Abstract
Shotgun proteomics is a powerful analytic method to characterize complex protein mixtures in combination with multi-dimensional liquid chromatography-tandem mass spectrometry (LC-MS/MS). We have used this platform for proteomic characterization of apoptotic bodies in efforts to define the complex protein mixtures found in primary cultures of human intrahepatic biliary epithelial cells (HiBEC), human renal proximal tubular epithelial cells, human bronchial epithelial cells, isolated intrahepatic biliary epithelial cells from explanted primary biliary cirrhosis (PBC) and control liver, using a total of 24 individual samples. Further, as additional controls and for purposes of comparison, proteomic signatures were also obtained from intact cells and apoptotic bodies. The data obtained from LC-MS/MS, combined with database searches and protein assembly algorithms, allowed us to address significant differences in protein spectral counts and identify unique pathways that may be a component to the induction of the signature inflammatory cytokine response against BECs, including the Notch signaling pathway, IL8, IL6, CXCR2 and integrin signaling. Indeed there are 11 proteins that localize specifically to apoptotic bodies of HiBEC and 8 proteins that were specifically absent in HiBEC apoptotic bodies. In conclusion, proteomic analysis of BECs from PBC liver compared to normal liver are significantly different, suggesting that an immunological attack affects the repertoire of proteins expressed and that such cells should be thought of as living in an environment undergoing continuous selection secondary to an innate and adaptive immune response, reflecting an almost “Darwinian” bias.
Keywords: apoptosis, shotgun proteomics, apoptotic bodies, biliary epithelial cells
We have previously postulated that the selective destruction of biliary epithelial cells (BECs) in PBC is related to the relationships between the apoptotic bodies of BEC, the innate immune system and mitochondrial autoantibodies (1–13). This model includes an MHC-independent mechanism as PBC can recur following liver transplantation, i.e. irrespective of HLA mismatch (14, 15). The bleb contents of biliary cells may contain components that stimulate pro-inflammatory responses or alternatively lack components that normally suppress such responses. Over the past decade, shotgun proteomics has emerged as a powerful analytical method to characterize complex protein mixtures (16–18). By combining multidimensional liquid chromatography-tandem mass spectrometry (LC-MS/MS) with database searches and protein assembly algorithms, shotgun proteomics platforms surpass other MS-based proteomics systems in the number and the diversity of proteins identified, and in the dynamic range of detection. Moreover, shotgun proteomic data sets from different tissue phenotypes are sufficiently rich in quantitative information that statistically significant differences in proteins spectral counts can identify differences in abundance of proteins in the samples (19, 20).
We report herein the proteomic characterization of the content of apoptotic bodies from human BECs. We identify 11 proteins localized specifically to apoptotic bodies of BECs but not similar apoptotic bodies from other epithelial tissues, and 8 proteins that were specifically absent in HiBEC apoptotic bodies. We also identify 6 proteins present in apoptotic blebs from BECs isolated from PBC patients when compared to those from BECs from healthy donors. These signature proteins and pathways may shed light on potential molecules for therapeutic intervention.
METHODS
Cell types and isolation of biliary epithelial cells
Primary cultures of normal HiBEC, Human Renal Proximal Tubular Epithelial cells (HRPTEpiC) and human bronchial epithelial cells (BrEPC) cells (ScienCell, San Diego, California) were primary cultures isolated from normal human tissue and cryopreserved immediately after purification. HiBEC were cultured in epithelial cell medium (ScienCell) supplemented with 2% fetal bovine serum (FBS), epithelial cell growth supplement (ScienCell) and 1% penicillin in flasks coated with poly-L-lysine (Sigma-Aldrich, St. Louis, MO). HiBEC were characterized using a previously described immunofluorescence microscopic method with antibodies to cytokeratin 7, cytokeratin 19 and vimentin, which labeled >90% of the cells (21, 22). The other epithelial cells were cultured under the same conditions without FBS. All experiments were performed using cells between passage 2 and 5 (8, 9).
