipid- and glucose metabolism–related biological processes were altered in aLpcat3-KO mice (Fig.2C), including response to insulin, the fatty acid catabolic process, and the glucose metabolic process. These results indicate that LPCAT3 may have a critical role in regulating insulin signaling.

Figure 2

Characterization of adipose tissue in aLpcat3-KO and WT mice. AC: Bulk RNA-seq analysis of sWAT from 8-week-old aLpcat3-KO and WT mice fed a normal chow diet (n = 4 per group; male). A: Volcano plot of differentially up- and downregulated genes in aLpcat3-KO and WT mice. B: Heat map of the 30 most significantly up- and downregulated genes in aLpcat3-KO and WT mice. C: PANTHER GO-slim analysis of gene ontology (GO) biological processes in aLpcat3-KO mouse sWAT relative to WT mouse sWAT. D and E: scRNA-seq analysis of the SVF from sWAT of pooled 8-week-old mice (n = 3 per group; male). D: Uniform manifold approximation and projection (UMAP) of sWAT SVF cell types in aLpcat3-KO and WT mice. E: Top 10 enriched GO terms in aLpcat3-KO ASPCs relative to WT ASPCs. F: Plasma adipokine profiling of HFHC diet–fed aLpcat3-KO and WT mice (pooled plasma from n = 6 mice in each group). FGF, fibroblast growth factor; IGFBP, IGF binding protein.

Figure 2

Characterization of adipose tissue in aLpcat3-KO and WT mice. AC: Bulk RNA-seq analysis of sWAT from 8-week-old aLpcat3-KO and WT mice fed a normal chow diet (n = 4 per group; male). A: Volcano plot of differentially up- and downregulated genes in aLpcat3-KO and WT mice. B: Heat map of the 30 most significantly up- and downregulated genes in aLpcat3-KO and WT mice. C: PANTHER GO-slim analysis of gene ontology (GO) biological processes in aLpcat3-KO mouse sWAT relative to WT mouse sWAT. D and E: scRNA-seq analysis of the SVF from sWAT of pooled 8-week-old mice (n = 3 per group; male). D: Uniform manifold approximation and projection (UMAP) of sWAT SVF cell types in aLpcat3-KO and WT mice. E: Top 10 enriched GO terms in aLpcat3-KO ASPCs relative to WT ASPCs. F: Plasma adipokine profiling of HFHC diet–fed aLpcat3-KO and WT mice (pooled plasma from n = 6 mice in each group). FGF, fibroblast growth factor; IGFBP, IGF binding protein.

Close modal

To better understand how LPCAT3 affects cellular heterogeneity and function within adipose tissue, we profiled the nonadipocyte fraction, also known as the SVF, from sWAT using scRNA-seq. By clustering gene expression profiles, we identified seven cell types in the SVF: adipose-derived stem and progenitor cells (ASPCs), macrophages, dendritic cells, mesothelial cells, T cells, mast cells, and endothelial cells (3133) (Fig.2D and Supplementary Fig. 3A). There was no major difference in abundance of each cell type when we compared the composition of the SVF from sWAT in aLpcat3-KO and WT mice (Supplementary Fig. 3B).

ASPCs play a critical role in adipose tissue homeostasis. Gene ontology enrichment analysis of genes in ASPC clusters that were differentially expressed between aLpcat3-KO and WT mice revealed over-representation of genes involved in pathways related to transmembrane receptor signaling and response to growth factor stimulus (Fig.2E). Single-cell transcriptome analysis showed that ASPCs are not homogeneous; they form three distinct clusters (ASPC groups 1–3) (Supplementary Fig. 3C). Group 1 cells expressed Dpp4 and Pi16, markers of adipose stem cells (Supplementary Fig. 3D) (34,35). Col4a1, the discriminative marker of matrix fibroblasts (that have properties of adipose stem cells), was also highly expressed in group 1 ASPCs (36). Group 2 cells expressed Fabp4, Col4a1, and Lpl, markers of preadipocytes (Supplementary Fig. 3D) (35,37,38). Preadipocytes are characterized by higher adipogenic capacity than adipose stem cells (34,39,40). Group 3 cells, comprising the smallest group, expressed the same canonical mesenchymal progenitor markers as cells in groups 1 and 2 and also highly expressed mt-Co1, mt-Co2, and Xist, genes characteristic of high mitochondrial activity that could be indicators of apoptotic processes (Supplementary Fig. 3A and D). However, group 3 did not show a strong association with previous classifications, and the function of these cells needs to be further studied. There were some differences in the compositions of these subpopulations between the aLpcat3-KO and WT groups (Supplementary Fig. 3E), indicating an increase in the fraction of preadipocytes in LPCAT3-deficient mice; gene ontology molecular function analysis of each group also indicated that the binding activity of insulin-like growth factor (IGF) is altered when LPCAT3 is deficient (Supplementary Fig. 3F). However, we need to confirm that these differences are caused by LPCAT3 deficiency and not by variation in sample preparation or other confounding factors.

We also assessed plasma adipokine expression using a mouse adipokine protein array in mice fed an HFHC diet for 8 weeks. Interestingly, the expression of fibroblast growth factor 21, IGFs, and IGF-binding proteins was higher in plasma samples from aLpcat3-KO mice compared with those from WT mice, but adiponectin and leptin levels were unaffected (Fig.2F). Fibroblast growth factor 21 protects against systemic insulin resistance (41). IGFs share significant structural homology with insulin (42), and both IGF-I and IGF-II promote glucose clearance (43). Taken together, the results of our plasma adipokine profiling imply that aLPCAT3 deficiency may influence insulin signaling and systemic glucose metabolism, which is consistent with our bulk RNA-seq analysis results.

