Neuropilin 1 (Nrp1), a coreceptor for class 3 semaphorins and growth factors, is highly expressed in vascular cells and myeloid cells, including macrophages. Unlike well-characterized proangiogenic functions of endothelial cell Nrp1, the contributions of macrophage Nrp1 within the context of metabolic dysfunction remain to be established. The aim of this study was to determine the contributions of macrophage Nrp1 in high-fat diet (HFD)-instigated insulin resistance in vivo. Insulin sensitivity and Nlrp3 inflammasome activation were monitored in wild-type (WT) and myeloid cell-specific Nrp1 knockout (Nrp1myel-KO) mice fed an HFD (60% kcal) for 16 weeks. HFD-fed mice exhibited insulin resistance with reduced levels of Nrp1 in macrophages compared with chow-fed mice. Further, HFD-fed Nrp1myel-KO mice displayed accentuated insulin resistance, enhanced systemic inflammation, and dramatically increased Nlrp3 inflammasome priming and activation. Importantly, knockout of Nlrp3 ablated HFD-induced insulin resistance and inflammation in Nrp1myel-KO mice, indicating that Nrp1 reduction in macrophages instigates insulin resistance by increasing macrophage Nlrp3 inflammasome activation. Mechanistically, Nrp1 deletion activates the nuclear factor-κB pathway, which in turn accentuates the priming of Nlrp3, promotes Nlrp3-ASC inflammasome assembly, and results in the activation of Nlrp3. We conclude that the HFD-instigated Nrp1 reduction in macrophages exacerbates insulin resistance by promoting Nlrp3 inflammasome priming and activation.

Obesity has become a global epidemic and a major cause of insulin resistance, which contributes to metabolic syndrome and type 2 diabetes (1). Chronic inflammation mediated by accumulated myeloid cells and lymphoid lineage in visceral adipose tissues is a major determinant of insulin resistance (24). Notably, macrophages infiltrated in visceral adipose tissues may constitute up to 40% of total obese visceral adipose tissue cells (3,4). Adipose tissue macrophages (ATMs) secrete proinflammatory cytokines, including tumor necrosis factor-α (TNF-α) (5) and chemokine (C-C motif) ligand 2 (Ccl2) (6). These cytokines inhibit insulin signaling in target tissues, impair glucose tolerance, and eventually instigate insulin resistance (5,7,8). Therefore, inhibition of macrophage-mediated chronic low-grade inflammation is an effective strategy to protect against obesity-induced insulin resistance and diabetes.

Emerging evidence supports an association between neuronal guidance cues and their receptors with insulin resistance. For example, axon-guiding class 3 semaphorin E (Sema3E) and its receptor plexin-D1 promote macrophage recruitment into adipose tissues, which triggers inflammation and systemic insulin resistance (9). Furthermore, another neuronal guidance cue, netrin-1 and its receptor Unc5, are highly expressed in visceral adipose tissues of obese human subjects and mice relative to lean ones (10). Netrin-1 has been shown to act as a local macrophage retention signal, leading to macrophage accumulation in adipose tissues (10). These studies suggest that the receptors of neuronal guidance cues may be required for inflammatory chemotaxis in adipose tissues. However, the direct links and molecular mechanisms of these receptors with respect to metabolic disorders remain largely unknown.

Neuropilin 1 (Nrp1), initially identified as a coreceptor for Sema3 and growth factors, including vascular endothelial growth factor, transforming growth factor-β, hepatocyte growth factor, and platelet-derived growth factor (1114), has been shown to be strongly expressed in immune cells and to regulate immune response (1517). In macrophages, ablation of Nrp1 results in decreased tumor growth and metastasis via enhanced infiltration of tumor-associated macrophages into normoxic tumor regions, which abolishes the proangiogenic and immunosuppressive functions of tumor-associated macrophages (18). Consistently, Miyauchi et al. (19) reported that Nrp1 ablation in glioma-associated microglia and macrophages suppresses glioma progression by promoting M1 macrophage polarization. Recent findings further establish the involvement of the Sema3A/Nrp1 axis, as well as M1/M2 macrophages, with respect to tumorigenic processes (20). However, specific functions of macrophage Nrp1 in the context of metabolic dysfunction, such as obesity-associated insulin resistance, have not been investigated. Here we report that a high-fat diet (HFD)–attenuated Nrp1 expression in macrophages correlates with accentuated insulin resistance, which is majorly caused by activated Nlrp3 inflammasome.

Mice

Nrp1flox/flox mice (The Jackson Laboratory, Bar Harbor, ME) were backcrossed to C57BL/6J for at least 10 generations before crossing with lysozyme M (LysM)–Cre mice (The Jackson Laboratory) to generate myeloid cell–specific knockout (Nrp1myel-KO) mice. Nrp1flox/flox mice with the LysM-Cre transgene were Nrp1myel-KO mice, and Nrp1flox/flox littermates without the LysM-Cre transgene served as wild-type (WT) controls. To generate Nrp1myel-KONlrp3−/− mice, Nlrp3−/− mice were crossed with Nrp1myel-KO mice. Nrp1myel-KONlrp3+/+ littermates served as controls. All mice were C57BL/6J background, housed in temperature-controlled cages with a 12-h light-dark cycle, and given free access to water and food. Mice of different genotypes were housed in the same cage. Mice were fed an HFD (60% fat kcal, D12492; Research Diets Inc., New Brunswick, NJ) or a normal chow diet for 16 weeks. Mice were sacrificed, and white adipose tissue (WAT), liver, and skeletal muscle were isolated for biochemical analyses. The Georgia State University Institutional Animal Care and Use Committee reviewed and approved the animal protocol.

Cell Culture

Peritoneal macrophages (PMs) were isolated as previously described with minor modification (21). Briefly, cells were collected by peritoneal lavage with 8 mL cold PBS with 10 mmol/L EDTA and 10% FBS. Macrophages were plated at 1.0 × 106 per mL RPMI-1640 with 10% FBS. After incubation for 3 h at 37°C, the nonadherent cells were washed away, and adherent cells were collected for experiments.

Primary bone marrow–derived macrophages (BMDMs) were prepared by flushing the bone marrow of the femur and tibia of mice. Cells were cultured with complete RPMI-1640 medium supplemented with mouse macrophage colony-stimulating factor recombinant protein (50 ng/mL; eBioscience, San Diego, CA) for 7 days (22).

RAW264.7 cells (American Type Culture Collection, Manassas, VA) were cultured with DMEM containing 10% FBS and maintained at 37°C in 5% CO2 and 95% air.

Reagents

Nuclear factor (NF)-κB activation inhibitor (481406) was obtained from EMD Millipore (Billerica, MA). Ultrapure lipopolysaccharide (LPS) was bought from InvivoGen (San Diego, CA). ATP was purchased from Roche Applied Science (Basel, Switzerland). Nigericin was obtained from Sigma-Aldrich (St. Louis, MO). Nrp1 plasmid (#21934) was purchased from Addgene (Cambridge, MA). A Transcription Factor Kit for NF-κB p65 (#89859) was obtained from Thermo Fisher Scientific (Waltham, MA).