Human BECs were isolated from PBC end stage liver tissue removed at transplantation and from non-diseased liver tissue that were surplus to transplant requirements (23). All tissue was obtained from fully informed patients with written consent. Ethical approval was granted by the South Birmingham Research Ethics Committee. Liver tissue underwent mechanical and enzymatic digestion with Collagenase type 1A (Sigma Aldrich, Dorset, UK) and density gradient centrifugation on 33%/77% Percoll. BECs were further purified by immunomagnetic isolation using antibodies against the cholangiocyte-specific receptor HEA 125 (Progen, Heidelberg, Germany). The cells were cultured in Dulbecco’s Modified Eagle medium, Hams F12 plus 10% heat-inactivated human serum, glutamine (2 mM), hepatocyte growth factor (10 ng/ml, Peprotech, London, UK), epidermal growth factor (10 ng/ml, Peprotech), hydrocortisone (2 µg/ml), cholera toxin (10 ng/ml, Sigma Aldrich), tri-iodo-thyronine (2 nM, Sigma Aldrich) and insulin (0.124 U/ml) with penicillin (100 IU/ml) and streptomycin (100 µg/ml). Cells were cultured to confluence in tissue culture flasks coated with rat tail collagen.
Apoptosis induction and isolation of apoptotic bodies
Apoptosis was induced as described (9); briefly HIBEC, HRPTEpiC and BrEPC were incubated in medium containing 1mM sodium glycochenodeoxycholate at 37°C for 4h. To establish optimal conditions for the induction of apoptosis, we previously incubated all cells types at 37°C for 1, 2, 3, and 4 hours using serial concentrations (100 µM, 500 µM, 1 mM, and 2mM) of GCDC added to normal culture medium, and in absence of serum and growth factors (24). The degree of apoptosis was confirmed for each experiment.
Apoptotic blebs were isolated as previously described (9) from normal primary cultures of HIBEC, HRPTEpiC and BrEPC, as well as from BECs isolated from PBC patients and healthy controls. Briefly, the cell culture supernatant fluid was collected after apoptosis induction. Two additional centrifugation steps (500g for 5 minutes) were performed to remove remaining cells. The supernatant fluid was thence passed through a 1.2 µm non-pyrogenic, hydrophilic syringe filter. After centrifugation at 100,000g for 30 minutes, the pellet containing apoptotic bodies was harvested and lysed in RIPA buffer (Thermo Scientific Pierce, Rockford, IL) supplemented with Complete Protease Inhibitor Cocktail (Roche, Indianapolis, IN). Protein concentration was determined by the BCA assay (Thermo Scientific Pierce). A total of 24 samples of apoptotic bodies were studied, including 4 independent preparations of HiBEC cells, 6 independent preparations of HRPTEpiC, 6 human BrERC, 3 independent preparations of BECs from PBC patients, and 5 from healthy controls. Six different samples of apoptotic blebs of each cell type were isolated. Finally to compare the expression level of proteins found in apoptotic bodies with those in intact cells, we ran an extensive proteome characterization of HiBEC, HRPTEpiC, and BrEPC.
Comparative Shotgun Proteomics
Protein content of the samples was determined by the BCA (bicinconic acid) assay and using a Nanodrop ND-1000 ultraviolet-visible Spectrophotometer (NanoDrop Technologies, Wilmington, DE). Purified apoptotic bodies containing normalized levels of protein were lysed in urea buffer (7M urea, 1M thiourea, 4% CHAPS, 4 mg/mL DTT, pH 8.0). Protein was precipitated using 25% TCA (w/v), washed with ice cold acetone, and re-suspended in 8M urea 100 mM Tris, pH 8.0. Proteins were reduced with TCEP, alkylated with iodoacetemide, diluted to 2M urea, and then digested with trypsin. The resulting peptide mixtures were split into duplicates and analyzed via MudPIT (25). Briefly, peptides were loaded onto a biphasic pre-column fitted using an Upchurch M-520 filter union. This 150 um fused silica microcapillary column was packed with 4 cm of 5-µm strong cation-exchange resin followed by 4 cm of C18 reverse-phase resin. Once loaded, it was then placed in-line with a 100 µm X 20 cm, C18 packed emitter tip column coupled to an LTQ ion trap mass spectrometer nanospray source. Separations were accomplished using 25–1000 mM pulses of ammonium acetate followed by a 115 min reversed phase gradient of 0–40% acetonitrile 0.1% formic acid. Tandem mass spectra were collected in a data dependent manner using dynamic exclusion. Spectra were extracted using ScanSifter and searched against the mouse Uniprot database that also contained reversed versions of the proteins using Sequest. Peptide to protein matches and spectral counts were assembled using IDPicker (9, 26) using a false discovery rate target of 5%. All shotgun proteomics experiments were performed at the Proteomics Laboratory at Vanderbilt University.