LPCAT3 Deficiency Enhances Insulin Sensitivity

We assessed fasting glucose levels in normal chow diet–fed aLpcat3-KO and WT mice, and no significant differences were found between the groups (Fig.3A). However, aLpcat3-KO mice showed improved glucose tolerance and clearance, as measured by glucose and insulin tolerance tests, respectively (Fig.3B and C), suggesting LPCAT3 deficiency in adipose tissue enhances systemic insulin sensitivity. To gain a better understanding of the role of LPCAT3 in insulin sensitivity, we next assessed insulin signaling by Western blot in various adipose tissues of aLpcat3-KO and WT mice after injection of insulin or saline as a basal control. Under the stimulation of insulin, LPCAT3 deficiency dramatically induced tyrosine-phosphorylated IR levels in sWAT without affecting total IR levels (Fig.3D). The downstream targets of insulin signaling pathways, phosphorylated AKT and caveolin-1, were also much higher in the sWAT of aLpcat3-KO mice compared with that of WT mice (Fig.3D). Similar to the results in sWAT, insulin-stimulated IR and AKT phosphorylation were also significantly higher in BAT, but expression of caveolin-1 was not affected (Fig.3E). Consistent with the results from LPCAT activity, insulin-stimulated AKT phosphorylation in the liver and skeletal muscle was not different between the groups (Supplementary Fig. 2G). Collectively, these results indicate that aLpcat3 deletion improves systemic insulin sensitivity caused by enhanced insulin signaling in adipose tissue alone.

Figure 3

Improved insulin sensitivity in aLpcat3-KO mice fed a normal chow diet. A: Fasting blood glucose levels (n = 8–10 per group; male). B: Glucose tolerance test (GTT; n = 7 per group; male). C: Insulin tolerance test (n = 8–10 per group; male). D: Western blot analysis of insulin-stimulated phosphorylated IR (p-IR; Y1162), p-AKT (S473), IR, AKT, caveolin-1, and Ponceau S staining in sWAT (n = 3 per group). Data are presented as mean ± SEM. Statistical significance was calculated by two-tailed Student t test. AU, arbitrary units; AUC, area under the curve; ns, not significant (P > 0.05). *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.

Figure 3

Improved insulin sensitivity in aLpcat3-KO mice fed a normal chow diet. A: Fasting blood glucose levels (n = 8–10 per group; male). B: Glucose tolerance test (GTT; n = 7 per group; male). C: Insulin tolerance test (n = 8–10 per group; male). D: Western blot analysis of insulin-stimulated phosphorylated IR (p-IR; Y1162), p-AKT (S473), IR, AKT, caveolin-1, and Ponceau S staining in sWAT (n = 3 per group). Data are presented as mean ± SEM. Statistical significance was calculated by two-tailed Student t test. AU, arbitrary units; AUC, area under the curve; ns, not significant (P > 0.05). *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.

Close modal

aLpcat3 Deletion Attenuates HFHC Diet–Induced Systemic Insulin Resistance

To gain insight into the effect of LPCAT3 on insulin resistance, aLpcat3-KO and WT mice were fed an HFHC diet for 16 weeks, starting at age 8 weeks, to induce obesity and impaired glucose tolerance. Both groups of mice had a similar time-dependent increase in body mass (Supplementary Fig. 4A) and sWAT and epididymal WAT mass (Supplementary Fig. 4B). Moreover, the depletion of LPCAT3 in adipose tissue did not affect the size or number of adipocytes in either group of mice fed an HFHC diet (Supplementary Fig. 4C and D). No significant differences were observed in plasma levels of phospholipids, triglycerides, or cholesterol in aLpcat3-KO mice compared with WT mice fed an HFHC diet (Supplementary Fig. 4E–G), similar to the results for mice fed a normal chow diet. Although there were no significant changes in fasting plasma glucose (Fig.4A) or free fatty acid levels (Supplementary Fig. 4H) in aLpcat3-KO mice compared with WT mice, fasting plasma insulin levels were dramatically reduced in aLpcat3-KO mice compared with WT mice (Fig.4B). Furthermore, aLpcat3-KO mice displayed a marked improvement in glucose and insulin tolerance (Fig.4C and D), similar to the results for mice fed a normal chow diet.

Figure 4

Improved insulin sensitivity in aLpcat3-KO mice fed an HFHC diet. A: Fasting blood glucose levels (n = 6–7 per group; male). B: Fasting plasma insulin levels (n = 5 per group; male). C: Glucose tolerance test (GTT) in HFHC diet–fed mice (n = 6 per group; male). D: Insulin tolerance test in HFHC diet–fed mice (n = 7 per group; male). E: Western blot analysis of insulin-stimulated phosphorylated IR (p-IR; Y1162), p-AKT (S473), IR, AKT, caveolin-1, and Ponceau S in sWAT (n = 4 per group). F: Immunofluorescence staining of sWAT from HFHC diet–fed mice stained with DAPI and antibodies against GLUT4, IR, and caveolin-1. Scale bar, 100 μm. G: Lpcat3 mRNA levels in differentiated mature adipocytes infected with Ad-shRNA-control and Ad-shRNA-LPCAT3 (n = 4 per group). H: Western blot analysis of insulin-stimulated p-IR (Y1162), p-AKT (S473), IR, AKT, and Ponceau S staining in differentiated mature adipocytes infected with Ad-shRNA-control and Ad-shRNA-LPCAT3 (n = 4 per group). I: Extraction of lipid raft fractions from pooled sWAT homogenates (pooled sWAT raft fractions from n = 4 mice per group). Lyn kinase and caveolin-1 were used to detect lipid raft and nonraft fractions, respectively. J: Western blot of p-IR (Y1162), IR, and caveolin-1 in lipid rafts (fractions 3–5; pooled sWAT raft fractions from n = 4 mice per group). Data are presented as mean ± SEM. Statistical significance was calculated by Student’s t test (two-tailed). AU, arbitrary units; AUC, area under the curve; Ctrl, control; ns, not significant (P > 0.05). *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.