Glucose and Insulin Tolerance Tests

Glucose and insulin tolerance tests were performed as previously described (10). Briefly, for the glucose tolerance test (GTT), the mice were fasted for 16 h. After determination of fasted blood glucose levels, each animal received a glucose intraperitoneal injection of 1 g/kg body weight of glucose (25% d-(+)-glucose; G7528, Sigma-Aldrich). Blood glucose levels were determined after 15, 30, 60, and 120 min. The mice were fasted for 4 h, and insulin tolerance tests (ITTs) were performed by an intraperitoneal injection of 1.5 units/kg body weight of insulin (I9278, Sigma-Aldrich). Blood glucose levels were measured at 0, 15, 30, 60, and 120 min.

Analytical Procedures

Glucose concentrations in whole venous blood were measured by an automatic glucose monitoring system (ReliOn Confirm). Serum insulin levels were measured by ELISA (ALPCO, Windham, NH). Serum free fatty acid (FFA) levels were measured using an enzyme kit (BioVision, Mountain View, CA). HOMA of insulin resistance (HOMA-IR) was calculated as (fasting insulin [mIU/L] × fasting glucose [mmol/L])/22.5 (23).

Statistical Analysis

Quantitative data are expressed as mean ± SEM of at least three independent experiments. The difference between two groups was analyzed by the Student t test. The difference among more than two groups was analyzed by one-way ANOVA, followed by the Newman-Keuls multiple comparison test. Comparisons of different parameters between each group were made by two-way ANOVA, followed by the Bonferroni posttest. Statistical significance was evaluated with GraphPad Prism 5 software. P values of <0.05 were considered statistically significant.

Obesity Decreases Nrp1 Expression in Macrophages

To establish the association between Nrp1 and obesity, we adopted a diet-induced obesity model by feeding C57BL/6J mice with an HFD (60% kcal) for 16 weeks. Control mice were fed a normal diet (chow) in parallel with the HFD group. As expected, body weight and serum total cholesterol were significantly elevated in HFD-fed mice compared with chow-fed mice (Supplementary Fig. 1A and B). Furthermore, GTT and ITT results showed significantly impaired glucose tolerance and markedly blunted insulin responsiveness in HFD-fed mice relative to chow-fed mice (Supplementary Fig. 1C and D). These results indicated that HFD-fed mice successfully developed obesity-associated insulin resistance.

Next, we determined the expression of Nrp1 in visceral WAT, and as shown in Fig. 1A, Nrp1 mRNA levels were profoundly decreased in WAT of HFD-fed compared with chow-fed mice. Immunoblot analysis confirmed decreased Nrp1 protein in visceral WAT of HFD-fed mice relative to chow-fed mice (Fig. 1B). Furthermore, we assessed the expression of Nrp1 in subcutaneous WAT under the same dietary conditions but failed to detect differential Nrp1 expression (Fig. 1C and D), suggesting the reduction of Nrp1 by HFD is specific to visceral WAT. To identify the specific cellular compartment of WAT with decreased Nrp1 expression, we isolated the stromal vascular fraction (SVF) and adipocyte-rich fraction (ARF) from visceral WAT of chow- and HFD-fed mice. As shown in Fig. 1E and G, mRNA levels of Nrp1 were dramatically downregulated in the SVF but not the ARF of HFD-fed mice compared with samples from chow-fed mice. Attenuated Nrp1 protein was confirmed in SVF but not ARF by immunoblot analysis (Fig. 1F and H). Although macrophages represent the dominant cells in the SVF of adipose tissues, there are a variety of other cell types, including preadipocytes, mesenchymal stem cells, endothelial cells, T cells, and B cells (24,25). We therefore used immunofluorescence microscopy to visualize the costaining of Nrp1 with various established cell-specific markers, such as von Willebrand factor (endothelial cell), CD3 (T cell), or CD11c (dendritic cell) in visceral WAT of chow- or HFD-fed mice. The results showed that the HFD did not affect Nrp1 expression in these cells of visceral WAT (Supplementary Fig. 2A–C). We eventually isolated PMs from chow-fed and HFD-fed mice to ensure the purity of macrophages. As shown in Fig. 1I, Nrp1 mRNA was significantly downregulated by the HFD. Consistently, decreased Nrp1 protein was confirmed in PMs of HFD-fed mice relative to chow-fed mice (Fig. 1J). Together, these data suggest that the HFD suppresses Nrp1 expression in macrophages.

Figure 1

Nrp1 is downregulated in macrophages of obese mice. C57BL/6J mice were fed a chow diet (lean) or an HFD (obese) for 16 weeks. A and B: Quantitative RT-PCR and immunoblot analysis of Nrp1 in visceral WAT (n = 5 mice per group). C and D: Quantitative RT-PCR and immunoblot analysis of Nrp1 in subcutaneous WAT (n = 6 mice per group). E and F: Quantitative RT-PCR and immunoblot analysis of Nrp1 in the SVF isolated from visceral WAT (n = 5 mice per group). G and H: Quantitative RT-PCR and immunoblot analysis of Nrp1 in the ARF isolated from visceral WAT (n = 5 mice per group). I and J: Quantitative RT-PCR and immunoblot analysis of Nrp1 in PMs (n = 5–7 mice per group). β-Tubulin or β-actin served as a loading control. Data are expressed as mean ± SEM. *P < 0.05 vs. chow.

Figure 1

Nrp1 is downregulated in macrophages of obese mice. C57BL/6J mice were fed a chow diet (lean) or an HFD (obese) for 16 weeks. A and B: Quantitative RT-PCR and immunoblot analysis of Nrp1 in visceral WAT (n = 5 mice per group). C and D: Quantitative RT-PCR and immunoblot analysis of Nrp1 in subcutaneous WAT (n = 6 mice per group). E and F: Quantitative RT-PCR and immunoblot analysis of Nrp1 in the SVF isolated from visceral WAT (n = 5 mice per group). G and H: Quantitative RT-PCR and immunoblot analysis of Nrp1 in the ARF isolated from visceral WAT (n = 5 mice per group). I and J: Quantitative RT-PCR and immunoblot analysis of Nrp1 in PMs (n = 5–7 mice per group). β-Tubulin or β-actin served as a loading control. Data are expressed as mean ± SEM. *P < 0.05 vs. chow.

Close modal

Insulin Resistance Is Exacerbated in HFD-Fed Nrp1myel-KO Mice

To investigate specific roles of macrophage Nrp1 in obesity-associated insulin resistance, Nrp1 was inactivated in myeloid cells by crossing Nrp1flox/flox mice into mice expressing LysM-Cre recombinase. Quantitative RT-PCR data showed significantly reduced expression of Nrp1 mRNA in Nrp1−/− BMDMs compared with WT BMDMs (Supplementary Fig. 3A). Decreased Nrp1 expression in Nrp1−/− BMDMs was further confirmed by immunofluorescence staining and immunoblot analysis (Supplementary Fig. 3B and C. We also assessed tissue-specific deletion of Nrp1 in WAT, liver, and skeletal muscle of WT and Nrp1myel-KO mice. Our data demonstrated that Nrp1 deletion was specific to macrophages (Supplementary Fig. 3D).