Statistical Analysis and Pathway definition
The data comparing the hepatic blebs to each of the control groups were individually plugged into Quasitel, a graphical interface software package that reads standard output from protein assemblies created by IDPicker, and analyzed with a quasi-likelihood Generalized Linear Modeling (GLM) system (19). All reported proteins were identified with a minimum of 2 distinct peptides. The fold change of each protein was calculated by comparing the log2 of total spectra per protein in blebs from HiBEC compared to the log2 ratio of spectra in HRPTEpiC or BrEPC. The results were then sorted by a metric (quasi-P) that takes into account both the difference between the two groups and how consistent the values are within that group. Only p<0.05 were considered significant. To handle the complications presented by simultaneously testing thousands of proteins, we applied the False Discovery Rate (FDR) method (27). The top 250 protein groups by spectral count were selected and unsupervised hierarchal clustering was performed using MeV (Multiple Experiment Viewer – http://www.tm4.org/mev/).
Pathway analysis was performed using The Pathway Interaction Database National Cancer Institute (http://pid.nci.nih.gov) (28) as well as the STRING (Search Tool for the Retrieval of Interacting Genes) software (v.9.0) (http://string-db.org/) (29). STRING is a large database of known and predicted protein interactions. All the edges were supported by at least a reference from the literature or from canonical information stored in the STRING dataset. A confidence score was fixed at 0.4 (medium level). Cluster networks were created using the MCL algorithm which is included in the STRING website and a value of 2 was selected for all the analyses. To simplify large datasets, all proteins that met the filter criteria by IDPicker groups were filtered to include proteins containing 25–300 total spectral counts. Only proteins exclusively detected in HiBEC blebs when compared to both HRPTEpiC and BrEPC were retained.
RESULTS
Characterization of the proteome of apoptotic bodies from primary cultures
Firstly, we note that samples were isolated and run independently. We identified a total of 40,843 distinct peptides and 6,160 protein groups within apoptotic bodies from HiBEC, BrEPC, and HRPTEpiC (Table 1A) and similar numbers in those from BECs from PBC patients and controls (Table 1B). To eliminate proteins that had too few spectral counts to be statistically different, we reduced the data set to protein groups that were identified by at least 5 peptides spectra. An unsupervised clustering of the top 250 proteins by spectral count was able to appropriately separate the samples (Figure 1), suggesting consistent results and distinguishing characteristics between groups.
Table 1.
Number of unique peptides, spectra, and proteins identified for a given run of apoptotic bodies isolated from HiBEC, BrEPC, and HRPTEpiC (A), and of apoptotic bodies from BECs isolated from PBC patients and healthy controls (B).
| A. | Distinct Peptides |
Distinct Matches |
Filtered Spectra |
Protein Groups |
|---|---|---|---|---|
| BrEPC | 7572 | 9120 | 14699 | 2881 |
| 7979 | 9148 | 14456 | 3003 | |
| 8842 | 10131 | 14444 | 3247 | |
| 10069 | 11531 | 16635 | 3361 | |
| 10528 | 12275 | 19153 | 3310 | |
| 10978 | 12761 | 18375 | 3190 | |
| HiBEC | 7005 | 8082 | 14270 | 2733 |
| 6074 | 7111 | 13849 | 2601 | |
| 5766 | 6833 | 10922 | 2544 | |
| 4305 | 5114 | 10493 | 2085 | |
| HRPTEpiC | 9875 | 11540 | 19218 | 3392 |
| 7747 | 9208 | 15159 | 2919 | |
| 7245 | 8713 | 14065 | 2919 | |
| 8858 | 10563 | 16675 | 3191 | |
| 9711 | 11038 | 14455 | 3105 | |
| 7265 | 8688 | 14245 | 2730 | |
| B. |
Distinct Peptides |
Distinct Matches |
Filtered Spectra |
Protein Groups |
| Healthy controls | 3743 | 4638 | 17479 | 1128 |
| 3920 | 4652 | 15153 | 1177 | |
| 8090 | 9929 | 21127 | 1843 | |
| 6852 | 8182 | 20275 | 1709 | |
| 5504 | 6790 | 17151 | 1466 | |
| PBC patients | 4923 | 6586 | 18949 | 1264 |
| 4848 | 6271 | 16567 | 1276 | |
| 6702 | 8557 | 20781 | 1655 | |
Figure 1.