Figure 4

Improved insulin sensitivity in aLpcat3-KO mice fed an HFHC diet. A: Fasting blood glucose levels (n = 6–7 per group; male). B: Fasting plasma insulin levels (n = 5 per group; male). C: Glucose tolerance test (GTT) in HFHC diet–fed mice (n = 6 per group; male). D: Insulin tolerance test in HFHC diet–fed mice (n = 7 per group; male). E: Western blot analysis of insulin-stimulated phosphorylated IR (p-IR; Y1162), p-AKT (S473), IR, AKT, caveolin-1, and Ponceau S in sWAT (n = 4 per group). F: Immunofluorescence staining of sWAT from HFHC diet–fed mice stained with DAPI and antibodies against GLUT4, IR, and caveolin-1. Scale bar, 100 μm. G: Lpcat3 mRNA levels in differentiated mature adipocytes infected with Ad-shRNA-control and Ad-shRNA-LPCAT3 (n = 4 per group). H: Western blot analysis of insulin-stimulated p-IR (Y1162), p-AKT (S473), IR, AKT, and Ponceau S staining in differentiated mature adipocytes infected with Ad-shRNA-control and Ad-shRNA-LPCAT3 (n = 4 per group). I: Extraction of lipid raft fractions from pooled sWAT homogenates (pooled sWAT raft fractions from n = 4 mice per group). Lyn kinase and caveolin-1 were used to detect lipid raft and nonraft fractions, respectively. J: Western blot of p-IR (Y1162), IR, and caveolin-1 in lipid rafts (fractions 3–5; pooled sWAT raft fractions from n = 4 mice per group). Data are presented as mean ± SEM. Statistical significance was calculated by Student’s t test (two-tailed). AU, arbitrary units; AUC, area under the curve; Ctrl, control; ns, not significant (P > 0.05). *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.

Close modal

We next examined the effect of LPCAT3 deficiency on adipose tissue insulin signaling by Western blot. As expected, we found a robust induction of IR and AKT phosphorylation in aLpcat3-KO mice compared with WT mice (Fig.4E). We also observed a significant induction of total IR but not AKT protein levels in aLpcat3-KO mice (Fig.4E). In line with this observation, the relative expression of caveolin-1 was significantly higher in aLpcat3-KO mice compared with WT mice (Fig.4E). Collectively, we suggest that LPCAT3 deficiency in adipose tissue attenuates HFHC diet–induced insulin resistance by increasing IR activation and expression.

To confirm our observations, we used immunofluorescence staining for IR and caveolin-1 on sWAT and found that the fluorescence intensities of IR and caveolin-1 were greatly enhanced on the adipocyte plasma membranes of aLpcat3-KO mice compared with on those of WT mice (Fig.4F). We also stained for glucose transporter 4 (GLUT4), a well-known downstream factor in insulin signaling (44,45). GLUT4 on adipocyte plasma membranes was highly induced in the absence of LPCAT3, suggesting the improvement of glucose uptake in adipose tissue in aLpcat3-KO mice (Fig.4F and Supplementary Fig. 4I).

To validate the role of LPCAT3 in adipocyte insulin action, we processed to knock down Lpcat3 expression using adenovirus-mediated shRNA against LPCAT3 in differentiated mature adipocytes from the WT mouse SVF (Fig.4G). LPCAT3 deficiency led to the significant induction of IR and AKT activation (Fig.4H). However, LPCAT3 knockdown in mature adipocytes did not affect lipogenesis or lipolytic pathways, which is consistent with observations in vivo (Supplementary Fig. 5A and B). These observations support our findings that inhibition of LPCAT3 activity improves adipose tissue insulin sensitivity.

Next, we investigated the mechanism of LPCAT3 deficiency–mediated induction of insulin sensitivity. We focused on the lipid rafts in which IR and caveolin-1 are located. We extracted lipid rafts from sWAT of aLpcat3-KO and WT mice fed a normal chow diet. As shown in Fig.4I, lipid rafts (fractions 3–5) and nonrafts (fractions 10–12) were distinguished based on the expression of caveolin-1 and Lyn kinase by Western blot. We next examined the phosphorylation of IR in lipid rafts (fractions 3–5) and found that higher amounts of activated IR and caveolin-1 were expressed in sWAT from aLpcat3-KO mice than in that from WT mice (Fig.4J). Moreover, LPCAT3 deficiency induced the enrichment of cholesterol and sphingomyelin in the lipid rafts on cell membranes (Supplementary Fig. 4J), suggesting an enlargement of the lipid rafts mediated with LPCAT3 deficiency.

LPCAT3 Activity Is Upregulated in Insulin-Resistant Mouse Models

To further evaluate the relationship between LPCAT3 activity and insulin resistance, we used genetic or diet-induced mouse models of insulin resistance: ob/ob (obese mice), db/db (diabetic mice), and HFHC diet–fed WT mice. First, we found that LPCAT3 mRNA levels and activity were upregulated in WAT and BAT of WT mice fed an HFHC diet compared with those of mice fed a normal chow diet (Fig.5A and B and Supplementary Fig. 6A), although the changes in LPCAT3 mRNA levels in BAT did not reach statistical significance (Fig.5A and B). Furthermore, we compared LPCAT3 mRNA levels and activity in sWAT in WT, ob/ob, and db/db mice and found that both ob/ob and db/db mice had significantly higher LPCAT3 mRNA levels and activity than WT mice (Fig.5C and D and Supplementary Fig. 6B). As expected, activation of IR and AKT in ob/ob and db/db adipose tissue was also dramatically inhibited (Fig.5E). Collectively, induction of LPCAT3 expression could be one of the reasons for the impaired insulin signaling observed in ob/ob and db/db mice and in mice with dietary-induced insulin resistance.

Figure 5

LPCAT3 induction in insulin-resistant mouse models. A: Relative Lpcat3 mRNA levels in sWAT and BAT from mice fed a normal chow diet (CD) or HFHC diet (HFHCD; n = 4–8 per group; male). B: Total LPCAT activity in sWAT and BAT from CD- and HFHCD-fed mice (n = 4 per group; male). C: Relative Lpcat3 mRNA levels in sWAT from C57BL/6J, ob/ob, and db/db mice (n = 4 per group; male). D: sWAT LPCAT activity in C57BL/6J, ob/ob, and db/db mice (n = 4 per group; male). E: Western blot analysis of insulin-stimulated phosphorylated IR (p-IR; Y1162), p-AKT (Ser 473), IR, AKT, and Ponceau S in sWAT from C57BL/6J, ob/ob, and db/db mice (n = 4 per group; male). Data are presented as mean ± SEM. Statistical significance was calculated by two-tailed Student t test (A and B) or one-way ANOVA followed by Tukey post hoc multiple comparison test (CE). *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.

