Abstract
Many metabolic diseases, including atherosclerosis, type 2 diabetes, pulmonary alveolar proteinosis (PAP), and obesity, have a chronic inflammatory component involving both innate and adaptive immunity. Mice lacking the ATP binding cassette (ABC) transporter ABCG1 develop chronic inflammation in the lungs, associated with lipid accumulation (cholesterol, cholesterol ester, and phospholipid) and cholesterol crystal deposition, characteristic of atherosclerotic lesions and PAP. Here we demonstrate that specific lipids, likely oxidized (Ox) phospholipids and/or sterols, elicit a lung-specific immune response in Abcg1−/− mice. Loss of ABCG1 results in increased levels of specific oxysterols, phosphatidylcholines and oxidized phospholipids, including 1-palmitoyl-2-(5’-oxovaleroyl)-sn-glycero-3-phosphocholine (POVPC), in the lungs. Further, we identify a niche-specific increase in natural antibody (NAb)-secreting B-1 B cells in response to this lipid accumulation that is paralleled by increased titers of IgM, IgA and IgG against oxidation specific epitopes such as those on OxLDL and malondialdehyde-modified LDL (MDA-LDL). Finally we identify a cytokine/chemokine signature reflective of increased B cell activation, antibody secretion and homing. Collectively, these data demonstrate that the accumulation of lipids in Abcg1−/− mice induces the specific expansion and localization of B-1 B cells, which secrete NAbs that may help protect against the development of atherosclerosis. Indeed, despite chronic lipid accumulation and inflammation, hyperlipidemic mice lacking ABCG1 develop smaller atherosclerotic lesions compared to controls. These data also suggest that Abcg1−/− mice may represent a new model in which to study the protective functions of B-1 B cells/NAbs, and suggest novel targets for pharmacologic intervention and treatment of disease.
Keywords: ABCG1, lipid mediators, B cells, autoantibodies, inflammation, lung
Introduction
B-2 B cells are hallmark effectors of the adaptive immune response, characterized by their production of specific antibodies (1–4). However, not all antibody production is triggered by a prior exposure and immune response. B-1 B cells, the innate-immune component of the B cell lineage, are the source of natural antibodies (NAbs), which are produced in the absence of exposure to an antigen (2, 5). B-1 B cells are primarily localized to the peritoneal cavity and pleural space (5, 6). B-1 B cells have been shown to affect the progression of multiple autoimmune diseases, human B-cell leukemias, and inflammatory disorders, such as atherosclerosis (7–9). Despite the importance of B cell homeostasis in human disease, factors that regulate B cell movement into specific compartments are not well understood.
The maintenance of tissue homeostasis and defense against mucosal pathogens is highly dependent on B-1 B cells and NAb production due to a B cell receptor (BCR) repertoire that is enriched for cross-reactive receptors that bind to both self and non-self (e.g. microbial) antigens (9–11). This selection for self-reactivity is restricted to B-1 B cells and yet contradicts the current paradigm of lymphocyte selection wherein self-reactive cells undergo deletion to avoid autoimmunity. The presence of such potentially auto-reactive B cells dictates the requirement for tightly regulated mechanisms to control their activation. Increasing evidences suggests that loss of cellular lipid homeostasis plays an essential role in regulating lymphocyte and hematopoietic cell proliferation, lymphocyte movement within the follicular regions of the spleen and immune responses (12–17).
Cellular cholesterol homeostasis is influenced by a number of proteins, including the sterol ATP binding cassette transporter G1 (ABCG1) (18, 19) (20, 21). Expression of ABCG1 in intracellular vesicles stimulates activity of the transcription factor SREBP-2 via the redistribution of sterols out of the ER (20, 22), and eventually results in the removal of cholesterol from the cell to extracellular acceptors, such as high-density lipoprotein (HDL) (18, 23–25). Analysis of the alveolar macrophages and/or brains of mice lacking ABCG1 demonstrated that loss of ABCG1 was associated with a marked accumulation not only of free and esterified cholesterol, but also 24-, 25-, and/or 27-hydroxycholesterols that are synthesized enzymatically by Cyp24a1, cholesterol 25-hydroxylase (Ch25h), and Cyp27a1, respectively (26–28). In addition, the levels of 7-ketocholesterol, a product of non-enzymatic autoxidation of cholesterol, are increased in both peritoneal macrophages and the aortas of Abcg1−/− mice (29, 30). Despite recent progress in understanding how lipid homeostasis impacts lymphocyte function, little is known about how lipid metabolism impacts B cell specific responses.
Herein, we demonstrate that loss of ABCG1 results in the accumulation of specific oxidized sterols and phospholipids, eliciting a lung-specific immune response. We show a niche-specific accumulation of B-1 B cells in the pleural cavity and lungs of Abcg1−/− mice, accompanied by increased IgM, IgA, and IgG titers to oxidized lipid epitopes in both plasma and whole lung. Additionally macrophage oxysterol production drives homing of B-1 B cells specifically to the lungs and pleural cavity. Our data suggest that ABCG1-dependent control of intracellular lipid homeostasis represents a previously unrecognized mechanism for the regulation of B-1 B cell movement and homing.
Materials and Methods
Animals
All animals were bred and maintained at UCLA in temperature-controlled, pathogen-free conditions under a 12-hour light/dark cycle. Abcg1−/− LacZ knock-in mice (Deltagen, (18, 31)) were backcrossed at least 10 times onto a C57BL/6 background. Control C57BL/6 mice (originally purchased from The Jackson Laboratory) were generated from Abcg1+/− breeding. Mice were fed a chow diet, or a Western diet (Research Diets #D12079B, containing 21% fat and 0.2% cholesterol) where indicated. Mice expressing the green fluorescent protein (GFP) C57BL/6-Tg(CAG-EGFP)1Osb/J were purchased from the Jackson Laboratory (Strain #003291). The Institutional Animal Care and Research Advisory Committee at UCLA approved all experimental protocols.
Adoptive Transfer
Cells were isolated with Ab-tagged magnetic beads and Auto-MACS (Miltenyi Biotec). Peritoneal CD19+CD23− B-1 B cells were isolated from C57BL/6-Tg(CAG-EGFP)1Osb/J mice by negative selection on a CD23+ column, followed by positive selection of CD19+ cells. Cell purity (>98%) was confirmed by FACS analysis using fluorochrome-labeled CD19, CD23 and CD5 Abs (eBioscience). Cell viability (>97%) was assessed by trypan blue exclusion. To obtain 10 × 106 B-1 B cells, a pool of peritoneal cells from 20 donor mice was used, and 1 × 106 B-1 B cells were adoptively transferred into 6 month old chow-fed wildtype and Abcg1−/− mice.
Surfactant Isolation
Pulmonary surfactant was isolated from 6 month old wildtype and Abcg1−/− mice by bronchoalveolar lavage as previously described (31). Briefly, tracheas were exposed and canulated before the lungs were flushed 3 times with 1 mL aliquots of BAL buffer (10 mmol/L Tris, 100 mmol/L NaCl, 0.2 mmol/L EGTA, pH 7.2). The aliquots were combined and centrifuged (200 × g, 5 min) to separate surfactant and cells.
Lipid Analyses
Cells, bronchoalveolar lavage fluid (BAL), or lung tissue were snap frozen in liquid nitrogen. Lung tissue was homogenized on ice in phosphate buffered saline. Cell suspensions, BAL fluid, or lung homogenates were subsequently subjected to a modified Bligh-Dyer lipid extraction (32) in the presence of lipid class internal standards including eicosanoic acid, 1-0-heptadecanoyl-sn-glycero-3-phosphocholine, 1,2-dieicosanoyl-sn-glycero-3-phosphocholine, and 1,2-ditetradecanoyl-sn-glycero-3-phosphoethanolamine (33). Fatty acids were converted to their pentafluorobenzyl esters and then were subsequently quantified using GC-MS with negative ion chemical ionization with methane as the reactant gas (34). For phospholipids, lipid extracts were diluted in methanol/chloroform (4/1, v/v) and molecular species were quantified using electrospray ionization mass spectrometry on a triple quadrupole instrument (Themo Fisher Quantum Ultra) employing shotgun lipidomics methodologies (35). Phosphatidylcholine molecular species were quantified as lithiated adducts in the positive ion mode using neutral loss scanning for 59.1 amu (collision energy = −28eV). Individual molecular species were quantified by comparing the ion intensities of the individual molecular species to that of the lipid class internal standard with additional corrections for type I and type II 13C isotope effects (35).