Unsupervised clustering. The top 250 protein groups by spectral count were selected and unsupervised hierarchal clustering using MeV (Multiple Experiment Viewer – http://www.tm4.org/mev/) was performed. The figure shows clustering of apoptotic bodies isolated from HiBEC, BrEPC, and HRPTEpiC (A), and of apoptotic bodies from BECs isolated from PBC patients and healthy controls (B).
Both data sets were tested for possible differences. First, we compared the proteome of apoptotic bodies from commercial primary cultures (i.e. HiBEC, BrEPC, and HRPTEpiC); which yielded a total of 589 protein database entries when apoptotic bodies from BrEPC were compared to those from HiBEC and a total of 663 protein database entries when apoptotic bodies from HRPTEpiC were compared to those from HiBEC, with a quasi p-value of less than 0.05 and at least a 4-fold difference in spectral counts (log2(rate1/rate2), or rate ratio, higher than 2 or lower than −2). This list includes a number of proteins with artificially low p-values either because no spectra were identified in one of the groups (reported with “cv” value of “NA”).
We also looked for proteins that were specifically identified in apoptotic bodies from HiBEC when compared to apoptotic bodies from both bronchial and renal epithelial cells (Table 2). Only 11 proteins were found to be specific for apoptotic bodies of HiBEC with a quasi p-value of less than 0.05 and at least a 4-fold difference in spectral counts. Table 3A summarizes the function of the identified proteins and the related genes. Further, we also investigated the presence of proteins unique to apoptotic bodies from BrEPC and HRPTEpiC, and absent from HiBEC. We identified eight such proteins and they are listed in Table 3B.
Table 2.
List of proteins specifically located in apoptotic bodies from HiBEC compared to epithelial controls.
| BrEPC vs HiBEC | HRPTEpiC vs HiBEC | |||||
|---|---|---|---|---|---|---|
| Proteins | 2log (λ1/λ2) | quasi p- value fdr |
Total Counts |
2log (λ1/λ2) | quasi p- value fdr |
Total Counts |
| A6NN80 | −3.05 | 0.000961 | 1061 | −2.21 | 0.002345 | 1189 |
| ANXA6 | −3.09 | 0.000880 | 1064 | −2.18 | 0.002086 | 1204 |
| B4DN38 | −2.39 | 0.018616 | 81 | −2.39 | 0.016801 | 81 |
| GPC6 | −4.16 | 0.007293 | 39 | −1.39 | 0.017383 | 66 |
| HSPB6 | −6.62 | 0.004719 | 67 | −2.20 | 0.001517 | 57 |
| LRP1 | −2.42 | 0.008753 | 55 | −6.62 | 0.003810 | 67 |
| PAPS2 | −6.35 | 0.001450 | 111 | −2.27 | 0.002121 | 97 |
| Q6ZR44 | −4.67 | 0.005526 | 72 | −2.75 | 0.002872 | 55 |
| RAB11A | −2.31 | 0.005947 | 160 | −1.16 | 0.002963 | 95 |
| SERPH | −2.41 | 0.000674 | 396 | −1.23 | 0.013308 | 95 |
| VGFR3 | −5.76 | 0.015027 | 521 | −6.98 | 0.011321 | 513 |
Top Ranked Proteins with differential spectral counts that are uniquely present in apoptotic bodies from HiBEC when compared to apoptotic bodies from both bronchial (BrEPC) and renal (HRPTEpiC) cells.