Figure 5

LPCAT3 induction in insulin-resistant mouse models. A: Relative Lpcat3 mRNA levels in sWAT and BAT from mice fed a normal chow diet (CD) or HFHC diet (HFHCD; n = 4–8 per group; male). B: Total LPCAT activity in sWAT and BAT from CD- and HFHCD-fed mice (n = 4 per group; male). C: Relative Lpcat3 mRNA levels in sWAT from C57BL/6J, ob/ob, and db/db mice (n = 4 per group; male). D: sWAT LPCAT activity in C57BL/6J, ob/ob, and db/db mice (n = 4 per group; male). E: Western blot analysis of insulin-stimulated phosphorylated IR (p-IR; Y1162), p-AKT (Ser 473), IR, AKT, and Ponceau S in sWAT from C57BL/6J, ob/ob, and db/db mice (n = 4 per group; male). Data are presented as mean ± SEM. Statistical significance was calculated by two-tailed Student t test (A and B) or one-way ANOVA followed by Tukey post hoc multiple comparison test (CE). *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.

Close modal

Polyunsaturated PC Treatment Attenuates Insulin Signaling in Mature Adipocytes

We cultured mature human or mouse adipocytes in vitro and treated them with polyunsaturated PCs to examine insulin signaling. First, we isolated the SVF from aLpcat3-KO mouse sWAT and induced preadipocyte differentiation. After the preadipocytes became mature adipocytes (Supplementary Fig. 7A), we treated the cells with different concentrations of 18:0/18:2 PC, a major product of LPCAT3 activity (46), and stimulated the cells with insulin or saline as a basal control. Under basal conditions, we did not detect phosphorylated IR or phosphorylated AKT. Under insulin-stimulated conditions, we clearly detected phosphorylated IR, phosphorylated AKT, and GLUT4, and all these factors were inhibited by 18:0/18:2 PC in a dose-dependent manner (Fig.6A). In line with this, 18:0/18:2 PC treatment also inhibited the ability of mature adipocytes to take up glucose in a dose-dependent manner (Fig.6B).

Figure 6

Polyunsaturated PC treatments impair adipocyte insulin signaling. A: Western blot analysis of phosphorylated IR (p-IR; Y1162), p-AKT (S473), IR, AKT, GLUT4, and Ponceau S in the SVF of mouse adipocytes differentiated from aLpcat3-KO mice and treated with 0, 10, 50, or 100 μmol/L 18:0/18:2 PC and stimulated with 100 nmol/L insulin for 30 min (n = 2–3 per group). B: Differentiated mouse adipocyte glucose uptake curve (n = 3 per group). C: Western blot analysis of phosphorylated IR (p-IR; Y1162), p-AKT (S473), IR, AKT, and Ponceau S in human adipocytes differentiated from human white preadipocytes and treated with 0, 10, 50, or 100 μmol/L 18:0/18:2 PC and stimulated with 100 nmol/L insulin for 30 min (n = 3 per group). D: Western blot analysis of p-IR (Y1162), p-AKT (S473), IR, AKT, and Ponceau S in mature adipocytes isolated from human WAT and treated with 0 or 100 μmol/L 18:0/18:2 PC and stimulated with 100 nmol/L insulin for 30 min (n = 3 per group). Data are presented as mean ± SEM. Statistical significance was calculated by two-tailed Student t test (D) or one-way ANOVA followed by Tukey post hoc multiple comparison test (AC). AU, arbitrary units; AUC, area under the curve; RLU, relative light units. *P < 0.05, **P < 0.01, ***P < 0.001.

Figure 6

Polyunsaturated PC treatments impair adipocyte insulin signaling. A: Western blot analysis of phosphorylated IR (p-IR; Y1162), p-AKT (S473), IR, AKT, GLUT4, and Ponceau S in the SVF of mouse adipocytes differentiated from aLpcat3-KO mice and treated with 0, 10, 50, or 100 μmol/L 18:0/18:2 PC and stimulated with 100 nmol/L insulin for 30 min (n = 2–3 per group). B: Differentiated mouse adipocyte glucose uptake curve (n = 3 per group). C: Western blot analysis of phosphorylated IR (p-IR; Y1162), p-AKT (S473), IR, AKT, and Ponceau S in human adipocytes differentiated from human white preadipocytes and treated with 0, 10, 50, or 100 μmol/L 18:0/18:2 PC and stimulated with 100 nmol/L insulin for 30 min (n = 3 per group). D: Western blot analysis of p-IR (Y1162), p-AKT (S473), IR, AKT, and Ponceau S in mature adipocytes isolated from human WAT and treated with 0 or 100 μmol/L 18:0/18:2 PC and stimulated with 100 nmol/L insulin for 30 min (n = 3 per group). Data are presented as mean ± SEM. Statistical significance was calculated by two-tailed Student t test (D) or one-way ANOVA followed by Tukey post hoc multiple comparison test (AC). AU, arbitrary units; AUC, area under the curve; RLU, relative light units. *P < 0.05, **P < 0.01, ***P < 0.001.

Close modal

Next, we sought to determine the consequence of polyunsaturated PC treatment on mature human adipocytes, which were derived from human adipose tissue preadipocytes (Supplementary Fig. 7B) or directly isolated from human WAT. We found that 18:0/18:2 PC treatment significantly attenuated insulin signaling by inhibiting the phosphorylation of IR and AKT and by reducing total IR content (Fig.6C and D). We also treated two mature human adipocytes with different concentrations of 18:0/20:4 PC, another major product of LPCAT3 activity (46), and found that the PC also could suppress IR and AKT activation (Supplementary Fig. 8A and B).

Adipose tissue is a key regulator of energy balance, plays an important role in lipid storage, and secretes a wide range of adipokines into the circulation that influence systemic metabolism (47). In the current study, we demonstrated that blocking adipocyte PC remodeling in mice promotes systemic insulin sensitivity, primarily by reducing the amount of polyunsaturated PCs in adipocyte plasma membranes.

One of the key findings of this study is that LPCAT3 is the major isoform of the LPCAT enzyme in adipose tissue, contributing >90% of the PC-remodeling activity. Adiponectin-Cre–mediated Lpcat3 deletion significantly reduced adipose tissue LPCAT3 activity and reduced polyunsaturated PC levels on the plasma membranes of adipocytes. Many reports, including ours (48), have indicated that the adiponectin-Cre–mediated approach can reduce protein or enzyme activity by ∼90%; however, we achieved only a 65 to 70% reduction of LPCAT3 activity in adipose tissue. Nevertheless, this reduction built the foundation for this study.