Flow Cytometry
Single-cell suspensions from lungs and spleen were depleted of red blood cells using hypotonic lysis. Cells were resuspended in phosphate buffered saline (PBS) with 0.2% bovine serum albumin (BSA) and 0.1% sodium azide (FACS buffer). Single cell suspensions were incubated for 15 min with anti-CD16/32 (Fc block), and stained for 30 min at 4°C. DAPI, anti-mouse CD3 (17A2), CD19 (1D3), CD11b (M1/70), CD5 (53-7.3), and IgM (II/41) were purchased from eBioscience. Cells were analyzed on LSRII (Becton Dickinson). More than 0.5 × 105 cells were analyzed per sample, with dead cells excluded by DAPI positive staining. Surface marker analysis was performed using FlowJo software (Treestar Inc.). B cells (CD19+), B-1 B cells (CD19+, sIgM+, CD11b+), B-1a B cells (CD19+, sIgM+, CD11b+, CD5+), and B-1b B cells (CD19+, sIgM+, CD11b+, CD5−) were identified with the appropriate gating. For gating strategy and controls see Figure 1.
RNA isolation and analysis
Total RNA was isolated from 50 mg lung tissue (tissue weight determined after blotting of excess buffer) using Qiazol (Qiagen). Gene expression profiling of inflammatory markers was determined using GEArray mouse inflammatory cytokines and receptors microarray system (SABiosciences) as previously described (36). Total RNA (0.5 µg) was reverse transcribed with random hexamers using the Taqman Reverse Transcription Reagents Kit (Applied Biosystems). Taqman real-time quantitative PCR assays were performed using an Applied Biosystems 7900HT sequence detector. Real Time qPCR (RT-qPCR) assays were performed on a Lightcycler 480 (Roche). Results show averages of triplicate experiments normalized to GAPDH (Taqman) or 36B4 (RT-qPCR). Primer sequences are available on request.
Measurement of Ab titers
Total and specific antibody titers were determined by chemiluminescent enzyme immunoassays as previously described (37). In brief, capture antigens were coated on plates at 5 µg/mL in PBS overnight at 4°C (AB-12, CuOxLDL, IgM (Goat anti-mouse-IgM; Sigma), IgG (Goat anti-mouse-IgG; Sigma), IgA (Rat anti-mouse-IgA; Sigma), and MDA-LDL). Plates were blocked with 1% BSA in tris-buffered saline (TBS), and serially diluted antisera from individual mice was added. Plates were incubated for 1.5 h at room temperature. Bound plasma immunoglobulin isotype levels were detected with various anti-mouse immunoglobulin isotype-specific alkaline phosphatase (AP) conjugates using LumiPhos 530 (Lumigen, Southfield, MI, USA) solution, and a Dynex Luminometer (Dynex Technologies, Chantilly, VA, USA). Several secondary antibodies were used at dilutions of 1:30,000. These included alkaline phosphatase–labeled (AP-labeled) Goat-α-mouse-IgM–AP (µ-chain specific) (Sigma), Goat-α-mouse-IgG–AP (γ-chain specific) (Sigma), Goat-α-mouse-IgA-AP (α-chain specific) (Sigma). Specific controls were used for each specific antibody, and formal antibody dilution curves were determined as an initial study to identify the linear range of each antibody titer measurement. It was determined from these dilution curves that plasma samples could be optimally measured at 1:100 dilutions and lung samples at 1:10 dilutions, yielding concentrations within the linear detection range for each assay.
Where indicated antibodies were extracted from lung tissue by a method modified from Ylä-Herttuala et al. (38). Lungs were extensively perfused with physiological saline to ensure removal of all blood prior to analysis. Briefly, a small piece of lung (50 mg) was homogenized (Homogenizers POLYTRON®) on ice, in 0.5 ml PBS, pH 7.2, containing the following preservatives: 2.7 mmol/L EDTA, 2 mmol/L benzamidine, 1 mmol/L PMSF, 40 µmol/L elastatinal, 10 µmol/L probucol, 0.01% aprotinin, 0.008% chloramphenicol, 0.008% gentamicin and Protease Inhibitor Cocktail (1:100; Sigma P8340). Samples were incubated overnight at 4°C. The extract was collected by low-speed centrifugation at 4°C (30 min at 3000 RPM) and re-centrifuged for 15 min at 4,000 RPM at 4°C prior to assay.
Protein-lipid overlay analysis
Total lung lipids were isolated by Folch extraction as previously described (31) from 25 mg tissue. Total lung lipid extracts were spotted on to nitrocellulose membrane and allowed to dry. Membranes were either incubated in the presence of total lung protein extracts (15 mg tissue) or E06-IgM antibody (1:500) for 16 h at 4°C. Extracts containing antibodies were removed by extensive washing in PBS, and cross-reactivity detected using HRP-conjugated anti-mouse IgM (Invitrogen).
Immunohistochemistry
Frozen-embedded sections (lungs) from wildtype and Abcg1−/− mice were fixed in 4% PFA, blocked with 5% goat serum, and stained with either HRP-conjugated anti-mouse IgM, IgG or IgA and detected with ECL. A Vectastain ABC-Alkaline phosphatase kit (Vector Laboratories) was used to visualize the antibody staining. Where indicated, slides were counter-stained with Harris Hematoxylin (Fisher Scientific). Frozen tissue sections of lungs from wildtype and Abcg1−/− mice were also stained with antibodies that recognize CXCL13 (Genetex), oxPL (E06), B220 (B cell marker; BD Biosciences Clone RA3–6B2) and PCNA (proliferative marker; Genetex), followed by anti-mouse IgM AlexaFluor 488, anti-rat AlexaFluor 594 or anti-rabbit AlexaFluor 488 secondary antibodies (Molecular Probes, Life Sciences). Immunostaining of adjacent sections in the absence of primary antibody was used as a negative control.
TUNEL staining
The presence of apoptotic cells was assessed by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay of frozen-embedded tissue sections or primary alveolar macrophages as previously described (39).
Statistics
Lipid parameters (cholesterol, oxysterols, phosphatidylcholine, oxidized phospholipids) were analyzed by two-way ANOVA, with genotype as one factor and lipid species as another. Where there was an effect of either genotype or lipid species with no apparent interaction, data were further analyzed by post hoc Bonferroni test to determine differential effects. Absolute cell numbers (determined using flow cytometry) were analyzed by unpaired Student t test. Antibody titers were analyzed by two-way ANOVA, with genotype as one factor and antigen (MDA-LDL, Cu-OxLDL, E06/T15) as another. Where there was an effect of either genotype or lipid species with no apparent interaction, data were further analyzed by post hoc Bonferroni test to determine differential effects. Ch25h, Cyp7b1, Cxcl13, Cxcr5, Gpr183, total IgM, and E06-IgM mRNA were analyzed by unpaired Student t test.
Results
ABCG1 regulates pulmonary B cell homeostasis
To investigate the role of ABCG1 in B cell homeostasis and innate immunity, we examined specific immunological properties of 6 month old Abcg1−/− mice. Flow cytometric analysis of the spleen demonstrated no significant difference in the number of B (Figure S1A) or T (Figure S1B) cells recovered from the spleens of wildtype and Abcg1−/− mice. We previously observed gross lymphocytic infiltrates consisting predominantly of B cells in the lungs of Abcg1−/− mice (36). Consistent with these observations, FACS analysis of the lungs revealed a significant increase in the B cell population (defined as CD19+) in the lungs of chow-fed Abcg1−/− mice (Figure 2A). In contrast, there was no significant difference in the number of T cells (defined as CD3+) in the lung (Figure 2B).
B cells can be subdivided into B-1 and B-2 B cells. B-1 B cells are primarily localized to the peritoneal cavity and pleural space whereas B-2 cells are localized to the spleen (1, 4). FACS analysis of cells recovered from the lung or pleural cavity demonstrated that B-1 B cells (CD19+, sIgM+, CD11b+) were increased in Abcg1−/−, compared to wildtype mice (Figure 2C, D). In contrast, there was no difference in the number of B-1 B cells present in the spleen or peritoneal cavity (PerC) (Figure 2E). Taken together, these data support the conclusion that a global loss of ABCG1 results in a niche-specific increase in B-1 B cells in the lungs and pleural space. Interestingly there was also a marked increase in the numbers of conventional B-2 cells in the lungs, but not the spleens, of Abcg1−/− mice (Figure 2F). Since B-2 and B-1 cells are generally considered to be mediators of adaptive immunity (40) and innate immunity (6, 41) respectively, these data demonstrate that loss of ABCG1 has a broad impact on pulmonary B cell biology in mice.
Innate B-1 B cells can also be further divided into B-1a (sIgM+, CD11b+, CD5+) and B-1b (sIgM+, CD11b+, CD5−) cells. In the absence of infection, B-1a cells are the source of most serum IgM, are present in serous cavities, and are known to generate NAbs, which for example specifically bind to phosphocholine (PC) containing oxidized phospholipids (OxPL) (42). FACS analysis of cells recovered from the pleural cavity and lungs of wild type and Abcg1−/− mice shows that there is a significant increase in B-1a cells recovered from Abcg1−/− mice (Figure 2G, H). In contrast to B1-a cells, B-1b cells are thought to be the primary source of T cell-independent antibody production, and provide delayed but long-term protection against infectious pathogens (1). The finding that B-1b cells are also increased in both the lungs and pleural cavities of Abcg1−/− mice, as compared to wildtype mice (Figure 2I), provides further support for the proposal that loss of ABCG1 affects multiple arcs of immunity. These effects are niche/lung-specific since there were was no significant difference in the numbers of B-1a (Figure S1C) or B-1b (Figure S1D) cells in the spleen or peritoneal cavity of wildtype and Abcg1−/− mice.