We next exposed WT and Nrp1myel-KO mice to chow or the HFD for 16 weeks. As would be expected, the body weight of HFD-fed mice was markedly increased (Supplementary Fig. 4). However, no significant difference was observed between WT and Nrp1myel-KO mice fed chow or the HFD (Supplementary Fig. 4). We used GTT and ITT to analyze possible alterations in glucose homeostasis in these mice. Although no significant difference was detected under chow-fed conditions in both mouse groups (Supplementary Fig. 5A and B), HFD-fed Nrp1myel-KO mice were more intolerant to glucose and more resistant to insulin than their WT counterparts (Fig. 2A and B). Consistently, HFD-fed Nrp1myel-KO mice displayed higher fasting serum insulin concentrations and elevated HOMA-IR index relative to their WT littermates (Fig. 2C and D). We also measured serum FFAs and observed no obvious difference between HFD-fed WT and Nrp1myel-KO mice (Fig. 2E). Because the metabolic effects of insulin largely depend on phosphoinositol 3 kinase–protein kinase B (AKT) signaling in target tissues, we measured the phosphorylation (p) of AKT at Ser473 in WAT, liver, and skeletal muscle. Consistent with impaired insulin sensitivity in Nrp1myel-KO mice fed the HFD, decreased p-AKT levels were evident in these tissues (Fig. 2F). Taken together, these data indicate that ablation of Nrp1 in myeloid cells leads to impaired maintenance of glucose homeostasis and blunted insulin sensitivity during obesity.

Figure 2

Insulin resistance is accentuated in HFD-fed Nrp1myel-KO mice. WT and Nrp1myel-KO mice were fed the HFD for 16 weeks (n = 7–8 mice per group). A and B: GTT and ITT. C and D: Fasting serum insulin concentrations and HOMA-IR index. E: Serum levels of FFAs. F: Immunoblot analysis of p-AKT (Ser473) and total AKT in WAT, liver, and skeletal muscle. Data are expressed as mean ± SEM. *P < 0.05 vs. WT.

Figure 2

Insulin resistance is accentuated in HFD-fed Nrp1myel-KO mice. WT and Nrp1myel-KO mice were fed the HFD for 16 weeks (n = 7–8 mice per group). A and B: GTT and ITT. C and D: Fasting serum insulin concentrations and HOMA-IR index. E: Serum levels of FFAs. F: Immunoblot analysis of p-AKT (Ser473) and total AKT in WAT, liver, and skeletal muscle. Data are expressed as mean ± SEM. *P < 0.05 vs. WT.

Close modal

HFD-Fed Nrp1myel-KO Mice Display Greater Systemic Inflammation

Obesity promotes the induction of a proinflammatory state and the accumulation of macrophages in adipose tissue (4,26). To investigate the potential inflammatory changes in WT and Nrp1myel-KO mice fed chow or the HFD, we analyzed the gene expression profile of WAT, liver, and skeletal muscle by quantitative RT-PCR. The data demonstrated that upon chow feeding, Nrp1myel-KO mice exhibited no significant difference with respect to inflammation profiles of WAT, liver, and skeletal muscle relative to these tissues of WT mice (Supplementary Fig. 5C–E). However, increased expression of genes encoding molecules typically linked to the development of insulin resistance (e.g., inducible nitric oxide synthase [iNOS], interleukin [IL]-13Rα1, Ccl2, and arginase II) were observed in HFD-fed Nrp1myel-KO mice (Fig. 3A–C). In addition, data from immunohistochemistry analysis of CD68 revealed that the number of macrophages significantly increased in WAT and liver of HFD-fed Nrp1myel-KO mice compared with the WT control group (Fig. 3D and E). In contrast, the number of T cells and dendritic cells was comparable in WAT of both mouse groups (Supplementary Fig. 6A and B). We also found that serum levels of TNF-α, IL-6, and IL-1β were dramatically increased in HFD-fed Nrp1myel-KO mice (Fig. 3F–H). As a result of the association of liver steatosis with inflammation, we examined the accumulation fat in the liver. Comparable levels of fat accumulation were detected within both mouse groups (Supplementary Fig. 7). Together, these findings indicate enhanced systemic inflammation in Nrp1myel-KO mice in response to the HFD.

Figure 3

HFD-fed Nrp1myel-KO mice show greater systemic inflammation. Relative mRNA levels of inflammatory genes in WAT (A), liver (B), and skeletal muscle (C) from HFD-fed WT (n = 8) and Nrp1myel-KO (n = 9) mice. D: Immunohistochemistry staining of CD68+ cells in visceral WAT from HFD-fed WT and Nrp1myel-KO mice (n = 7 mice per group). Scale bar, 100 μm. E: Immunofluorescence staining of CD68+ cells in liver from HFD-fed WT and Nrp1myel-KO mice (n = 6 mice per group). Nuclei were stained with DAPI (blue). Scale bar, 50 μm. Serum levels of TNFα (F), IL-6 (G), and IL-1β (H) in HFD-fed WT and Nrp1myel-KO mice (n = 6 mice per group). Data are expressed as mean ± SEM. *P < 0.05 vs. WT.

Figure 3

HFD-fed Nrp1myel-KO mice show greater systemic inflammation. Relative mRNA levels of inflammatory genes in WAT (A), liver (B), and skeletal muscle (C) from HFD-fed WT (n = 8) and Nrp1myel-KO (n = 9) mice. D: Immunohistochemistry staining of CD68+ cells in visceral WAT from HFD-fed WT and Nrp1myel-KO mice (n = 7 mice per group). Scale bar, 100 μm. E: Immunofluorescence staining of CD68+ cells in liver from HFD-fed WT and Nrp1myel-KO mice (n = 6 mice per group). Nuclei were stained with DAPI (blue). Scale bar, 50 μm. Serum levels of TNFα (F), IL-6 (G), and IL-1β (H) in HFD-fed WT and Nrp1myel-KO mice (n = 6 mice per group). Data are expressed as mean ± SEM. *P < 0.05 vs. WT.

Close modal

Priming and Activation of Nlrp3 Inflammasome Are Enhanced in HFD-Fed Nrp1myel-KO Mice In Vivo

Inflammasomes were assessed in tissues to explore potential involvement of inflammasomes in the heightened systemic inflammation of Nrp1myel-KO mice. As shown in Fig. 4A, the expression of several inflammasomes, such as Nlrp1a, absent in melanoma 2 (AIM2), and Nlrc4, was similar in WAT of WT and Nrp1myel-KO mice. Moreover, relative mRNA levels of inflammasome ASC adaptor and caspase 11 effector were comparable in WAT of both groups. However, relative mRNA levels of Nlrp3, caspase 1, IL-1β, and IL-18 were higher in WAT of Nrp1myel-KO mice compared with WT littermates (Fig. 4A). In parallel, protein levels of Nlrp3, cleaved caspase 1 p20, pro–IL-1β, IL-1β p17, and IL-18 were profoundly elevated in WAT of Nrp1myel-KO mice compared with WT littermates (Fig. 4B). Importantly, we also observed elevated priming and activation of Nlrp3 inflammasome in the liver and skeletal muscle of Nrp1myel-KO mice (Fig. 4C–F). Taken together, myeloid cell–specific Nrp1 deficiency results in increased Nlrp3 inflammasome priming and activation in vivo.