RNA-seq analysis showed that LPCAT3 deficiency resulted in significant upregulation of 414 genes and downregulation of 180 genes in adipose tissue. Importantly, PANTHER GO-slim analysis revealed adipocyte enrichment of lipid- and glucose metabolism–related genes in the upregulated gene set, indicating that LPCAT3 activity plays a critical role in regulating insulin signaling. This was confirmed by the adipokine array. We also isolated the SVF from aLpcat3-KO and WT mice and performed unbiased scRNA-seq to explore cellular diversity in adipose tissue microenvironments. We did not observe dramatic changes in cell population structure, because LPCAT3 depletion may affect only mature adipocytes that are not part of the SVF. Although the influence of LPCAT3-deficient mature adipocytes on the SVF could be minor, there was a noticeable increase in the preadipocyte fraction in LPCAT3-deficient mice. We observed an increased tendency for transmembrane receptor kinase signaling by gene ontology analysis of ASPCs, which implies insulin signaling in ASPCs, especially in group 1 ASPCs. ASPCs play a critical role in adipose tissue homeostasis and influence adipose tissue expansion during the transition into mature adipocytes (49). Insulin and IR play important roles in adipose tissue development (50). Moreover, ASPCs can differentiate into insulin-producing cells (51). Some studies suggest that abnormal insulin activity negatively affects ASPCs, causing the loss of their proliferative potential and weakening their differentiation potential (52). ASPCs also have a close linkage with human obesity (53). It is possible that LPCAT3 deficiency–mediated polyunsaturated PC reduction on the membrane of ASPCs could influence insulin signaling. This aspect deserves further investigation.

The fatty acids of membrane PCs in mammalian cells exhibit considerable structural diversity (7,54). Saturated PCs promote membrane molecules to pack tightly, thereby decreasing membrane fluidity. The interaction between cholesterol and sphingolipids drives the formation of lipid rafts, which constitute a specific microdomain on the plasma membrane (55). Rafts containing caveolin proteins, called caveolae, are particularly abundant in adipocytes (56). We previously found that manipulation of lipid organization in lipid rafts by reducing sphingomyelin content improves tissue and whole-body insulin sensitivity (57). In addition, caveolin-1 can interact with the IR and enhance insulin-mediated phosphorylation of IR substrate 1 (58). Although sphingolipids and cholesterol are the components of lipid rafts, many PCs are still the major lipids found in raft regions. Saturated and monounsaturated PCs are dominant in the lipid raft regions, whereas polyunsaturated PCs are dominant in nonraft regions (59,60). We hypothesize that increased concentrations of saturated PCs in adipose tissue result in enlargement of lipid rafts where IR is located, thereby enhancing insulin signaling. However, our results were separated by diet conditions. In mice fed a normal chow diet, we could clearly see activation of IR and AKT by their phosphorylation, but their total protein content was unchanged. This could be caused by a reduction in polyunsaturated PC amounts on lipid rafts, which could provide a microenvironment conducive to more IR activation. We indirectly confirmed this hypothesis by treatment of cultured mature human adipocytes with 18:0/18:2 PC. In contrast, the increased lipid content of the HFHC diet leads HFHC diet–fed mice to store more fat in adipose tissue. LPCAT3 deficiency can result in lipid rafts that are enlarged in size and that have greatly reduced amounts of polyunsaturated PCs and more tightly packed IRs. In agreement with these observations, we saw greater protein levels of activated IRs, IRs, and caveolin-1 in LPCAT3-deficient mice.

GLUT4 sits downstream of the insulin signaling pathway and promotes the uptake of glucose when it moves from intracellular storage vesicles to the plasma membrane after insulin signaling is activated (44,45). We confirmed the LPCAT3 deficiency–mediated induction of insulin signaling by GLUT4 immunostaining and polysaturated PC–associated glucose uptake. It is also possible that LPCAT3 deficiency–mediated enlargement of lipid rafts could provide more space for GLUT4 (61). Our glucose and insulin tolerance studies also confirmed that LPCAT3 depletion can promote insulin signaling.

Another key finding of this study is that LPCAT3 is upregulated under three metabolic disorders. Mice fed an HFHC diet had significantly higher LPCAT3 mRNA expression and adipose tissue LPCAT activity (LPCAT3 being the major isoform) than mice fed a normal chow diet. Liver X receptor is one of the important transcription factors that control LPCAT3 expression in liver tissue via a liver X receptor response element in the proximal promoter region of the Lpcat3 gene (17). Furthermore, Lpcat3 is a direct target of peroxisome proliferator–activated receptor δ, which suggests peroxisome proliferator–activated receptor δ functions as a regulator of phospholipid metabolism through LPCAT3 (62). Our results provide further evidence for both regulations. Leptin-deficient ob/ob mice and leptin receptor-deficient db/db mice are widely used as animal models to study human-like obesity and related metabolic disorders (63). We found adipose tissue LPCAT activity was significantly greater in ob/ob and db/db mice than in WT mice, and blockage of insulin signaling observed in these animals could be partially attributed to induction of LPCAT3 activity. The mechanisms of these phenomena also deserve further investigation.

We used two sources of human mature adipocytes to evaluate the human relevance of this study. Results from a previously published in vitro assay determined that 18:0/18:2 PC and 18:0/20:4 PC are major products of human LPCAT3 (46). We found that treatment of both types of mature human adipocytes with 18:0/18:2 PC and 18:0/20:4 PC significantly attenuated insulin signaling by reducing IR activation, IR content, and AKT activation (Fig. 6 and Supplementary Fig. 8). However, 18:0/20:4 PC was not as effective as 18:0/18:2 PC. This could be because 1) 18:0/18:2 PC is the most abundant PC in adipose tissue and could be the most abundant PC in lipid rafts, so its changes could have larger effects than 18:0/20:4 PC, and 2) exogenous PC supplementation may affect insulin signaling in a different manner than endogenous PC supplementation.