Lipid-driven expansion of B-1 B cells in the lungs of Abcg1−/− mice
To test the hypothesis that the lung-specific expansion of B cells is dependent upon lipid accumulation, we analyzed cells isolated from lungs, spleens, peritoneal and pleural cavities of chow-fed 12 week old wildtype and Abcg1−/− mice. Importantly there was little/no lipid accumulation in the lungs of these young Abcg1−/− mice (18). Consistent with our hypothesis, in the absence of pulmonary lipid accumulation, there was no significant difference in the number of B-1 B cells in the tissues of wildtype and Abcg1−/− mice (Figure 3A). We previously demonstrated that the pulmonary lipidosis in Abcg1−/− mice can be accelerated by feeding mice a Western diet (18). Consequently, 4 week old wildtype and Abcg1−/− mice were fed a Western diet for 8 weeks to promote lipid deposition. FACS analysis of cells recovered from the pleural cavity and lungs shows that there was a significant increase in the number of B-1 B cells recovered from Abcg1−/− mice, with no change observed in spleen and peritoneal cavity (Figure 3B, C).
It is possible that there are off-target effects from feeding a Western diet. Further, it is known that Abcg1−/− mice >12 weeks old exhibit increasing pulmonary lipidosis on a chow diet (18). Therefore, to test the requirement of the pulmonary lipidosis for B-1 B cell expansion in Abcg1−/− mice, we isolated B-1 B cells from 12 week old transgenic mice expressing green fluorescent protein (GFP) under the control of the chicken β-actin promoter. These GFP+ B-1 B cells were injected (1×106 cells/mouse) intraperitoneally into 6 month old chow-fed wildtype and Abcg1−/− mice. After 10 weeks reconstitution, FACS analysis of cells recovered from lung, spleen, pleural and peritoneal cavities shows that there were significantly increased numbers of GFP+ cells specifically in the lungs and pleural cavities of Abcg1−/− mice, compared to wildtype mice (Figure 3D, E). In contrast, there was no difference in the number of GFP+ cells in either the spleens or peritoneal cavities of wildtype and Abcg1−/− mice (Figure 3E).
Accumulation of oxysterols and oxidized phospholipids in the lungs of Abcg1−/− mice
To better define the lipids that accumulate in the lungs of Abcg1−/− mice, we utilized GC/MS and ESI-MS/MS to analyze lipids extracted under conditions that minimize autoxidation. Figure 4A–F shows for the first time that both the lungs and surfactant from 6 month old chow-fed Abcg1−/− mice contain significantly increased levels of a number of enzymatically synthesized oxysterols (24-, 25- and 27-hydroxycholesterol), and autoxidation derivatives of cholesterol (7α-hydroxycholesterol, 7β-hydroxycholesterol, 3,5,6-triolcholesterol and 7-ketocholesterol), in addition to total cholesterol. Further, we demonstrate that the lungs of Abcg1−/− mice contain 3- to 6-fold higher levels of numerous phosphatidylcholine species that differ in their content of different fatty acids at the sn-1 and/or sn-2 positions (Figure 4G). Phosphatidylcholines, especially di-palmitoyl phosphatidylcholine, represent the major phospholipids in the mammalian lung (43, 44). Thus, whole body loss of ABCG1 leads to a generalized increase in all lung phosphatidylcholines, rather than affecting the metabolism of specific species.
Given the increase in autoxidation products of cholesterol in the lungs and surfactant of Abcg1−/− mice (Figure 4A, D), we used ESI-MS/MS to identify PC-containing oxidized phospholipids (OxPL), possible oxidation products of phosphatidylcholine. These analyses identified two OxPL, 1-palmitoyl-2-(5’-oxovaleroyl)-sn-glycero-3-phosphocholine (POVPC) and 1-palmitoyl-2-(9’-oxononanoyl)-sn-glycero-3-phosphocholine (PoxnoPC), which were increased 2–15 fold in the lungs of Abcg1−/− mice (Figure 4H). Interestingly, we have previously reported that POVPC and PoxnoPC are able to serve as antigens for B-1 cells (42), a cell type that has a critical role in innate immunity (1, 4).
ABCG1 regulates B-1 B cell homing
Despite chronic inflammation in the lungs of Abcg1−/− mice (18, 36), pathway analysis of inflammatory gene expression identified a signature of chemokines and cytokines, present in Abcg1−/− mice compared to wildtype animals, reflective of B cell activation and homing (Table 1). Importantly, lipids are now widely accepted to act as both signaling molecules and mediators produced locally in response to various stimuli (45). Indeed, 25-hydroxycholesterol is a metabolite of cholesterol, produced and secreted by macrophages, that has potent and diverse effects on the immune system (46). Catabolism of 25-hydroxycholesterol by the enzyme CYP7B1 results in the formation of 7α,25-dihydroxycholesterol, a ligand for the G protein-coupled receptor (GPCR) GPR183/EBI2, expressed on the surface of B cells (47). The data of Figure 4B and (27) demonstrate that 25-hydroxycholesterol levels are significantly increased in the lungs and alveolar macrophages of Abcg1−/− mice, respectively, suggesting there are enhanced B cell homing signals in the lungs of Abcg1−/− mice. Indeed, mRNA levels of the enzymes cholesterol 25-hydroxylase (Ch25h) and Cyp7b1 are significantly increased in the lungs of Abcg1−/− mice (Figure 5A).
Table 1.
Gene Symbol | Gene Description | Fold Change (Abcg1−/−/ Wildtype) |
---|---|---|
Cxcl13 | Chemokine (C-X-C motif) ligand 13 | 3.12 ± 1.23 *** |
Ifng | Interferon gamma | 21.32 ± 3.42 *** |
Il10 | Interleukin 10 | 5.48 ± 1.07 ** |
Il10ra | Interleukin 10 receptor, alpha | 4.41 ± 0.89 ** |
Il10rb | Interleukin 10 receptor, beta | 1.11 ± 0.23 |
Il12a | Interleukin 12A | 1.98 ± 0.42 * |
Il12b | Interleukin 12B | 3.13 ± 0.93 ** |
Il12rb2 | Interleukin 12 receptor, beta 2 | 10.10 ± 1.56 *** |
Il13 | Interleukin 13 | 18.41 ± 1.64 *** |
Il13ra1 | Interleukin 13 receptor, alpha 1 | 14.74 ± 1.43 *** |
Il17 | Interleukin 17 | 4.56 ± 0.76 ** |
Il17b | Interleukin 17B | 4.92 ± 0.55 ** |
Il1b | Interleukin 1 beta | 8.24 ± 1.37 *** |
Il4 | Interleukin 4 | 4.06 ± 0.69 ** |
Il5 | Interleukin 5 | 13.68 ± 1.78 *** |
Il5ra | Interleukin 5 receptor, alpha | 2.30 ± 0.34 * |
Il6 | Interleukin 6 | 4.08 ± 0.97 ** |
Tgfb1 | Transforming growth factor, beta 1 | 3.11 ± 0.85 ** |
Tnf | Tumor necrosis factor | 4.45 ± 0.79 ** |
p<0.05
p<0.01
p<0.001.
Naïve B cells, expressing the CXCL13 receptor CXCR5, migrate in response to the chemokine CXCL13, and mice lacking CXCL13 fail to migrate B cells into the peritoneal and pleural cavities (48). The data of Figure 5B (left panel) show that in young 12-week old mice, that lack significant lung lipidosis there is no change in Cxcl13 mRNA expression. In contrast, Cxcl13 mRNA levels were increased in Abcg1−/− mice fed a Western diet for 8 weeks to induce lipidosis (Figure 5B; right panel). Six month old chow-fed Abcg1−/− mice also exhibit pulmonary lipidosis (18, 31). Compared to wildtype mice, the lungs of these older, chow-fed Abcg1−/− mice also have elevated levels of Cxcl13 mRNA levels (Figure 5C). Additionally, B-1 B cells isolated from the pleural cavities of Abcg1−/− mice expressed significantly increased levels of Gpr183/EBI2 and Cxcr5 mRNA compared to cells from wildtype mice (Figure S2A), which is in agreement with increased homing of B-1 B cells to the lungs and pleural cavity. Finally, immunohistochemical analysis of the lungs of 6 month old chow-fed mice demonstrates significantly increased levels of CXCL13 in the lungs of Abcg1−/− mice, compared to wildtype animals (Figure 5D).