Figure 4

Priming and activation of Nlrp3 inflammasome are enhanced in HFD-fed Nrp1myel-KO mice in vivo. A: Relative mRNA levels of Nlrp3, caspase 1, ASC, IL-1β, IL-18, Nlrp1a, AIM2, Nlrc4, and caspase 11 in visceral WAT of HFD-fed WT and Nrp1myel-KO mice. B: Immunoblot analysis of Nlrp3, cleaved caspase 1 p20, ASC, pro IL-1β, IL-1β p17, IL-18, Nlrc4, and AIM2 in visceral WAT of HFD-fed WT and Nrp1myel-KO mice. C: Relative mRNA levels of Nlrp3, caspase 1, ASC, IL-1β, IL-18, Nlrp1a, AIM2, Nlrc4, and caspase 11 in liver of HFD-fed WT and Nrp1myel-KO mice. D: Immunoblot analysis of Nlrp3, cleaved caspase 1 p20, pro IL-1β, and IL-18 in liver of HFD-fed WT and Nrp1myel-KO mice. E: Relative mRNA levels of Nlrp3, caspase 1, ASC, IL-1β, IL-18, Nlrp1a, AIM2, Nlrc4, and caspase 11 in skeletal muscle of HFD-fed WT and Nrp1myel-KO mice. F: Immunoblot analysis of Nlrp3, pro IL-1β, IL-1β p17, and IL-18 in skeletal muscle of HFD-fed WT and Nrp1myel-KO mice. β-Actin served as a loading control. Data are expressed as mean ± SEM. *P < 0.05 vs. WT.

Figure 4

Priming and activation of Nlrp3 inflammasome are enhanced in HFD-fed Nrp1myel-KO mice in vivo. A: Relative mRNA levels of Nlrp3, caspase 1, ASC, IL-1β, IL-18, Nlrp1a, AIM2, Nlrc4, and caspase 11 in visceral WAT of HFD-fed WT and Nrp1myel-KO mice. B: Immunoblot analysis of Nlrp3, cleaved caspase 1 p20, ASC, pro IL-1β, IL-1β p17, IL-18, Nlrc4, and AIM2 in visceral WAT of HFD-fed WT and Nrp1myel-KO mice. C: Relative mRNA levels of Nlrp3, caspase 1, ASC, IL-1β, IL-18, Nlrp1a, AIM2, Nlrc4, and caspase 11 in liver of HFD-fed WT and Nrp1myel-KO mice. D: Immunoblot analysis of Nlrp3, cleaved caspase 1 p20, pro IL-1β, and IL-18 in liver of HFD-fed WT and Nrp1myel-KO mice. E: Relative mRNA levels of Nlrp3, caspase 1, ASC, IL-1β, IL-18, Nlrp1a, AIM2, Nlrc4, and caspase 11 in skeletal muscle of HFD-fed WT and Nrp1myel-KO mice. F: Immunoblot analysis of Nlrp3, pro IL-1β, IL-1β p17, and IL-18 in skeletal muscle of HFD-fed WT and Nrp1myel-KO mice. β-Actin served as a loading control. Data are expressed as mean ± SEM. *P < 0.05 vs. WT.

Close modal

Nrp1 Inhibits the Priming and Activation of Nlrp3 Inflammasome in Macrophages In Vitro

Nlrp3 inflammasome function involves a twofold process: an initial priming phase and subsequent inflammasome activation (27). We investigated the effect of Nrp1 on LPS-induced Nlrp3 priming. As shown in Fig. 5A–D, Nrp1 deficiency markedly enhanced LPS-induced mRNA expression of Nlrp3, caspase 1, IL-1β, and IL-18. In line with these results, immunoblot analysis confirmed that pro–IL-1β, cleaved IL-1β p17, IL-18, Nlrp3, and cleaved caspase 1 p20 were substantially higher in LPS-primed Nrp1−/− macrophages than those in WT cells, whereas ASC was not differentially modulated in the two genotypes (Fig. 5E and F). In contrast, LPS-induced mRNA expression of Nlrp3, IL-1β, and IL-18 was downregulated in RAW264.7 cells upon overexpression of Nrp1 (Supplementary Fig. 8A–C). Moreover, Nrp1 overexpression inhibited LPS-induced pro–IL-1β, cleaved IL-1β p17, and IL-18 protein in RAW264.7 macrophages (Supplementary Fig. 8D).

Figure 5

Nrp1 deletion promotes Nlrp3 inflammasome priming and activation in macrophages in vitro. WT and Nrp1−/− BMDMs were treated with or without 100 ng/mL LPS for 4 h, and then relative mRNA levels of Nlrp3 (A), caspase 1 (B), IL-1β (C), and IL-18 (D) were analyzed by quantitative RT-PCR. E and F: WT and Nrp1−/− BMDMs were incubated with indicated concentrations of LPS for 4 h before cell extracts were prepared and immunoblotted with the indicated antibodies. G: WT and Nrp1−/− BMDMs were primed with or without 100 ng/mL LPS for 3 h and then treated with 5 mmol/L ATP for 30 min or 20 μmol/L nigericin for 30 min. Cell lysates were analyzed by immunoblot for the indicated antibodies. H: Resting or 100 ng/mL LPS-primed WT and Nrp1−/− BMDMs were stimulated with BSA-PA as indicated for 24 h before cell lysates and supernatants were analyzed by immunoblot for the indicated antibodies. β-Actin served as a loading control. I: Representative images of immunofluorescent staining of ASC (green) in WT or Nrp1−/− BMDMs primed with or without 100 ng/mL LPS for 3 h and followed by 20 μmol/L nigericin treatment for 30 min. Nuclei were stained with DAPI (blue). Scale bar, 5 µm. J: ASC was immunoprecipitated (IP) from lysed unstimulated WT and Nrp1−/− BMDMs or after 100 ng/mL LPS priming for 3 h and followed by 20 μmol/L nigericin (Nig) treatment for 30 min. Data are expressed as mean ± SEM. *P < 0.05 vs. WT.