Although we focused our study on WAT, we also found that LPCAT3 is the major isoform in BAT and that LPCAT3 deficiency enhanced insulin signaling. Although both BAT and WAT secrete factors that act as hormones to affect liver and skeletal muscle metabolism, BAT differs fundamentally from WAT, because BAT can directly take up large quantities of glucose (64). Thus, BAT and WAT differ mechanistically in their glucose uptake and metabolism pathways. The insulin-sensitive GLUT4 translocation pathway operates in BAT, but in the context of a more active GLUT1 translocation pathway (GLUT1 being regulated by norepinephrine) (65). Nonetheless, insulin substantially stimulates glucose uptake in BAT (66). Silencing of GLUT4 and GLUT1 in brown adipocytes shows that both contribute extensively to overall glucose uptake (67). Thus, LPCAT3 deficiency in BAT can also promote systemic insulin sensitivity, as we observed in this study.

There are some limitations to this study. Although a 65 to 70% reduction of LPCAT3 activity in adipose tissue built the foundation for the current study, we still do not know why the approach could not achieve >90% reduction, as in other gene KO studies. Also, it should be important to study why 18:0/18:2 PC was more effective than 18:0/20:4 PC at blocking insulin signaling in mature adipocytes, although both PCs are the products of LPCAT3 activity.

Can inhibition of LPCAT3 be used as a treatment for metabolic diseases, such as insulin resistance and dyslipidemia? The current aLPCAT3 deficiency study, together with a previous skeletal muscle–specific LPCAT3 deficiency study (68), clearly indicates depletion of LPCAT3 in both tissues can significantly increase systemic insulin sensitivity. In our previous inducible small intestine– and liver-specific studies (14,15), we found that 1) small intestine LPCAT3 deficiency has a bigger effect on plasma lipid levels than that of liver deficiency, 2) the deficiency has an effect on lipid retention in both tissue, and 3) the deficiency has a marginal effect on endoplasmic reticulum stress. Taken together, we believe that inhibition of LPCAT3 activity could be a novel approach for treating insulin resistance and hyperlipidemia.

Funding. This work was supported by Veterans Affairs Merit Award 000900-01, National Institutes of Health (NIH) grants RO1 HL139582 and RO1 HL149730 grants to X.-C.J., and NIH grants P30 AG013319 and P30 AG044271 to the Lipidomics Core (X.H.).

Duality of Interest. No potential conflicts of interest relevant to this article were reported.

Author Contributions. M.H. and Z.L. performed 85% of the experiments, analyzed data, and modified the manuscript. V.S.K.T. and O.E. performed single-cell sequence analyses and modified the manuscript. M.P. and X.H. measured lipids using liquid chromatography–coupled tandem mass spectrometry and modified the manuscript. X.-C.J. conceived the ideas, designed and discussed the experiments, supervised progress, and wrote the manuscript. X.-C.J. is the guarantor of this work and, as such, had full access to all the data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis.

This article contains supplementary material online at https://doi.org/10.2337/figshare.24018636.

M.H., Z.L., and V.S.K.T. contributed equally to this work.