Increased immunoglobulins in the lungs of Abcg1−/− mice
The data of Figures 2–5 suggest that the lungs of Abcg1−/− mice may contain increased titers of antibodies that function in innate (IgM, IgA, or IgG3 from B-1 cells) and adaptive (IgG from B-2 cells) immunity. Consistent with this hypothesis, immunohistochemical analysis of the lungs and spleens of wildtype and Abcg1−/− mice demonstrate that the lungs, but not spleens, of the knockout mice contain elevated levels of IgM, IgA and IgG, with the most positive staining observed adjacent to alveolar sacs (Figure 6A; arrows). Western blot analysis confirmed that the levels of IgM and IgA were markedly increased, and IgG slightly increased in the lungs of the knockout mice (Figure 6B). Further, titers of total IgM and IgA determined by ELISA were also increased in the lungs of Abcg1−/− mice (Figure 6C). Thus, loss of ABCG1 results in increased levels of multiple immunoglobulins that function in both innate and adaptive immunity.
Loss of ABCG1 results in pulmonary accumulation of lipid antigens and specific IgM antibodies
We next performed a modification of the protein-lipid-overlay (PLO) technique (49) to determine if the lungs of Abcg1−/− mice contained increased levels of lipid antigens and corresponding immunoglobulins. Total lipids were isolated from perfused lungs of 6 month old chow-fed wildtype and Abcg1−/− mice, and spotted onto a nitrocellulose membrane. The membrane containing the bound lipids was then incubated with protein extracts from wildtype or Abcg1−/− mice before addition of anti-IgM, -IgG or -IgA HRP-conjugated secondary antibodies for detection. Figure 6D (right panels) shows that the lungs of Abcg1−/− mice, but not wildtype mice, contain IgM antibodies that bind to an endogenous lipid antigen(s) present in the lungs of knockout mice. This observation is consistent with the presence of elevated levels of POVPC and PoxnoPC (Figure 4H), known to be antigens for B-1 B cells, in the lungs of Abcg1−/− mice. In contrast, the lungs of wildtype and knockout mice contain both IgA and IgM antibodies that bind lipid antigens present only in the lungs of Abcg1−/− mice (Figure 6D).
Increased antibody titers to oxidation-specific epitopes (OSE) in the lungs of Abcg1−/− mice
The increase in specific oxidized sterols and phospholipids (Figure 4) in the lungs of Abcg1−/− mice suggested an enhanced oxidative environment and the generation of oxidation-specific epitopes (OSE). Consequently, we performed chemiluminescent ELISAs, using various antigens, to specifically determine whether the lungs of Abcg1−/− mice contained antibodies binding to OSE. Figure 7A–C shows that titers of IgM, IgA and IgG to MDA-LDL and Cu-OxLDL were increased 2.5- to 6-fold in the lungs of knockout mice. In addition, using the anti-idiotypic antibody AB1–2, which binds specifically to the E06/T15 idiotype that binds OxPL (50), we measured titers of the specific E06 IgM and T15 IgA NAbs. There were markedly increased titers of both E06 IgM (Figure 7B) and T15 IgA (Figure 7C) in the lungs of Abcg1−/− mice. We also noted markedly increased titers of IgA to the classic B-1 cell antigen α1,3-dextran in the lungs of Abcg1−/− mice. The latter finding suggests a generalized increase in B-1 cell derived IgA antibodies, which are typically mucosal, but a preferential increase in IgM NAbs to OSE. Further, the lungs of Abcg1−/− mice exhibited a 2-fold increase in total IgM mRNA but a 6-fold increase in E06-IgM-specific mRNA (Figure 7D), consistent with the idea that there are increased levels of E06-secreting B-1a B cells in the Abcg1−/− lungs. These data indicate that the lungs of Abcg1−/− mice contain increased levels of B-1a B cells (Figures 2, 3), oxidized lipid antigens (Figure 4H), IgA and IgM NAbs (Figures 6, 7) that are part of innate immunity, and IgGs that are likely part of adaptive immunity (Figures 6, 7).
Elevated plasma titers of natural and adaptive antibodies in Abcg1−/− mice
To assess whether loss of ABCG1 leads to a more global expansion of antibodies involved in innate and/or adaptive immunity, we used chemiluminescent ELISAs to measure total and OSE antibody titers in the plasma of wildtype and Abcg1−/− mice. Table 2 shows that plasma from Abcg1−/− mice contained increased titers of total IgG and IgM, increased titers of IgA and IgG to MDA-LDL, and increased IgM titers to OxLDL and α-1,3-dextran, as well as increased titers of the IgM E06 to OxPL. Interestingly, although IgA are predominantly serosal antibodies, there were also increased plasma titers of IgA to MDA-LDL in Abcg1−/− mice.
TABLE 2.
Antibody Specificity |
Antibody Class |
Wildtype (ng/mL) |
Abcg1−/− (ng/mL) |
---|---|---|---|
Total | IgA | 325000 ±37571 | 305432 ±136999 |
IgG | 343513 ±36994 | 441131 ±52656 * | |
IgM | 372777 ±50347 | 581597 ±70206 * | |
MDA-LDL | IgA | n.d. | n.d. |
IgG | 79518 ±6608 | 135757 ±8440 ** | |
IgM | 251904 ±78329 | 306314 ±58127 | |
Cu-OxLDL | IgA | n.d. | n.d. |
IgG | 59390 ±619 | 64395 ±2626 | |
IgM | 74639 ±27931 | 212358 ±25990 ** | |
E06/T15 | IgA | n/a | n/a |
IgG | n/a | n/a | |
IgM | 87647 ±14887 | 298786 ±39055 ** | |
α-1,3-dextran | IgA | n.d. | n.d. |
IgG | n/a | n/a | |
IgM | 31609 ±11698 | 45252 ±28432 |
p<0.05
p<0.01.
Selective stimulation and increased proliferation of B-1a B cells in the lungs of Abcg1−/− mice
Together, the data of Figures 2–7 and Table 2 demonstrate that loss of ABCG1 results in increased titers of NAbs in the lungs and plasma of Abcg1−/− mice, and to a niche-specific pulmonary expansion of B cells and especially B-1 B cells. However, we cannot rule out the possibility that in addition to increased homing of B-1 B cells to the lungs and pleural cavity (Figure 5, Table 1), there may also be local expansion of B-1 B cells. To directly test whether a localized increase of B-1a B cells might be related to a selective stimulation of these clones with their respective antigens, (e.g. the presence of OxPL in the lungs leading to enhanced secretion of IgM E06 and IgA T15), we performed immunohistochemical analyses of lung sections to determine the presence of OxPL that are recognized and bound by E06. In agreement with the increased titers of E06 NAbs (Figure 7) and increased OxPL antigens (Figure 4H), we observed positive E06 staining in the lungs of 6 month old chow-fed Abcg1−/− mice compared to wildtype mice (Figure 8A). We also determined whether there was increased proliferation of B cells in situ in the lung using proliferative and B cell marker co-staining on lung tissue sections from wildtype and Abcg1−/− mice that were injected with GFP+ B-1 B cells (as in Figure 3). First, consistent with the data of Figure 2 and (36) we observed increased staining of B-1 B cells in the lungs of Abcg1−/− mice compared to wildtype mice, using GFP expression levels (Figure 8B, left panel). We also observed increased proliferation of B-1 B cells, indicated by the increased staining and co-localization of the proliferative marker PCNA with GFP (Figure 8B, right panel merged image). Finally, we also observed that cells expressing PCNA also express IgM, a marker of B-1 B cells (Figure S2B). Taken together, these data demonstrate that loss of ABCG1 and tissue sterol homeostasis profoundly affects both adaptive and innate immune responses.
Discussion
Innate immunity provides an essential role in the initial defense against exogenous pathogens and in maintaining a level of homeostasis against the generation of self-antigens. NAbs are an important part of innate immunity, and it is increasingly recognized that OSE are a major target of these responses (51). NAbs are the product of natural selection and in normal, uninfected mice, most plasma IgM Abs are NAbs secreted from B-1 cells (9, 41, 52, 53). Although little is known about the maturation and expansion of B-1 cells, it is generally accepted that antigen selection during fetal development leads to a positive selection (9, 54). This selection occurs in both wild type mice and in mice raised in germ-free environments suggesting this selection is caused by endogenous self-antigens (52). Therefore, the repertoire of B-1 cells in an animal is selected to bind to evolutionarily important epitopes. We have previously demonstrated that OSE constitute a significantly large portion of these self-antigens and are a major target of innate NAbs in both mice and humans (50, 55).
ABCG1 is highly expressed in many cell types, including macrophages, lymphocytes (T and B cells), and type II pneumocytes that play essential roles in maintaining normal lung homeostasis (18, 31, 36). Consistent with important roles for ABCG1 in these cells, we and others have previously reported that the lungs of Abcg1−/− mice contain increased numbers of lipid-filled, Oil red O-positive macrophage foam cells, abnormal lamellae (phospholipid)-loaded type II pneumocytes, increased lymphocytic infiltrates, and elevated cytokines (31, 36, 56). Studies in Abcg1−/− mice have demonstrated that whole-body deletion of ABCG1 leads to cell-specific phenotypes that include increased lymphocyte proliferation (12, 13), leukocytosis (15), increased macrophage apoptosis (27), altered signaling of endothelial cells (30), and reduced insulin secretion from β-cells (21). These data suggest that ABCG1 is required for the normal function of many different cell types. Whether all these changes are a result of altered control of intracellular sterol homeostasis (20, 27) remains unknown.