Figure 5

Nrp1 deletion promotes Nlrp3 inflammasome priming and activation in macrophages in vitro. WT and Nrp1−/− BMDMs were treated with or without 100 ng/mL LPS for 4 h, and then relative mRNA levels of Nlrp3 (A), caspase 1 (B), IL-1β (C), and IL-18 (D) were analyzed by quantitative RT-PCR. E and F: WT and Nrp1−/− BMDMs were incubated with indicated concentrations of LPS for 4 h before cell extracts were prepared and immunoblotted with the indicated antibodies. G: WT and Nrp1−/− BMDMs were primed with or without 100 ng/mL LPS for 3 h and then treated with 5 mmol/L ATP for 30 min or 20 μmol/L nigericin for 30 min. Cell lysates were analyzed by immunoblot for the indicated antibodies. H: Resting or 100 ng/mL LPS-primed WT and Nrp1−/− BMDMs were stimulated with BSA-PA as indicated for 24 h before cell lysates and supernatants were analyzed by immunoblot for the indicated antibodies. β-Actin served as a loading control. I: Representative images of immunofluorescent staining of ASC (green) in WT or Nrp1−/− BMDMs primed with or without 100 ng/mL LPS for 3 h and followed by 20 μmol/L nigericin treatment for 30 min. Nuclei were stained with DAPI (blue). Scale bar, 5 µm. J: ASC was immunoprecipitated (IP) from lysed unstimulated WT and Nrp1−/− BMDMs or after 100 ng/mL LPS priming for 3 h and followed by 20 μmol/L nigericin (Nig) treatment for 30 min. Data are expressed as mean ± SEM. *P < 0.05 vs. WT.

Close modal

We also checked the effects of Nrp1 on Nlrp3 activation. WT and Nrp1−/− BMDMs were primed, with or without LPS, and then treated with ATP or nigericin to trigger the activation of Nlrp3 inflammasome. As shown in Fig. 5G, pro–IL-1β, cleaved IL-1β p17, and IL-18 levels were markedly elevated in Nrp1−/− macrophages compared with WT macrophages. Given the report of palmitate (PA)-mediated activation of the Nlrp3 inflammasome (28), WT and Nrp1−/− BMDMs were primed with or without LPS to mimic elevated serum FFAs during obesity and then treated with BSA-PA. As shown in Fig. 5H, protein levels of pro–IL-1β and cleaved IL-1β were markedly increased in cell lysates and supernatants of Nrp1−/− macrophages relative to WT macrophages. We further examined whether Nrp1 affected ASC cytoplasmic foci formation, an upstream event of caspase-1 cleavage. Consistent with previous study (29), ASC was diffusely localized in unstimulated WT and Nrp1−/− BMDMs (Fig. 5I). However, the ASC cytoplasmic foci became obvious upon Nlrp3 activation with LPS and nigericin, (Fig. 5I). Importantly, Nrp1 deletion significantly promoted ASC foci formation (Fig. 5I). A similar pattern of Nlrp3-ASC complex formation was confirmed in WT BMDMs and markedly enhanced upon Nrp1 deletion (Fig. 5J). Taken together, these results suggest that Nrp1 deficiency promotes Nlrp3 inflammasome priming and activation in macrophages in vitro.

Activated NF-κB Signaling Contributes to Enhanced Nlrp3 Priming in Nrp1-Deficient Macrophages

The essential role of the proinflammatory NF-κB pathway in priming the Nlrp3 inflammasome has been established (30). To determine whether increased Nlrp3 priming in Nrp1-deficient macrophages was dependent on the NF-κB pathway, we investigated potential functions of Nrp1 in NF-κB signaling. After LPS stimulation, Nrp1−/− BMDMs exhibited enhanced phosphorylation of p65 and p105 compared with WT BMDMs (Fig. 6A). However, no significant difference in c-Jun N-terminal kinase or extracellular signal–regulated kinase 1/2 phosphorylation was observed between both groups (Fig. 6A). In line with these data, transcription factor NF-κB p65 activity detected by luminescence assay was significantly higher in Nrp1−/− BMDMs than that in WT BMDMs (Fig. 6B). Next, we overexpressed Nrp1 in RAW264.7 cells to test direct roles of Nrp1 in the LPS-mediated signaling pathway. As shown in Fig. 6C, Nrp1 overexpression significantly inhibited LPS-induced p-IκBα (inhibitor of κBα) and iNOS levels in macrophages, suggesting that Nrp1 functions as an endogenous suppressor of LPS–NF-κB signaling. Finally, blockade of the NF-κB pathway could reverse Nrp1 knockdown-elevated Nlrp3, caspase 1 p20, pro–IL-1β, and cleaved IL-1β p17 levels in macrophages (Fig. 6D). Taken together, these data indicate that activated NF-κB signaling contributes to Nrp1 deficiency–induced Nlrp3 priming.

Figure 6

Activated NF-κB signaling contributes to Nrp1 deficiency–induced Nlrp3 priming. A: Immunoblot analysis of NF-κB and mitogen-activated protein kinase signaling molecules was performed in WT and Nrp1−/− BMDMs treated with 100 ng/mL LPS for 0, 15, 30, or 60 min. B: WT and Nrp1−/− BMDMs were treated with or without 100 ng/mL LPS for 4 h. Cells were lysed by radioimmunoprecipitation assay and cell lysates were subjected to transcription factor NF-κB p65 detection. C: RAW264.7 cells were transfected with or without Nrp1 plasmid for 48 h and then treated with 100 ng/mL LPS for 6 h. Immunoblot analysis was used to determine p-IκBα (Ser32) and iNOS. D: RAW264.7 cells were transfected with scramble small interfering (si)RNA or Nrp1 siRNA for 48 h, pretreated with or without NF-κB inhibitor (10 μmol/L) for 30 min, and then treated with or without LPS (200 ng/mL) for 4 h. Cell lysates were analyzed with immunoblot for the indicated antibodies. β-Actin served as a loading control. ERK, extracellular signal–regulated kinase; JNK, c-Jun N-terminal kinase. Data are expressed as mean ± SEM. *P < 0.05 vs. WT.

Figure 6

Activated NF-κB signaling contributes to Nrp1 deficiency–induced Nlrp3 priming. A: Immunoblot analysis of NF-κB and mitogen-activated protein kinase signaling molecules was performed in WT and Nrp1−/− BMDMs treated with 100 ng/mL LPS for 0, 15, 30, or 60 min. B: WT and Nrp1−/− BMDMs were treated with or without 100 ng/mL LPS for 4 h. Cells were lysed by radioimmunoprecipitation assay and cell lysates were subjected to transcription factor NF-κB p65 detection. C: RAW264.7 cells were transfected with or without Nrp1 plasmid for 48 h and then treated with 100 ng/mL LPS for 6 h. Immunoblot analysis was used to determine p-IκBα (Ser32) and iNOS. D: RAW264.7 cells were transfected with scramble small interfering (si)RNA or Nrp1 siRNA for 48 h, pretreated with or without NF-κB inhibitor (10 μmol/L) for 30 min, and then treated with or without LPS (200 ng/mL) for 4 h. Cell lysates were analyzed with immunoblot for the indicated antibodies. β-Actin served as a loading control. ERK, extracellular signal–regulated kinase; JNK, c-Jun N-terminal kinase. Data are expressed as mean ± SEM. *P < 0.05 vs. WT.