1.
van Meer
G
,
Voelker
DR
,
Feigenson
GW.
Membrane lipids: where they are and how they behave
.
Nat Rev Mol Cell Biol
2008
;
9
:
112
124
2.
Schiller
J
,
Zschörnig
O
,
Petković
M
,
Müller
M
,
Arnhold
J
,
Arnold
K.
Lipid analysis of human HDL and LDL by MALDI-TOF mass spectrometry and (31)P-NMR
.
J Lipid Res
2001
;
42
:
1501
1508
3.
Lands
WE.
Stories about acyl chains
.
Biochim Biophys Acta
2000
;
1483
:
1
14
4.
Shindou
H
,
Shimizu
T.
Acyl-CoA:lysophospholipid acyltransferases
.
J Biol Chem
2009
;
284
:
1
5
5.
Kennedy
EP
,
Weiss
SB.
The function of cytidine coenzymes in the biosynthesis of phospholipides
.
J Biol Chem
1956
;
222
:
193
214
6.
Lands
WE.
Metabolism of glycerolipides; a comparison of lecithin and triglyceride synthesis
.
J Biol Chem
1958
;
231
:
883
888
7.
MacDonald
JI
,
Sprecher
H.
Phospholipid fatty acid remodeling in mammalian cells
.
Biochim Biophys Acta
1991
;
1084
:
105
121
8.
Clandinin
MT
,
Cheema
S
,
Field
CJ
,
Garg
ML
,
Venkatraman
J
,
Clandinin
TR.
Dietary fat: exogenous determination of membrane structure and cell function
.
FASEB J
1991
;
5
:
2761
2769
9.
Ntambi
JM.
The regulation of stearoyl-CoA desaturase (SCD)
.
Prog Lipid Res
1995
;
34
:
139
150
10.
Lee
HC
,
Inoue
T
,
Imae
R
, et al
.
Caenorhabditis elegans mboa-7, a member of the MBOAT family, is required for selective incorporation of polyunsaturated fatty acids into phosphatidylinositol
.
Mol Biol Cell
2008
;
19
:
1174
1184
11.
Hishikawa
D
,
Shindou
H
,
Kobayashi
S
,
Nakanishi
H
,
Taguchi
R
,
Shimizu
T.
Discovery of a lysophospholipid acyltransferase family essential for membrane asymmetry and diversity
.
Proc Natl Acad Sci USA
2008
;
105
:
2830
2835
12.
Rong
X
,
Wang
B
,
Dunham
MM
, et al
.
Lpcat3-dependent production of arachidonoyl phospholipids is a key determinant of triglyceride secretion
.
eLife
2015
;
4
:
e06557
13.
Hashidate-Yoshida
T
,
Harayama
T
,
Hishikawa
D
, et al
.
Fatty acid remodeling by LPCAT3 enriches arachidonate in phospholipid membranes and regulates triglyceride transport
.
eLife
2015
;
4
:
e06328
14.
Kabir
I
,
Li
Z
,
Bui
HH
,
Kuo
MS
,
Gao
G
,
Jiang
XC.
Small intestine but not liver lysophosphatidylcholine acyltransferase 3 (Lpcat3) deficiency has a dominant effect on plasma lipid metabolism
.
J Biol Chem
2016
;
291
:
7651
7660
15.
Li
Z
,
Jiang
H
,
Ding
T
, et al
.
Deficiency in lysophosphatidylcholine acyltransferase 3 reduces plasma levels of lipids by reducing lipid absorption in mice
.
Gastroenterology
2015
;
149
:
1519
1529
16.
Ishibashi
M
,
Varin
A
,
Filomenko
R
, et al
.
Liver X receptor regulates arachidonic acid distribution and eicosanoid release in human macrophages: a key role for lysophosphatidylcholine acyltransferase 3
.
Arterioscler Thromb Vasc Biol
2013
;
33
:
1171
1179
17.
Rong
X
,
Albert
CJ
,
Hong
C
, et al
.
LXRs regulate ER stress and inflammation through dynamic modulation of membrane phospholipid composition
.
Cell Metab
2013
;
18
:
685
697
18.
Samra
JS.
Sir David Cuthbertson Medal Lecture. Regulation of lipid metabolism in adipose tissue
.
Proc Nutr Soc
2000
;
59
:
441
446
19.
Fairley
C
,
Wilson
JW
,
Barraclough
D.
Pulmonary involvement in Behçet’s syndrome
.
Chest
1989
;
96
:
1428
1429
20.
Li
Z
,
Ding
T
,
Pan
X
, et al
.
Lysophosphatidylcholine acyltransferase 3 knockdown-mediated liver lysophosphatidylcholine accumulation promotes very low density lipoprotein production by enhancing microsomal triglyceride transfer protein expression
.
J Biol Chem
2012
;
287
:
20122
20131
21.
Liu
L
,
Zheng
LD
,
Donnelly
SR
,
Emont
MP
,
Wu
J
,
Cheng
Z.
Isolation of mouse stromal vascular cells for monolayer culture
.
Methods Mol Biol
2017
;
1566
:
9
16
22.
Alexandersson
I
,
Harms
MJ
,
Boucher
J.
Isolation
and culture of human mature adipocytes using membrane mature adipocyte aggregate cultures (MAAC)
.
J Vis Exp.
13 February
2020
[Epub ahead of print]. DOI: 10.3791/60485
23.
Huo
H
,
Guo
X
,
Hong
S
,
Jiang
M
,
Liu
X
,
Liao
K.
Lipid rafts/caveolae are essential for insulin-like growth factor-1 receptor signaling during 3T3-L1 preadipocyte differentiation induction
.
J Biol Chem
2003
;
278
:
11561
11569
24.
Yuan
T
,
Hong
S
,
Yao
Y
,
Liao
K.
Glut-4 is translocated to both caveolae and non-caveolar lipid rafts, but is partially internalized through caveolae in insulin-stimulated adipocytes
.
Cell Res
2007
;
17
:
772
782
25.
Li
Z
,
He
M
,
Chen
G
,
Souaiaia
T
,
Worgall
TS
,
Jiang
XC.
Effect of total SMS activity on LDL catabolism in mice
.
Arterioscler Thromb Vasc Biol
2023
;
43
:
1251
1261
26.
Beliveau
BJ
,
Joyce
EF
,
Apostolopoulos
N
, et al
.
Versatile design and synthesis platform for visualizing genomes with Oligopaint FISH probes
.
Proc Natl Acad Sci USA
2012
;
109
:
21301
21306
27.
Love
MI
,
Huber
W
,
Anders
S.
Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2
.
Genome Biol
2014
;
15
:
550
28.
Ashburner
M
,
Ball
CA
,
Blake
JA
, et al;
The Gene Ontology Consortium
.
Gene ontology: tool for the unification of biology
.
Nat Genet
2000
;
25
:
25
29
29.
Gene Ontology Consortium
.
The Gene Ontology resource: enriching a GOld mine
.
Nucleic Acids Res
2021
;
49
:
D325
D334
30.
Stuart
T
,
Butler
A
,
Hoffman
P
, et al
.
Comprehensive integration of single-cell data
.
Cell
2019
;
177
:
1888
1902.e21
31.
Cui
K
,
Gao
X
,
Wang
B
, et al
.
Epsin nanotherapy regulates cholesterol transport to fortify atheroma regression
.
Circ Res
2023
;
132
:
e22
e42
32.
Emont
MP
,
Jacobs
C
,
Essene
AL
, et al
.
A single-cell atlas of human and mouse white adipose tissue
.
Nature
2022
;
603
:
926
933
33.
Vijay
J
,
Gauthier
MF
,
Biswell
RL
, et al
.
Single-cell analysis of human adipose tissue identifies depot and disease specific cell types
.
Nat Metab
2020
;
2
:
97
109
34.
Merrick
D
,
Sakers
A
,
Irgebay
Z
, et al
.
Identification of a mesenchymal progenitor cell hierarchy in adipose tissue