In this study we demonstrate for the first time that there is a niche-specific increase in B-1 B cells in the lungs and pleural cavities of Abcg1−/− mice, and that this is accompanied by parallel increases in IgM and E06-specific IgM mRNA, consistent with increased NAb generation (Figure 2). In addition, we demonstrate that the lungs and pleural cavities of these knockout mice contain increased levels of B-1a B cells, a subset of B-1 B cells that are known to specifically secrete IgM and IgA NAbs and to be involved in innate immunity. Consistent with this latter finding, we also noted increased pulmonary levels of IgM and IgA NAbs. These changes are also cell-specific, as the levels of T cells were unaltered. Collectively these data are in agreement with the hypothesis that loss of ABCG1 leads to changes in innate immunity. Since we also show that there are elevated levels of B-2 cells and IgG antibody titers in the lungs of these knockout mice, we propose that loss of ABCG1 is associated with increases in both innate and adaptive immunity.
What are the antigens that lead to the increase in B-1/B-1a B cell number and to the increase in NAbs that include E06 IgM and T15 IgA, in the lungs and/or plasma of Abcg1−/− mice? Despite the critical importance of B-1 B cells, there is a little known regarding how these cells enter or accumulate in specific body cavities. It is well established that chemokines and chemokine receptors play an essential role in the homing of lymphocytes. Homing of B cells to the lymphoid follicles in the spleen is regulated by the chemokine CXCL13, and mice lacking CXCL13 or its receptor CXCR5 fail to generate lymphoid follicles (17, 48). CXCL13 is constitutively expressed by cells in both the peritoneal and pleural cavities, including macrophages, and mice lacking CXCL13 have a severe deficiency of B cells in the peritoneal and pleural cavities (48). Consistent with the role of CXCL13 in B-1 B cell homing, we have shown increased levels of CXCL13 in the lungs of Abcg1−/− mice, compared to wildtype mice (Figure 5).
The CXCL13-mediated accumulation of B-1 B cells has significant consequences for NAb production and local immunity, as Cxcl13−/− mice have reduced circulating PC-specific IgM and T15 idiotype-containing IgM levels, and impaired antibody response to intraperitoneal challenge with bacterial antigens (48). B-1 B cells display increased sensitivity to CXCL13 compared to B-2 B cells (48), which may further contribute to their selective movement to specific body cavities. Our data suggest that the observed increase in CXCL13 may contribute to the increased NAbs present in the lungs of Abcg1−/− mice.
Contrastingly, ectopic expression of CXCL13 was not sufficient to mediate the selective recruitment of B-1 B cells (57), indicating the presence of additional mechanisms/signals that contribute to homing. Indeed, recent data has suggested that specific oxidized derivatives of cholesterol (the most potent being 7α,25-dihydroxycholesterol) act as chemoattractants for immune cells expressing the G protein-coupled receptor EBI2 (GPR183) (47). In addition to increased levels of CXCL13, we also demonstrate increased concentrations of oxidized cholesterol derivatives (Figure 4), that may act as chemoattractants to B-1 B cells expressing GPR183/EBI2 (Figure S2A).
It has been previously shown that mycobacterium infection in the lung leads to the homing of innate immune B-1 cells to the site of infection (58) and in addition, there are other examples of tissue specific homing of B-1 cells, mostly to bacterial antigens (e.g. LPS) (59–62). However, to our knowledge the current report is the first demonstration of a site-specific expansion of B-1 cells, in response to the accumulation of an oxidized lipid antigen. Our data suggest that changes in the lipid content of the lung affects specific homing mechanisms (Figure 9), although additional studies will be required to determine if B cell signaling is also altered. Of likely direct relevance, in a manuscript published while this paper was in revision, Weber et al. show that innate response activator (IRA) B cells, a particular subset of B-1 like cells that respond to LPS stimulation via GM-CSF autocrine signaling, migrate from the pleural space into the lung parenchyma in response to infection (63). Once there, these IRA B cells secrete IgM antibodies that bind to and neutralize the bacteria, including IgM that bind to S. pneumonia (63). As noted below, E06, which binds to OxPL also binds S. pneumonia and provides optimal protection to mice from this pathogen.
The identification of the E06 IgM NAb was the first example that an endogenous oxidized lipid antigen could in fact be responsible for the positive selection of a B-1 cell clone (64). Indeed, E06 was initially identified for its ability to bind to OxLDL and only after sequencing of its variable heavy/light genes was it discovered that E06 was identical to the previously described IgA NAb termed T15 (50). T15 was well known because it provides optimal protection to mice against lethal infection with pathogens such as S. pneumonia. Both IgM E06 and IgA T15 bind to the same phosphocholine (PC) moiety, present either as the PC head group of OxPL, or as the PC covalently linked to a carbohydrate on the cell wall polysaccharide of pathogens (65). Remarkably, E06/T15 do not bind to the same PC present on unoxidized PC containing phospholipids. It was further shown that E06 bound strongly to apoptotic cells and apoptotic debris, which have increased content of such OxPL (66). In contrast, E06 did not bind to viable cells that lack OxPL. We have previously suggested that the need to provide homeostasis against apoptotic cells, which are proinflammatory and immunogenic if not promptly removed, has provided a selective pressure for the expansion of E06 and related NAbs to OSE (66, 67). In this context, it will be important to determine if the IgM antibodies secreted by IRA B-1 cells in the paper by Swirksi and colleagues noted above (63) include E06 and related IgM that bind also to OSE.
Consistent with the idea that endogenously produced oxidized lipids could drive selection and/or expansion of specific B-cell subsets, we showed that PC containing OxPL were increased in the lungs of Abcg1−/− mice (Figure 4H). Furthermore, we also showed that titers of antibodies to specific oxidized lipid antigens were increased in the lungs and plasma of Abcg1−/− mice, when compared to wild type mice (Figures 6–7, Table 2). We have previously reported that POVPC and PoxnoPC can serve as antigens for B-1 B cells resulting in increased secretion of E06 IgM (42). Indeed, we now demonstrate that IgM-E06-specific mRNA was increased 6-fold in the lungs of Abcg1−/− mice, compared to a 2-fold increase in total IgM mRNA, suggesting that there has been a specific, localized expansion of E06-secreting B-1 B cells, possibly from a selective increase in specific oxidized lipids. In turn, this B-1 cell expansion appears to be due to both homing of B-1 cells as well as localized proliferation. Consequently, our data are in agreement with a model in which loss of ABCG1 results in accumulation of both sterols and phospholipids and that some of these lipids, once oxidized, act as both homing signals for B-1 B cell movement into the lungs and pleural cavity, and that OSE drive expansion of B-1 cells and increased secretion of NAbs (Figure 9).
In addition to changes in IgM NAbs, we also observed significant increases in IgG Abs to OSE in Abcg1−/− mice. These changes in adaptive immune IgG responses are not unexpected given the significant changes in the lungs, and are reflected in the IgG increases in both the plasma and lung of Abcg1−/− mice. Most likely, these changes represent IgG generation from sites outside the lung. Collectively, these data suggest that loss of ABCG1 has broad consequences for innate and adaptive immunity. Draper et al. previously demonstrated that Abcg1−/− mice display altered IL-17 signaling and a reduced Th2 response, suggesting a role for ABCG1 in the adaptive immune response (68). Furthermore, Bensinger et al. reported that the availability of intracellular sterols is an essential check point for T cell activation and is dependent on ABCG1 (13). The rapid proliferation of T cells during activation is an important characteristic of the adaptive immune response in response to an antigen challenge, further implicating a significant role for ABCG1 in adaptive immunity. It has also been previously reported that, consistent with the data presented here, Abcg1−/− mice show no differences in T cell number, however increased T cell cycling was reported in these mice (12). It is conceivable that the increased T cell cycling could underlie an increased T cell activation, which may account for the increased B-2 B cells and IgG Abs. Nevertheless, our findings provide strong evidence for the hypothesis that changes in intracellular sterols/lipids have profound effects on the immune system (12–15, 69–71).
We have previously reported that atherosclerotic lesions of mice lacking ABCG1 display increased numbers of apoptotic macrophages in atherosclerotic lesions (27, 39). Indeed, we have shown that E06/T15 NAbs bind OxPL neo-determinants on apoptotic cells, as well as those present on OxLDL (50, 55, 66, 67, 72, 73). Both the lungs and alveolar macrophages obtained by broncho-alveolar lavage of Abcg1−/− mice show increased levels of apoptosis, when compared to wildtype mice (Figure S3). Given the elevated levels of OxPL in the lungs of Abcg1−/− mice, we propose that the combined effect of an increase in apoptotic cells and oxidized lipids drives a lung-specific expansion of B-1 B cells that could explain in part the attenuated atherosclerotic lesion development. This proposal is also consistent with studies by Kyaw et al. (74) that demonstrated that adoptive transfer of B-1 cells into mice lacking endogenous B-1 cells reduced atherosclerotic plaque burden. Importantly, transfer of sIgM−/− B-1 cells, which are unable to secrete IgM, were unable to provide such atheroprotection identifying the secreted IgM component of the B-1 cell function as being critical for protection against atherosclerosis (74). Furthermore, sIgM−/− mice on the Ldlr−/− background developed significantly more atherosclerosis (75).