Close modal

Nlrp3 Deletion Rescues Insulin Resistance in HFD-Fed Nrp1myel-KO Mice

To test whether excessive activation of Nlrp3 inflammasome was a major contributor of insulin resistance in HFD-fed Nrp1myel-KO mice, Nlrp3−/− mice were crossed with Nrp1myel-KO mice to generate Nrp1myel-KONlrp3−/− mice. Nrp1myel-KONlrp3+/+ (Nrp1myel-KO) and Nrp1myel-KONlrp3−/− mice were fed the HFD for 16 weeks. Body weight was comparable in both groups (Fig. 7A). However, glucose tolerance and insulin sensitivity detected by GTT and ITT were improved in Nrp1myel-KONlrp3−/− mice compared with those in Nrp1myel-KONlrp3+/+ mice (Fig. 7B and C). Moreover, the improvement in glucose homeostasis of Nrp1myel-KONlrp3−/− mice fed the HFD was also validated by the lower fasting serum insulin concentrations and reduced HOMA-IR index in Nrp1myel-KONlrp3−/− mice compared with those in their Nrp1myel-KONlrp3+/+ littermates (Fig. 7D and E). However, the level of serum FFAs was not different between HFD-fed Nrp1myel-KONlrp3+/+ and Nrp1myel-KONlrp3−/− mice (Fig. 7F).

Figure 7

Nlrp3 deletion rescues insulin resistance in HFD-fed Nrp1myel-KO mice. Nrp1myel-KONlrp3+/+ and Nrp1myel-KONlrp3−/− mice were fed the HFD for 16 weeks. A: Body weight. B and C: GTT and ITT (n = 7 mice per group). D and E: Fasting serum insulin concentrations and HOMA-IR index. F: Serum levels of FFAs (n = 7 mice per group). G: Immunoblot analysis of p-AKT (Ser473) and total AKT in WAT, liver, and skeletal muscle. Serum levels of TNF-α (H), IL-6 (I), and IL-1β (J) (n = 6–7 mice per group). K: Schematic illustration of Nrp1-limited dietary insulin resistance. HFD-instigated Nrp1 attenuation in macrophages exaggerates Nlrp3 inflammasome via promoting NF-κB–mediated Nlrp3 priming and Nlrp3-ASC inflammasome assembly, which consequently increases systemic inflammation and triggers insulin resistance. Data are expressed as mean ± SEM. *P < 0.05 vs. Nrp1myel-KONlrp3+/+.

Figure 7

Nlrp3 deletion rescues insulin resistance in HFD-fed Nrp1myel-KO mice. Nrp1myel-KONlrp3+/+ and Nrp1myel-KONlrp3−/− mice were fed the HFD for 16 weeks. A: Body weight. B and C: GTT and ITT (n = 7 mice per group). D and E: Fasting serum insulin concentrations and HOMA-IR index. F: Serum levels of FFAs (n = 7 mice per group). G: Immunoblot analysis of p-AKT (Ser473) and total AKT in WAT, liver, and skeletal muscle. Serum levels of TNF-α (H), IL-6 (I), and IL-1β (J) (n = 6–7 mice per group). K: Schematic illustration of Nrp1-limited dietary insulin resistance. HFD-instigated Nrp1 attenuation in macrophages exaggerates Nlrp3 inflammasome via promoting NF-κB–mediated Nlrp3 priming and Nlrp3-ASC inflammasome assembly, which consequently increases systemic inflammation and triggers insulin resistance. Data are expressed as mean ± SEM. *P < 0.05 vs. Nrp1myel-KONlrp3+/+.

Close modal

Next, we measured the p-AKT (Ser473) in WAT, liver, and skeletal muscle of HFD-fed Nrp1myel-KONlrp3+/+ and Nrp1myel-KONlrp3−/− mice. Consistent with improved insulin sensitivity of HFD-fed Nrp1myel-KONlrp3−/− mice, there was an increased p-AKT in WAT, liver, and skeletal muscle of HFD-fed Nrp1myel-KONlrp3−/− mice compared with that in HFD-fed Nrp1myel-KONlrp3+/+ controls (Fig. 7G).

Eventually, we detected the secretary proinflammatory cytokines in serum by ELISA. As shown in Fig. 7H–J, the levels of TNF-α, IL-6, and IL-1β were dramatically decreased in the serum of Nrp1myel-KONlrp3−/− mice compared with that in Nrp1myel-KONlrp3+/+ mice, respectively. Taken together, these data indicate that loss of Nlrp3 rescues dysregulated glucose homeostasis and insulin sensitivity during obesity in Nrp1myel-KO mice, suggesting that the macrophage Nlrp3 pathway is at least partly responsible for HFD-induced insulin resistance in the mice.

In this study, we have uncovered a novel role for macrophage Nrp1 in repressing HFD-induced insulin resistance by suppressing Nlrp3 inflammasome priming and activation. Nrp1 expression in macrophages was markedly downregulated in HFD-induced mouse obesity. Importantly, the myeloid cell–specific Nrp1 deletion (Nrp1myel-KO) in mice accentuated HFD-induced aberrant activation of Nlrp3, systemic inflammation, and insulin resistance. Finally, deletion of Nlrp3 was observed to markedly protect against both diet-induced inflammation and insulin resistance in Nrp1myel-KO mice. Collectively, our data demonstrate that Nrp1 deficiency–accentuated Nlrp3 inflammasome in mouse models and macrophages is mediated by two interrelated mechanisms: NF-κB–mediated Nlrp3 priming and Nlrp3-ASC assembly. HFD-induced Nrp1 attenuation in macrophages promotes insulin resistance via Nlrp3-mediated chronic inflammation (Fig. 7K).

Nrp1 is a coreceptor for class 3 semaphorins, including Sema3A, B, C, D, E, and F, which possess axon guidance cue functions in neuronal cells (14). Nrp1 has been shown to promote endothelial tip rather than stalk cell function during sprouting angiogenesis (31). In this study, we have for the first time demonstrated a novel role for macrophage Nrp1 in attenuating insulin resistance in HFD-induced obesity. We found that Nrp1 levels were significantly decreased in macrophages of obese mice. Furthermore, in response to the HFD, Nrp1myel-KO mice displayed impaired glucose homeostasis and blunted insulin sensitivity relative to WT mice. Unlike previous studies that majorly showed the involvement of macrophage Nrp1 in the regulation of tumor growth (19,20), our current study demonstrates the causative role of macrophage Nrp1 in HFD-induced insulin resistance. Our results are in line with other reports in which Sema3 family members and their cognate receptors have been linked to insulin resistance. For example, Shimizu et al. (9) described that Sema3E and its plexinD1 receptor are markedly increased in adipose tissue of obese mice. Sema3E was shown to function as a chemoattractant for plexinD1-expressing macrophages, resulting in the infiltration and accumulation of macrophages in adipose tissue and consequent adipose inflammation and insulin resistance (9). Another neuroimmune guidance cue, netrin-1 and its receptor Unc5b, have been shown to be upregulated in ATMs of obese human visceral adipose tissue (10). Netrin-1 promotes macrophage retention in adipose tissue via Unc5b and exaggerates metabolic dysfunction (10). Sema3A was previously identified as a chemoattractant for cortical apical dendrites (32) and regulates neuronal polarization by inhibiting axon formation and increasing dendrite growth (33). Recently, Casazza et al. (18) demonstrated that Sema3A is a chemoattractant for macrophages and that Nrp1 deletion significantly inhibits Sema3A-induced macrophage migration. Nrp1-dependent myeloid cell chemoattraction was also confirmed in neovascular retinal disease (34). Interestingly, we observed that myeloid cell–specific Nrp1 deficiency contributed to a higher accumulation of macrophages in WAT and liver of Nrp1myel-KO mice.