.
Science
2019
;
364
:
eaav2501
35.
Ferrero
R
,
Rainer
P
,
Deplancke
B.
Toward a consensus view of mammalian adipocyte stem and progenitor cell heterogeneity
.
Trends Cell Biol
2020
;
30
:
937
950
36.
Xie
T
,
Wang
Y
,
Deng
N
, et al
.
Single-cell deconvolution of fibroblast heterogeneity in mouse pulmonary fibrosis
.
Cell Rep
2018
;
22
:
3625
3640
37.
Cristancho
AG
,
Lazar
MA.
Forming functional fat: a growing understanding of adipocyte differentiation
.
Nat Rev Mol Cell Biol
2011
;
12
:
722
734
38.
Tang
W
,
Zeve
D
,
Suh
JM
, et al
.
White fat progenitor cells reside in the adipose vasculature
.
Science
2008
;
322
:
583
586
39.
Gustafson
B
,
Hedjazifar
S
,
Gogg
S
,
Hammarstedt
A
,
Smith
U.
Insulin resistance and impaired adipogenesis
.
Trends Endocrinol Metab
2015
;
26
:
193
200
40.
Dong
H
,
Sun
W
,
Shen
Y
, et al
.
Identification of a regulatory pathway inhibiting adipogenesis via RSPO2
.
Nat Metab
2022
;
4
:
90
105
41.
Li
H
,
Wu
G
,
Fang
Q
, et al
.
Fibroblast growth factor 21 increases insulin sensitivity through specific expansion of subcutaneous fat
.
Nat Commun
2018
;
9
:
272
42.
Blundell
TL
,
Bedarkar
S
,
Rinderknecht
E
,
Humbel
RE.
Insulin-like growth factor: a model for tertiary structure accounting for immunoreactivity and receptor binding
.
Proc Natl Acad Sci USA
1978
;
75
:
180
184
43.
Zapf
J
,
Schoenle
E
,
Froesch
ER.
Insulin-like growth factors I and II: some biological actions and receptor binding characteristics of two purified constituents of nonsuppressible insulin-like activity of human serum
.
Eur J Biochem
1978
;
87
:
285
296
44.
Bryant
NJ
,
Govers
R
,
James
DE.
Regulated transport of the glucose transporter GLUT4
.
Nat Rev Mol Cell Biol
2002
;
3
:
267
277
45.
Huang
S
,
Czech
MP.
The GLUT4 glucose transporter
.
Cell Metab
2007
;
5
:
237
252
46.
Zhao
Y
,
Chen
YQ
,
Bonacci
TM
, et al
.
Identification and characterization of a major liver lysophosphatidylcholine acyltransferase
.
J Biol Chem
2008
;
283
:
8258
8265
47.
Romacho
T
,
Elsen
M
,
Röhrborn
D
,
Eckel
J.
Adipose tissue and its role in organ crosstalk
.
Acta Physiol (Oxf)
2014
;
210
:
733
753
48.
Lee
SY
,
Lee
HY
,
Song
JH
, et al
.
Adipocyte-specific deficiency of de novo sphingolipid biosynthesis leads to lipodystrophy and insulin resistance
.
Diabetes
2017
;
66
:
2596
2609
49.
Câmara
DAD
,
Shibli
JA
,
Müller
EA
, et al
.
Adipose tissue-derived stem cells: the biologic basis and future directions for tissue engineering
.
Materials (Basel)
2020
;
13
:
3210
50.
Cignarelli
A
,
Genchi
VA
,
Perrini
S
,
Natalicchio
A
,
Laviola
L
,
Giorgino
F.
Insulin and insulin receptors in adipose tissue development
.
Int J Mol Sci
2019
;
20
:
759
51.
Wada
Y
,
Ikemoto
T
,
Morine
Y
, et al
.
The differences in the characteristics of insulin-producing cells using human adipose-tissue derived mesenchymal stem cells from subcutaneous and visceral tissues
.
Sci Rep
2019
;
9
:
13204
52.
Skubis-Sikora
A
,
Sikora
B
,
Witkowska
A
,
Mazurek
U
,
Gola
J.
Osteogenesis of adipose-derived stem cells from patients with glucose metabolism disorders
.
Mol Med
2020
;
26
:
67
53.
Yang
H
,
Li
C
,
Li
Y
,
Tai
R
,
Sun
C.
Adipose-derived stem cells and obesity: the spear and shield relationship
.
Genes Dis
2021
;
10
:
175
186
54.
Holub
BJ
,
Kuksis
A.
Metabolism of molecular species of diacylglycerophospholipids
.
Adv Lipid Res
1978
;
16
:
1
125
55.
Simons
K
,
Ikonen
E.
Functional rafts in cell membranes
.
Nature
1997
;
387
:
569
572
56.
Razani
B
,
Woodman
SE
,
Lisanti
MP.
Caveolae: from cell biology to animal physiology
.
Pharmacol Rev
2002
;
54
:
431
467
57.
Li
Z
,
Zhang
H
,
Liu
J
, et al
.
Reducing plasma membrane sphingomyelin increases insulin sensitivity
.
Mol Cell Biol
2011
;
31
:
4205
4218
58.
Yamamoto
M
,
Toya
Y
,
Schwencke
C
,
Lisanti
MP
,
Myers
MG
Jr
,
Ishikawa
Y.
Caveolin is an activator of insulin receptor signaling
.
J Biol Chem
1998
;
273
:
26962
26968
59.
Hattersley
KJ
,
Hein
LK
,
Fuller
M.
Lipid composition of membrane rafts, isolated with and without detergent, from the spleen of a mouse model of Gaucher disease
.
Biochem Biophys Res Commun
2013
;
442
:
62
67
60.
de Almeida
RF
,
Fedorov
A
,
Prieto
M.
Sphingomyelin/phosphatidylcholine/cholesterol phase diagram: boundaries and composition of lipid rafts
.
Biophys J
2003
;
85
:
2406
2416
61.
Ros-Baro
A
,
Lopez-Iglesias
C
,
Peiro
S
, et al
.
Lipid rafts are required for GLUT4 internalization in adipose cells
.
Proc Natl Acad Sci USA
2001
;
98
:
12050
12055
62.
Singh
AB
,
Liu
J.
Identification of hepatic lysophosphatidylcholine acyltransferase 3 as a novel target gene regulated by peroxisome proliferator-activated receptor δ
.
J Biol Chem
2017
;
292
:
884
897
63.
Suriano
F
,
Vieira-Silva
S
,
Falony
G
, et al
.
Novel insights into the genetically obese (ob/ob) and diabetic (db/db) mice: two sides of the same coin
.
Microbiome
2021
;
9
:
147
64.
Hankir
MK
,
Cowley
MA
,
Fenske
WK.
A BAT-centric approach to the treatment of diabetes: turn on the brain
.
Cell Metab
2016
;
24
:
31
40
65.
Czech
MP.
Mechanisms of insulin resistance related to white, beige, and brown adipocytes
.
Mol Metab
2020
;
34
:
27
42
66.
Orava
J
,
Nuutila
P
,
Lidell
ME
, et al
.
Different metabolic responses of human brown adipose tissue to activation by cold and insulin
.
Cell Metab
2011
;
14
:
272
279
67.
Winther
S
,
Isidor
MS
,
Basse
AL
, et al
.
Restricting glycolysis impairs brown adipocyte glucose and oxygen consumption
.
Am J Physiol Endocrinol Metab
2018
;
314
:
E214
E223
68.
Ferrara
PJ
,
Rong
X
,
Maschek
JA
, et al
.
Lysophospholipid acylation modulates plasma membrane lipid organization and insulin sensitivity in skeletal muscle
.
J Clin Invest
2021
;
131
:
e135963
Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. More information is available at https://www.diabetesjournals.org/journals/pages/license.