In summary, to our knowledge, our results are the first demonstration of a niche-specific expansion of B cells, particularly B-1 B cells, in response to oxidized lipid antigens. We also demonstrate increases in titers of NAbs indicative of enhanced innate immunity, coupled with changes in adaptive immune Abs. We suggest that Abcg1−/− mice represent a previously unrecognized model system in which to study the atheroprotective effects of NAbs, lipid-driven immune responses, and B-1 B cell biology.
Supplementary Material
Acknowledgements
We thank Drs. Peter Edwards, Peter Tontonoz, Ken Dorshkind, Thomas Vallim and Steven Bensinger for critical and insightful discussions, and Irene H. Hernandez for technical assistance.
This work was supported in part by National Institutes of Health Grants HL107794 (to A.B.), HL30568 (to P.A.E.), HL074214 and HL111906 (to D.A.F.), HL088093 and GM69338-06 (to J.L.W.), and HL118161 (to E.J.T.). E.J.T. was partially supported by an American Heart Association (Western States Affiliate) Postdoctoral Fellowship (11POST7300060), and Beginning Grant in Aid (13BGIA17080038). A.G. was partially supported by an American Heart Association (Western States Affiliate) Postdoctoral Fellowship (12POST9580023).
Footnotes
Disclosures
The authors have no conflicts of interest or disclosures.
J.L.W. and X.Q. have patents and disclosures related to the use of oxidation-specific antibodies, which are owned by the University of California San Diego.
References
- 1.Baumgarth N. The double life of a B-1 cell: self-reactivity selects for protective effector functions. Nat. Rev. Immunol. 2011;11:34–46. doi: 10.1038/nri2901. [DOI] [PubMed] [Google Scholar]
- 2.Hardy RR. B-1 B Cell Development. J. Immunol. 2006;177:2749–2754. doi: 10.4049/jimmunol.177.5.2749. [DOI] [PubMed] [Google Scholar]
- 3.Montecino-Rodriguez E, Dorshkind K. Formation of B-1 B Cells from Neonatal B-1 Transitional Cells Exhibits NFkB Redundancy. J. Immunol. 2011;187:5712–5719. doi: 10.4049/jimmunol.1102416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Montecino-Rodriguez E, Dorshkind K. B-1 B cell development in the fetus and adult. Immunity. 2012;36:13–21. doi: 10.1016/j.immuni.2011.11.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Martin F, Kearney JF. B-cell subsets and the mature preimmune repertoire. Marginal zone and B1 B cells as part of a “natural immune memory”. Immunol. Rev. 2000;175:70–79. [PubMed] [Google Scholar]
- 6.Viau M, Zouali M. B-lymphocytes, innate immunity, and autoimmunity. Clin. Immunol. 2005;114:17–26. doi: 10.1016/j.clim.2004.08.019. [DOI] [PubMed] [Google Scholar]
- 7.Hardy RR. B-1 B cells: development, selection, natural autoantibody and leukemia. Curr. Opin. Immunol. 2006;18:547–555. doi: 10.1016/j.coi.2006.07.010. [DOI] [PubMed] [Google Scholar]
- 8.Baumgarth N, Herman OC, Jager GC, Brown LE, Herzenberg LA, Chen J. B-1 and B-2 Cell-Derived Immunoglobulin M Antibodies Are Nonredundant Components of the Protective Response to Influenza Virus Infection. J. Exp. Med. 2000;192:271–280. doi: 10.1084/jem.192.2.271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Baumgarth N, Tung JW, Herzenberg LA. Inherent specificities in natural antibodies: a key to immune defense against pathogen invasion. Springer Semin. Immunopathol. 2005;26:347–362. doi: 10.1007/s00281-004-0182-2. [DOI] [PubMed] [Google Scholar]
- 10.Bouvet JP, Dighiero G. From natural polyreactive autoantibodies to a la carte monoreactive antibodies to infectious agents: is it a small world after all? Infect. Immun. 1998;66:1–4. doi: 10.1128/iai.66.1.1-4.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Stewart J. Immunoglobulins did not arise in evolution to fight infection. Immunology today. 1992;13:396–399. doi: 10.1016/0167-5699(92)90088-O. discussion 399–400. [DOI] [PubMed] [Google Scholar]
- 12.Armstrong AJ, Gebre AK, Parks JS, Hedrick CC. ATP-binding cassette transporter G1 negatively regulates thymocyte and peripheral lymphocyte proliferation. J. Immunol. 2010;184:173–183. doi: 10.4049/jimmunol.0902372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Bensinger SJ, Bradley MN, Joseph SB, Zelcer N, Janssen EM, Hausner MA, Shih R, Parks JS, Edwards PA, Jamieson BD, Tontonoz P. LXR signaling couples sterol metabolism to proliferation in the acquired immune response. Cell. 2008;134:97–111. doi: 10.1016/j.cell.2008.04.052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Westerterp M, Gourion-Arsiquaud S, Murphy AJ, Shih A, Cremers S, Levine RL, Tall AR, Yvan-Charvet L. Regulation of Hematopoietic Stem and Progenitor Cell Mobilization by Cholesterol Efflux Pathways. Cell stem cell. 2012;11:195–206. doi: 10.1016/j.stem.2012.04.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Yvan-Charvet L, Pagler TA, Gautier EL, Avagyan S, Siry RL, Han S, Welch C, Wang N, Randolph GJ, Snoeck HW, Tall AR. ATP-binding cassette transporters and HDL suppress hematopoietic stem cell proliferation. Science. 2010;328:1689–1693. doi: 10.1126/science.1189731. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Cyster JG. Leukocyte migration: scent of the T zone. Curr. Biol. 2000;10:R30–R33. doi: 10.1016/s0960-9822(99)00253-5. [DOI] [PubMed] [Google Scholar]
- 17.Cyster JG, Ansel KM, Reif K, Ekland EH, Hyman PL, Tang HL, Luther SA, Ngo VN. Follicular stromal cells and lymphocyte homing to follicles. Immunol. Rev. 2000;176:181–193. doi: 10.1034/j.1600-065x.2000.00618.x. [DOI] [PubMed] [Google Scholar]
- 18.Kennedy MA, Barrera GC, Nakamura K, Baldan A, Tarr P, Fishbein MC, Frank J, Francone OL, Edwards PA. ABCG1 has a critical role in mediating cholesterol efflux to HDL and preventing cellular lipid accumulation. Cell Metab. 2005;1:121–131. doi: 10.1016/j.cmet.2005.01.002. [DOI] [PubMed] [Google Scholar]
- 19.Wang N, Lan D, Chen W, Matsuura F, Tall AR. ATP-binding cassette transporters G1 and G4 mediate cellular cholesterol efflux to high-density lipoproteins. Proc. Natl. Acad. Sci. U.S.A. 2004;101:9774–9779. doi: 10.1073/pnas.0403506101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Tarling EJ, Edwards PA. ATP binding cassette transporter G1 (ABCG1) is an intracellular sterol transporter. Proc. Natl. Acad. Sci. U.S.A. 2011;108:19719–19724. doi: 10.1073/pnas.1113021108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Sturek JM, Castle JD, Trace AP, Page LC, Castle AM, Evans-Molina C, Parks JS, Mirmira RG, Hedrick CC. An intracellular role for ABCG1-mediated cholesterol transport in the regulated secretory pathway of mouse pancreatic beta cells. J. Clin. Invest. 2010;120:2575–2589. doi: 10.1172/JCI41280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Tarr PT, Edwards PA. ABCG1 and ABCG4 are coexpressed in neurons and astrocytes of the CNS and regulate cholesterol homeostasis through SREBP-2. J. Lipid Res. 2008;49:169–182. doi: 10.1194/jlr.M700364-JLR200. [DOI] [PubMed] [Google Scholar]
- 23.Vaughan AM, Oram JF. ABCG1 Redistributes Cell Cholesterol to Domains Removable by High Density Lipoprotein but Not by Lipid-depleted Apolipoproteins. J Biol Chem. 2005;280:30150–30157. doi: 10.1074/jbc.M505368200. [DOI] [PubMed] [Google Scholar]
- 24.Tarling EJ, Edwards PA. Dancing with the sterols: Critical roles for ABCG1, ABCA1, miRNAs, and nuclear and cell surface receptors in controlling cellular sterol homeostasis. Biochim Biophys Acta - Molecular and Cell Biology of Lipids. 2012;1821:386–395. doi: 10.1016/j.bbalip.2011.07.011. [DOI] [PubMed] [Google Scholar]