One of the most important findings of this study is the novel function of Nrp1 as an important negative regulator of the NF-κB pathway. We found that Nrp1 deletion correlates with enhanced NF-κB activation, which in turn exerts proinflammatory effects through a Nlrp3 inflammasome-dependent and -independent manner. Nrp1 deletion–induced activation of NF-κB promotes Nlrp3 inflammasome via Nlrp3 priming; that is, NF-κB activation by Nrp1 loss increases the levels of inflammasome precursors. In addition, Nrp1 deletion promotes Nlrp3-ASC inflammasome assembly. Although the exact mechanisms are unclear, we have observed that Nrp1 deficiency results in attenuated LPS-induced 26S proteasome activation (data not shown). Because HOIL-1L is required for the assembly of Nlrp3-ASC inflammasome (29), we speculate that Nrp1 deletion induces proteasome inhibition, thereby delaying HOIL-1L degradation. Consequently, accumulated HOIL-1L promotes aberrant assembly of Nlrp3-ASC inflammasome. Further investigation is warranted to further delineate this complex and dynamic process.

Another key finding of this study is that Nlrp3 inflammasome is responsible for accentuated insulin resistance in HFD-fed Nrp1myel-KO mice. The critical role of Nlrp3 within the condition was substantiated using Nlrp3-deficient/Nrp1myel-KO mice. Marked protection against HFD-induced insulin resistance and decreased systemic inflammation was demonstrated in the double-KO mice. Together, these results reveal that Nrp1 may act as a critical endogenous inhibitor of Nlrp3 inflammasome. A published report by Wen et al. (28) has established a role of Nlrp3 inflammasome in HFD-induced insulin resistance. The authors showed that Nlrp3−/− or Pycard−/− mice exhibit improved insulin sensitivity compared with WT controls (28). Consistent with these observations, we found that Nlrp3 deficiency reversed HFD-induced insulin resistance in Nrp1myel-KO mice, suggesting that the macrophage Nlrp3 pathway is at least partly responsible for HFD-induced insulin resistance in the mice. However, we cannot exclude the possibilities that Nlrp3-independent pathways may also contribute to insulin resistance in HFD-fed Nrp1myel-KO mice.

Although this study provides insight on novel metabolic Nrp1 functions, a puzzling phenomenon remains unresolved about why Nrp1 levels become downregulated in macrophages but not in endothelial cells, T cells, or dendritic cells of HFD-fed obese mice. Recent studies have shown that serum FFAs are significantly increased during obesity and activate the proinflammatory NF-κB pathway (35) through fetuin-A, an endogenous ligand of Toll-like receptor 4 (36). Moreover, NF-κB p65 and p50 translocate to the nucleus upon activation and suppress the Nrp1 promoter in BMDM/macrophage precursor cells (37). We speculate that obesity may downregulate Nrp1 expression through activation of NF-κB signaling. However, future studies will be required to dissect whether obesity mediates Nrp1 regulation via other mechanisms. Potential consequences of Nrp1 with respect to additional obesity-related disease conditions, such as diabetes, should provide useful information on critical Nrp1 functions in metabolic disease.

In conclusion, our study identifies a novel function of Nrp1 as a negative regulator of Nlrp3 inflammasome in macrophages during obesity, suggesting potential therapeutic targeting of Nrp1 in chronic inflammation and insulin resistance.

Funding. This study was partly supported by grants from the National Heart, Lung, and Blood Institute (HL-079584, HL-080499, HL-089920, HL-110488, HL-128014, and HL-132500), National Cancer Institute (CA-213022), and National Institute on Aging (AG-047776). M.-H.Z. is an eminent scholar of Georgia Research Alliance.

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

Author Contributions. X.D., I.O., P.S., and M.-H.Z. conceived experiments and wrote the manuscript. X.D., Z.L., T.B., Q.W., T.R., and M.Z. performed experiments and analyzed data. All authors had final approval of the submitted and published versions. M.-H.Z. 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.