Table 3.
Encoded protein, gene and description of the specific HiBEC apoptotic bodies proteome (A) and of proteins located in apoptotic bodies from both bronchial (BrEPC) and renal (HRPTEpiC) cells but absent in HiBEC apoptotic bodies (B).
| A. | ||
|---|---|---|
| Protein | Gene | Description |
| A6NN80 | ANXA6 | Putative uncharacterized protein |
| ANXA6 | ANXA6 | Annexin A6, Lipocortin VI, p68, p70, Protein III, Chromobindin-20, 67 kDa calelectrin, Calphobindin-II |
| B4DN38 | TSC22D2 | highly similar to Glucosamine--fructose-6-phosphateaminotransferase (isomerizing) |
| GPC6 | GPC6 | Secreted glypican-6 |
| HSPB6 | HSPB6 | Heat shock protein beta-6, Heat shock 20 kDa-like protein p20 |
| LRP1 | LRP1 | Low-density lipoprotein receptor-related protein 1, Alpha-2-macroglobulin receptor, Apolipoprotein E receptor, Low-density lipoprotein receptor-related protein 1 intracellular domain |
| PAPS2 | PAPSS2 | 3’-phosphoadenosine 5’-phosphosulfate synthase 2, isoform CRAb, FLJ92929, highly similar to Homo sapiens 3’-phosphoadenosine 5’-phosphosulfate synthase 2 |
| Q6ZR44 | C20orf151 | highly similar to Thioredoxin reductase |
| RAB11A | RAB11A | RAB 11A, member oncogene family; rab-11; ras-related protein Rab-11A |
| SERPH | SERPINH1 | highly similar to Collagen-binding protein 2, Serpin peptidase inhibitor, clade H (Heat shock protein 47), member 1, (Collagen binding protein 1), isoform CRAa |
| VGFR3 | SERPINB4 | Tyrosine-protein kinase receptor |
| B. | ||
| Protein | Gene | Description |
| ANXA3 | ANXA3 | Annexin A3 |
| H2AW | H2AFY2 | Core histone H2A Inositol 1,4,5-trisphosphate receptor type 3, Type 3 inositol |
| ITPR3 | ITPR3 | 1,4,5-trisphosphate receptor, Type 3 InsP3 receptor, IP3 receptor isoform 3, InsP3R3 |
| K1C19 | KRT19 | Keratin, type I cytoskeletal 19, Cytokeratin-19, Keratin-19 |
| K2C7 | KRT7 | Keratin, type II cytoskeletal 7, Cytokeratin-7 Laminin subunit gamma-2, Laminin 5 gamma 2 subunit, Kalinin/nicein/epiligrin 100 kDa subunit, Laminin B2t chain, |
| LAMC2 | LAMC2 | Cell-scattering factor 140 kDa subunit, Large adhesive scatter factor 140 kDa subunit |
| PYGB | PYGB | Glycogen phosphorylase, brain form |
| Q9HA11 | ACAD9 | Histone H2A |
Proteome of intact cells versus apoptotic bodies
The presence of specific proteins within the apoptotic bodies was compared to the global proteome of naïve non apoptotic cells from the three epithelial cell types included in this study (HiBEC, HRPTEpiC, and BrEPC). We identified a total of 3,152 protein groups within HiBECs, HRPTEpiCs, and BrEPCs. Of the 11 proteins uniquely found in the apoptotic bodies of HiBEC cells, 4 of the 11 (ANXA6, LRP1, PAPS2, and SERPH) were found to be present in all three intact cell lines. One protein, HSPB6, was found only in intact HiBEC, but not intact HRPTEpiC or BrEPC. Interestingly, the other 6 proteins that were uniquely found in the blebs of HiBEC (A6NN80, B4DN38, GPC6, Q6ZR44, RAB11A AND VGFR3), were not found in intact cells. Finally, of the 3,152 protein groups, there were only 3 proteins found in intact HiBEC cells, but not in HiBEC apoptotic bodies (ANXA3, PYGB, and ITPR3). Hence, the presence of proteins specifically located in the blebs of isolated HiBEC cells appear secondary to apoptosis.