- 25.Tarling EJ. Expanding the roles of ABCG1 and sterol transport. Curr. Opin. Lipidol. 2013;24:138–146. doi: 10.1097/MOL.0b013e32835da122. [DOI] [PubMed] [Google Scholar]
- 26.Bojanic DD, Tarr PT, Gale GD, Smith DJ, Bok D, Chen B, Nusinowitz S, Lovgren-Sandblom A, Bjorkhem I, Edwards PA. Differential expression and function of ABCG1 and ABCG4 during development and ageing. J. Lipid Res. 2009;51:169–181. doi: 10.1194/jlr.M900250-JLR200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Tarling EJ, Bojanic DD, Tangirala RK, Wang X, Lovgren-Sandblom A, Lusis AJ, Bjorkhem I, Edwards PA. Impaired development of atherosclerosis in Abcg1−/−Apoe−/− mice: identification of specific oxysterols that both accumulate in Abcg1−/−Apoe−/− tissues and induce apoptosis. Arterioscler. Thromb. Vasc. Biol. 2010;30:1174–1180. doi: 10.1161/ATVBAHA.110.205617. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Bjorkhem I. Five decades with oxysterols. Biochimie. 2012 doi: 10.1016/j.biochi.2012.02.029. [DOI] [PubMed] [Google Scholar]
- 29.Terasaka N, Wang N, Yvan-Charvet L, Tall AR. High-density lipoprotein protects macrophages from oxidized low-density lipoprotein-induced apoptosis by promoting efflux of 7-ketocholesterol via ABCG1. Proc. Natl. Acad. Sci. U.S.A. 2007;104:15093–15098. doi: 10.1073/pnas.0704602104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Terasaka N, Yu S, Yvan-Charvet L, Wang N, Mzhavia N, Langlois R, Pagler T, Li R, Welch CL, Goldberg IJ, Tall AR. ABCG1 and HDL protect against endothelial dysfunction in mice fed a high-cholesterol diet. J. Clin. Invest. 2008;118:3701–3713. doi: 10.1172/JCI35470. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Baldan A, Tarr P, Vales CS, Frank J, Shimotake TK, Hawgood S, Edwards PA. Deletion of the Transmembrane Transporter ABCG1 Results in Progressive Pulmonary Lipidosis. J. Biol. Chem. 2006;281:29401–29410. doi: 10.1074/jbc.M606597200. [DOI] [PubMed] [Google Scholar]
- 32.Bligh EG, Dyer WJ. A rapid method for total lipid extraction and purification. Can. J. Biochem. Physiol. 1959;37:911–917. doi: 10.1139/o59-099. [DOI] [PubMed] [Google Scholar]
- 33.Demarco VG, Ford DA, Henriksen EJ, Aroor AR, Johnson MS, Habibi J, Ma L, Yang M, Albert CJ, Lally JW, Ford CA, Prasannarong M, Hayden MR, Whaley-Connell AT, Sowers JR. Obesity-related alterations in cardiac lipid profile and nondipping blood pressure pattern during transition to diastolic dysfunction in male db/db mice. Endocrinology. 2013;154:159–171. doi: 10.1210/en.2012-1835. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Quehenberger O, Armando A, Dumlao D, Stephens DL, Dennis EA. Lipidomics analysis of essential fatty acids in macrophages. Prostaglandins, leukotrienes, and essential fatty acids. 2008;79:123–129. doi: 10.1016/j.plefa.2008.09.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Han X, Gross RW. Shotgun lipidomics: electrospray ionization mass spectrometric analysis and quantitation of cellular lipidomes directly from crude extracts of biological samples. Mass Spectrom. Rev. 2005;24:367–412. doi: 10.1002/mas.20023. [DOI] [PubMed] [Google Scholar]
- 36.Baldan A, Gomes AV, Ping P, Edwards PA. Loss of ABCG1 Results in Chronic Pulmonary Inflammation. J. Immunol. 2008;180:3560–3568. doi: 10.4049/jimmunol.180.5.3560. [DOI] [PubMed] [Google Scholar]
- 37.Binder CJ, Horkko S, Dewan A, Chang M-K, Kieu EP, Goodyear CS, Shaw PX, Palinski W, Witztum JL, Silverman GJ. Pneumococcal vaccination decreases atherosclerotic lesion formation: molecular mimicry between Streptococcus pneumoniae and oxidized LDL. Nat. Med. 2003;9:736–743. doi: 10.1038/nm876. [DOI] [PubMed] [Google Scholar]
- 38.Yla-Herttuala S, Palinski W, Butler S, Picard S, Steinberg D, Witztum J. Rabbit and human atherosclerotic lesions contain IgG that recognizes epitopes of oxidized LDL. Arterioscler. Thromb. Vasc. Biol. 1994;14:32–40. doi: 10.1161/01.atv.14.1.32. [DOI] [PubMed] [Google Scholar]
- 39.Baldan A, Pei L, Lee R, Tarr P, Tangirala RK, Weinstein MM, Frank J, Li AC, Tontonoz P, Edwards PA. Impaired Development of Atherosclerosis in Hyperlipidemic Ldlr−/− and ApoE−/− Mice Transplanted With Abcg1−/− Bone Marrow. Arterioscler Thromb Vasc Biol. 2006;26:2301–2307. doi: 10.1161/01.ATV.0000240051.22944.dc. [DOI] [PubMed] [Google Scholar]
- 40.Allman D, Pillai S. Peripheral B cell subsets. Curr. Opin. Immunol. 2008;20:149–157. doi: 10.1016/j.coi.2008.03.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Kearney JF. Innate-like B cells. Springer Semin. Immunopathol. 2005;26:377–383. doi: 10.1007/s00281-004-0184-0. [DOI] [PubMed] [Google Scholar]
- 42.Horkko S, Bird DA, Miller E, Itabe H, Leitinger N, Subbanagounder G, Berliner JA, Friedman P, Dennis EA, Curtiss LK, Palinski W, Witztum JL. Monoclonal autoantibodies specific for oxidized phospholipids or oxidized phospholipid-protein adducts inhibit macrophage uptake of oxidized low-density lipoproteins. J. Clin. Invest. 1999;103:117–128. doi: 10.1172/JCI4533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Goerke J. Pulmonary surfactant: functions and molecular composition. Biochim. Biophys. Acta. 1998;1408:79–89. doi: 10.1016/s0925-4439(98)00060-x. [DOI] [PubMed] [Google Scholar]
- 44.Veldhuizen R, Nag K, Orgeig S, Possmayer F. The role of lipids in pulmonary surfactant. Biochim. Biophys. Acta. 1998;1408:90–108. doi: 10.1016/s0925-4439(98)00061-1. [DOI] [PubMed] [Google Scholar]
- 45.Murakami M. Lipid Mediators in Life Science. Exp. Anim. 2011;60:7–20. doi: 10.1538/expanim.60.7. [DOI] [PubMed] [Google Scholar]
- 46.McDonald JG, Russell DW. Editorial: 25-Hydroxycholesterol: a new life in immunology. J. Leukoc. Biol. 2010;88:1071–1072. doi: 10.1189/jlb.0710418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Hannedouche S, Zhang J, Yi T, Shen W, Nguyen D, Pereira JP, Guerini D, Baumgarten BU, Roggo S, Wen B, Knochenmuss R, Noel S, Gessier F, Kelly LM, Vanek M, Laurent S, Preuss I, Miault C, Christen I, Karuna R, Li W, Koo DI, Suply T, Schmedt C, Peters EC, Falchetto R, Katopodis A, Spanka C, Roy MO, Detheux M, Chen YA, Schultz PG, Cho CY, Seuwen K, Cyster JG, Sailer AW. Oxysterols direct immune cell migration via EBI2. Nature. 2011;475:524–527. doi: 10.1038/nature10280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Ansel KM, Harris RBS, Cyster JG. CXCL13 Is Required for B1 Cell Homing, Natural Antibody Production, and Body Cavity Immunity. Immunity. 2002;16:67–76. doi: 10.1016/s1074-7613(01)00257-6. [DOI] [PubMed] [Google Scholar]
- 49.Dowler S, Kular G, Alessi DR. Protein Lipid Overlay Assay. Sci. STKE. 2002;2002:l6. doi: 10.1126/stke.2002.129.pl6. [DOI] [PubMed] [Google Scholar]
- 50.Shaw PX, Horkko S, Chang M-K, Curtiss LK, Palinski W, Silverman GJ, Witztum JL. Natural antibodies with the T15 idiotype may act in atherosclerosis, apoptotic clearance, and protective immunity. J. Clin. Invest. 2000;105:1731–1740. doi: 10.1172/JCI8472. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Pluddemann A, Neyen C, Gordon S. Macrophage scavenger receptors and host-derived ligands. Methods. 2007;43:207–217. doi: 10.1016/j.ymeth.2007.06.004. [DOI] [PubMed] [Google Scholar]
- 52.Haury M, Sundblad A, Grandien A, Barreau C, Coutinho A, Nobrega A. The repertoire of serum IgM in normal mice is largely independent of external antigenic contact. Eur. J. Immunol. 1997;27:1557–1563. doi: 10.1002/eji.1830270635. [DOI] [PubMed] [Google Scholar]