1.
Johnson
AM
,
Olefsky
JM
.
The origins and drivers of insulin resistance
.
Cell
2013
;
152
:
673
684
[PubMed]
2.
Nishimura
S
,
Manabe
I
,
Nagasaki
M
, et al
.
CD8+ effector T cells contribute to macrophage recruitment and adipose tissue inflammation in obesity
.
Nat Med
2009
;
15
:
914
920
[PubMed]
3.
Weisberg
SP
,
McCann
D
,
Desai
M
,
Rosenbaum
M
,
Leibel
RL
,
Ferrante
AW
 Jr
.
Obesity is associated with macrophage accumulation in adipose tissue
.
J Clin Invest
2003
;
112
:
1796
1808
[PubMed]
4.
Xu
H
,
Barnes
GT
,
Yang
Q
, et al
.
Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance
.
J Clin Invest
2003
;
112
:
1821
1830
[PubMed]
5.
Hotamisligil
GS
,
Shargill
NS
,
Spiegelman
BM
.
Adipose expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance
.
Science
1993
;
259
:
87
91
[PubMed]
6.
Weisberg
SP
,
Hunter
D
,
Huber
R
, et al
.
CCR2 modulates inflammatory and metabolic effects of high-fat feeding
.
J Clin Invest
2006
;
116
:
115
124
[PubMed]
7.
Arkan
MC
,
Hevener
AL
,
Greten
FR
, et al
.
IKK-beta links inflammation to obesity-induced insulin resistance
.
Nat Med
2005
;
11
:
191
198
[PubMed]
8.
Cai
D
,
Yuan
M
,
Frantz
DF
, et al
.
Local and systemic insulin resistance resulting from hepatic activation of IKK-beta and NF-kappaB
.
Nat Med
2005
;
11
:
183
190
[PubMed]
9.
Shimizu
I
,
Yoshida
Y
,
Moriya
J
, et al
.
Semaphorin3E-induced inflammation contributes to insulin resistance in dietary obesity
.
Cell Metab
2013
;
18
:
491
504
[PubMed]
10.
Ramkhelawon
B
,
Hennessy
EJ
,
Ménager
M
, et al
.
Netrin-1 promotes adipose tissue macrophage retention and insulin resistance in obesity
.
Nat Med
2014
;
20
:
377
384
[PubMed]
11.
He
Z
,
Tessier-Lavigne
M
.
Neuropilin is a receptor for the axonal chemorepellent semaphorin III
.
Cell
1997
;
90
:
739
751
[PubMed]
12.
Kolodkin
AL
,
Levengood
DV
,
Rowe
EG
,
Tai
YT
,
Giger
RJ
,
Ginty
DD
.
Neuropilin is a semaphorin III receptor
.
Cell
1997
;
90
:
753
762
[PubMed]
13.
Soker
S
,
Takashima
S
,
Miao
HQ
,
Neufeld
G
,
Klagsbrun
M
.
Neuropilin-1 is expressed by endothelial and tumor cells as an isoform-specific receptor for vascular endothelial growth factor
.
Cell
1998
;
92
:
735
745
[PubMed]
14.
Kumanogoh
A
,
Kikutani
H
.
Immunological functions of the neuropilins and plexins as receptors for semaphorins
.
Nat Rev Immunol
2013
;
13
:
802
814
[PubMed]
15.
Tordjman
R
,
Lepelletier
Y
,
Lemarchandel
V
, et al
.
A neuronal receptor, neuropilin-1, is essential for the initiation of the primary immune response
.
Nat Immunol
2002
;
3
:
477
482
[PubMed]
16.
Bruder
D
,
Probst-Kepper
M
,
Westendorf
AM
, et al
.
Neuropilin-1: a surface marker of regulatory T cells
.
Eur J Immunol
2004
;
34
:
623
630
[PubMed]
17.
Delgoffe
GM
,
Woo
SR
,
Turnis
ME
, et al
.
Stability and function of regulatory T cells is maintained by a neuropilin-1-semaphorin-4a axis
.
Nature
2013
;
501
:
252
256
[PubMed]
18.
Casazza
A
,
Laoui
D
,
Wenes
M
, et al
.
Impeding macrophage entry into hypoxic tumor areas by Sema3A/Nrp1 signaling blockade inhibits angiogenesis and restores antitumor immunity
.
Cancer Cell
2013
;
24
:
695
709
[PubMed]
19.
Miyauchi
JT
,
Chen
D
,
Choi
M
, et al
.
Ablation of neuropilin 1 from glioma-associated microglia and macrophages slows tumor progression
.
Oncotarget
2016
;
7
:
9801
9814
[PubMed]
20.
Wallerius
M
,
Wallmann
T
,
Bartish
M
, et al
.
Guidance molecule SEMA3A restricts tumor growth by differentially regulating the proliferation of tumor-associated macrophages
.
Cancer Res
2016
;
76
:
3166
3178
[PubMed]
21.
Takahashi
N
,
Kawada
T
,
Goto
T
, et al
.
Abietic acid activates peroxisome proliferator-activated receptor-gamma (PPARgamma) in RAW264.7 macrophages and 3T3-L1 adipocytes to regulate gene expression involved in inflammation and lipid metabolism
.
FEBS Lett
2003
;
550
:
190
194
[PubMed]
22.
Bruscia
EM
,
Zhang
PX
,
Satoh
A
, et al
.
Abnormal trafficking and degradation of TLR4 underlie the elevated inflammatory response in cystic fibrosis
.
J Immunol
2011
;
186
:
6990
6998
[PubMed]
23.
Matthews
DR
,
Hosker
JP
,
Rudenski
AS
,
Naylor
BA
,
Treacher
DF
,
Turner
RC
.
Homeostasis model assessment: insulin resistance and beta-cell function from fasting plasma glucose and insulin concentrations in man
.
Diabetologia
1985
;
28
:
412
419
[PubMed]
24.
Riordan
NH
,
Ichim
TE
,
Min
WP
, et al
.
Non-expanded adipose stromal vascular fraction cell therapy for multiple sclerosis
.
J Transl Med
2009
;
7
:
29
[PubMed]
25.
Schipper
HS
,
Prakken
B
,
Kalkhoven
E
,
Boes
M
.
Adipose tissue-resident immune cells: key players in immunometabolism
.
Trends Endocrinol Metab
2012
;
23
:
407
415
[PubMed]
26.
Wellen
KE
,
Hotamisligil
GS
.
Obesity-induced inflammatory changes in adipose tissue
.
J Clin Invest
2003
;
112
:
1785
1788
[PubMed]
27.
Lamkanfi
M
,
Dixit
VM
.
Mechanisms and functions of inflammasomes
.
Cell
2014
;
157
:
1013
1022
[PubMed]
28.
Wen
H
,
Gris
D
,
Lei
Y
, et al
.
Fatty acid-induced NLRP3-ASC inflammasome activation interferes with insulin signaling
.
Nat Immunol
2011
;
12
:
408
415
[PubMed]
29.
Rodgers
MA
,
Bowman
JW
,
Fujita
H
, et al
.
The linear ubiquitin assembly complex (LUBAC) is essential for NLRP3 inflammasome activation
.
J Exp Med
2014
;
211
:
1333
1347
[PubMed]
30.
Bauernfeind
FG
,
Horvath
G
,
Stutz
A
, et al
.
Cutting edge: NF-kappaB activating pattern recognition and cytokine receptors license NLRP3 inflammasome activation by regulating NLRP3 expression
.
J Immunol
2009
;
183
:
787
791
[PubMed]
31.
Fantin
A
,
Vieira
JM
,
Plein
A
, et al
.
NRP1 acts cell autonomously in endothelium to promote tip cell function during sprouting angiogenesis
.
Blood
2013
;
121
:
2352
2362
[PubMed]
32.
Polleux
F
,
Morrow
T
,
Ghosh
A
.
Semaphorin 3A is a chemoattractant for cortical apical dendrites
.
Nature
2000
;
404
:
567
573
[PubMed]
33.
Shelly
M
,
Cancedda
L
,
Lim
BK
, et al
.
Semaphorin3A regulates neuronal polarization by suppressing axon formation and promoting dendrite growth
.
Neuron
2011
;
71
:
433
446
[PubMed]
34.
Dejda
A
,
Mawambo
G
,
Cerani
A
, et al
.
Neuropilin-1 mediates myeloid cell chemoattraction and influences retinal neuroimmune crosstalk
.
J Clin Invest
2014
;
124
:
4807
4822
[PubMed]
35.
Boden
G
,
She
P
,
Mozzoli
M
, et al
.
Free fatty acids produce insulin resistance and activate the proinflammatory nuclear factor-kappaB pathway in rat liver
.
Diabetes
2005
;
54
:
3458
3465
[PubMed]
36.
Pal
D
,
Dasgupta
S
,
Kundu
R
, et al
.
Fetuin-A acts as an endogenous ligand of TLR4 to promote lipid-induced insulin resistance
.
Nat Med
2012
;
18
:
1279
1285
[PubMed]
37.
Hayashi
M
,
Nakashima
T
,
Taniguchi
M
,
Kodama
T
,
Kumanogoh
A
,
Takayanagi
H
.
Osteoprotection by semaphorin 3A
.
Nature
2012
;
485
:
69
74
[PubMed]
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 http://www.diabetesjournals.org/content/license.

Supplementary data