- 53.Hardy RR, Hayakawa K. Development of B cells producing natural autoantibodies to thymocytes and senescent erythrocytes. Springer Semin. Immunopathol. 2005;26:363–375. doi: 10.1007/s00281-004-0183-1. [DOI] [PubMed] [Google Scholar]
- 54.Hardy RR, Wei C-J, Hayakawa K. Selection during development of VH11+ B cells: a model for natural autoantibody-producing CD5+ B cells. Immunol. Rev. 2004;197:60–74. doi: 10.1111/j.0105-2896.2004.0100.x. [DOI] [PubMed] [Google Scholar]
- 55.Chou M-Y, Fogelstrand L, Hartvigsen K, Hansen LF, Woelkers D, Shaw PX, Choi J, Perkmann T, Backhed F, Miller YI, Horkko S, Corr M, Witztum JL, Binder CJ. Oxidation-specific epitopes are dominant targets of innate natural antibodies in mice and humans. J. Clin. Invest. 2009;119:1335–1349. doi: 10.1172/JCI36800. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Wojcik AJ, Skaflen MD, Srinivasan S, Hedrick CC. A Critical Role for ABCG1 in Macrophage Inflammation and Lung Homeostasis. J. Immunol. 2008;180:4273–4282. doi: 10.4049/jimmunol.180.6.4273. [DOI] [PubMed] [Google Scholar]
- 57.Luther SA, Lopez T, Bai W, Hanahan D, Cyster JG. BLC expression in pancreatic islets causes B cell recruitment and lymphotoxin-dependent lymphoid neogenesis. Immunity. 2000;12:471–481. doi: 10.1016/s1074-7613(00)80199-5. [DOI] [PubMed] [Google Scholar]
- 58.Russo RT, Mariano M. B-1 cell protective role in murine primary Mycobacterium bovis bacillus Calmette-Guerin infection. Immunobiol. 2010;215:1005–1014. doi: 10.1016/j.imbio.2010.01.003. [DOI] [PubMed] [Google Scholar]
- 59.Itakura A, Szczepanik M, Campos RA, Paliwal V, Majewska M, Matsuda H, Takatsu K, Askenase PW. An hour after immunization peritoneal B-1 cells are activated to migrate to lymphoid organs where within 1 day they produce IgM antibodies that initiate elicitation of contact sensitivity. J. Immunol. 2005;175:7170–7178. doi: 10.4049/jimmunol.175.11.7170. [DOI] [PubMed] [Google Scholar]
- 60.Meyer-Bahlburg A, Rawlings DJ. Differential impact of Toll-like receptor signaling on distinct B cell subpopulations. Front. Biosci. 2012;17:1499–1516. doi: 10.2741/4000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Moon H, Lee JG, Shin SH, Kim TJ. LPS-induced migration of peritoneal B-1 cells is associated with upregulation of CXCR4 and increased migratory sensitivity to CXCL12. J. Korean Med. Sci. 2012;27:27–35. doi: 10.3346/jkms.2012.27.1.27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Watanabe N, Ikuta K, Fagarasan S, Yazumi S, Chiba T, Honjo T. Migration and differentiation of autoreactive B-1 cells induced by activated gamma/delta T cells in antierythrocyte immunoglobulin transgenic mice. J. Exp. Med. 2000;192:1577–1586. doi: 10.1084/jem.192.11.1577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Weber GF, Chousterman BG, Hilgendorf I, Robbins CS, Theurl I, Gerhardt LM, Iwamoto Y, Quach TD, Ali M, Chen JW, Rothstein TL, Nahrendorf M, Weissleder R, Swirski FK. Pleural innate response activator B cells protect against pneumonia via a GM-CSF-IgM axis. J. Exp. Med. 2014;211:1243–1256. doi: 10.1084/jem.20131471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Palinski W, Horkko S, Miller E, Steinbrecher UP, Powell HC, Curtiss LK, Witztum JL. Cloning of monoclonal autoantibodies to epitopes of oxidized lipoproteins from apolipoprotein E-deficient mice. Demonstration of epitopes of oxidized low density lipoprotein in human plasma. J. Clin. Invest. 1996;98:800–814. doi: 10.1172/JCI118853. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Chou MY, Hartvigsen K, Hansen LF, Fogelstrand L, Shaw PX, Boullier A, Binder CJ, Witztum JL. Oxidation-specific epitopes are important targets of innate immunity. J. Intern. Med. 2008;263:479–488. doi: 10.1111/j.1365-2796.2008.01968.x. [DOI] [PubMed] [Google Scholar]
- 66.Chang MK, Binder CJ, Miller YI, Subbanagounder G, Silverman GJ, Berliner JA, Witztum JL. Apoptotic cells with oxidation-specific epitopes are immunogenic and proinflammatory. J. Exp. Med. 2004;200:1359–1370. doi: 10.1084/jem.20031763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Miller YI, Choi S-H, Wiesner P, Fang L, Harkewicz R, Hartvigsen K, Boullier A, Gonen A, Diehl CJ, Que X, Montano E, Shaw PX, Tsimikas S, Binder CJ, Witztum JL. Oxidation-Specific Epitopes Are Danger-Associated Molecular Patterns Recognized by Pattern Recognition Receptors of Innate Immunity. Circ. Res. 2011;108:235–248. doi: 10.1161/CIRCRESAHA.110.223875. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Draper DW, Gowdy KM, Madenspacher JH, Wilson RH, Whitehead GS, Nakano H, Pandiri AR, Foley JF, Remaley AT, Cook DN, Fessler MB. ATP Binding Cassette Transporter G1 Deletion Induces IL-17-Dependent Dysregulation of Pulmonary Adaptive Immunity. J. Immunol. 2012 doi: 10.4049/jimmunol.1101605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Sag D, Wingender G, Nowyhed H, Wu R, Gebre AK, Parks JS, Kronenberg M, Hedrick CC. ATP-binding cassette transporter G1 intrinsically regulates invariant NKT cell development. J. Immunol. 2012;189:5129–5138. doi: 10.4049/jimmunol.1201570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Kidani Y, Elsaesser H, Hock MB, Vergnes L, Williams KJ, Argus JP, Marbois BN, Komisopoulou E, Wilson EB, Osborne TF, Graeber TG, Reue K, Brooks DG, Bensinger SJ. Sterol regulatory element-binding proteins are essential for the metabolic programming of effector T cells and adaptive immunity. Nat. Immunol. 2013;14:489–499. doi: 10.1038/ni.2570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Williams KJ, Argus JP, Zhu Y, Wilks MQ, Marbois BN, York AG, Kidani Y, Pourzia AL, Akhavan D, Lisiero DN, Komisopoulou E, Henkin AH, Soto H, Chamberlain BT, Vergnes L, Jung ME, Torres JZ, Liau LM, Christofk HR, Prins RM, Mischel PS, Reue K, Graeber TG, Bensinger SJ. An essential requirement for the SCAP/SREBP signaling axis to protect cancer cells from lipotoxicity. Cancer Res. 2013;73:2850–2862. doi: 10.1158/0008-5472.CAN-13-0382-T. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Chang MK, Bergmark C, Laurila A, Horkko S, Han KH, Friedman P, Dennis EA, Witztum JL. Monoclonal antibodies against oxidized low-density lipoprotein bind to apoptotic cells and inhibit their phagocytosis by elicited macrophages: evidence that oxidation-specific epitopes mediate macrophage recognition. Proc. Natl. Acad. Sci. U.S.A. 1999;96:6353–6358. doi: 10.1073/pnas.96.11.6353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Tuominen A, Miller YI, Hansen LF, Kesaniemi YA, Witztum JL, Horkko S. A Natural Antibody to Oxidized Cardiolipin Binds to Oxidized Low-Density Lipoprotein, Apoptotic Cells, and Atherosclerotic Lesions. Arterioscler. Thromb. Vasc. Biol. 2006;26:2096–2102. doi: 10.1161/01.ATV.0000233333.07991.4a. [DOI] [PubMed] [Google Scholar]
- 74.Kyaw T, Tay C, Krishnamurthi S, Kanellakis P, Agrotis A, Tipping P, Bobik A, Toh B-H. B1a B Lymphocytes Are Atheroprotective by Secreting Natural IgM That Increases IgM Deposits and Reduces Necrotic Cores in Atherosclerotic Lesions / Novelty and Significance. Circ. Res. 2011;109:830–840. doi: 10.1161/CIRCRESAHA.111.248542. [DOI] [PubMed] [Google Scholar]
- 75.Lewis MJ, Malik TH, Ehrenstein MR, Boyle JJ, Botto M, Haskard DO. Immunoglobulin M is required for protection against atherosclerosis in low-density lipoprotein receptor-deficient mice. Circulation. 2009;120:417–426. doi: 10.1161/CIRCULATIONAHA.109.868158. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.