Proteome of apoptotic bodies from BECs from patients and controls
Comparison of the proteome of BECs from PBC patients and healthy controls yielded a total of 165 protein database entries, with a quasi p-value of less than 0.05 and at least a 4-fold difference in spectral counts (log2(rate1/rate2), or rate ratio, higher than 2 or lower than −2). We looked for proteins that were specifically located in apoptotic bodies from PBC patients compared to apoptotic bodies from healthy controls (Table 4). Six proteins were found to be specific for apoptotic bodies of PBC patients with a quasi p-value of less than 0.05 and at least a 4-fold difference in spectral counts. Further, we also investigated the presence of proteins unique to apoptotic bodies from healthy controls that are absent in those from PBC patients. We identified only 2 such proteins and they are listed in Table 4. Table 5 summarizes the function of the proteins and related genes.
Table 4.
List of proteins specifically located in apoptotic bodies isolated from BECs from PBC patients and controls.
| A. | |||
|---|---|---|---|
| Proteins | 2log (λ1/λ2) | quasi p-value fdr | Total Counts |
| CPN1 | −2.28 | 0.00017 | 52 |
| ITIH2 | −2.10 | 0.00134 | 1493 |
| C9 | −2.44 | 0.00180 | 288 |
| FGG | −2.96 | 0.00180 | 192 |
| FGA | −2.69 | 0.01155 | 155 |
| SERPINF2 | −2.08 | 0.01752 | 94 |
| B. | |||
| Proteins | 2log (λ1/λ2) | quasi p-value fdr | Total Counts |
| APOC2 | 32.44 | 0.007316 | 16 |
| DRG1 | 32.03 | 0.015156 | 12 |
Top ranked proteins with differential spectral counts that are uniquely present in apoptotic bodies from BECs from PBC patients when compared to blebs from BECs from healthy donors (A) and uniquely present in apoptotic blebs from BECs from healthy donors but absent in those from PBC patients (B).
Table 5.
Gene, encoded protein and description of proteins located in apoptotic bodies from BECs from PBC patients and healthy controls.
| Protein | Gene | Description |
|---|---|---|
| CPN1 | CPN1 | Carboxypeptidase N catalytic chain |
| ITIH2 | ITIH2 | Inter-alpha (Globulin) inhibitor H2 |
| C9 | C9 | Complement component C9 |
| FGG | FGG | Fibrinogen gamma chain |
| FGA | FGA | Fibrinogen alpha chain |
| SERPINF2 | SERPINF2 | Alpha-2-antiplasmin |
| APOC2 | APOC2 | Apolipoprotein C-II |
| DRG1 | DRG1 | Developmentally-regulated GTP-binding protein 1 |
Pathway analysis
Using the STRING database a proteome-scale interaction network was created. This database includes interactions from the published literature describing interactions as well as genome analysis based on domain fusion, phylogenetic profiling and gene neighborhood concepts. Accordingly, a confidence score for every protein-protein association was assigned to the network. To minimize false positives as well as false negatives, all interactions tagged as “low-confidence” (<0.4) in STRING database were eliminated from this study. The analysis resulted in the Protein interaction network for LRP1 (also known as A2MR), RAB11A and ANXA6 using the STRING software as illustrated in Figure 2. Stronger associations are represented by thicker lines. We also examined the pathways found in apoptotic bodies using The Pathway Interaction Database, NIH. This proteomic pathway analysis identifies inflammation pathways (Table 6).
Figure 2.
LRP1, ANXA6, RAB11A, FGG, and FGA network display. Nodes are either colored (if they are directly linked to the input protein) or white (nodes of a higher iteration/depth). Edges, i.e. predicted functional links, consist of up to eight active prediction methods: neighborhood, gene fusion, co-occurrence, co-expression, experiments, databases, textmining.
Table 6.
Pathway analysis of the BEC apoptotic bodies proteome.
| Protein | Pathway |
|---|---|
| ANXA6 | - EGFR/Ras signaling pathway |
| LRP1 | - amb2 Integrin signaling |
| - PDGFR-beta signaling pathway | |
| - Urokinase-type plasminogen activator (uPA) and uPAR-mediated signaling | |
| - ERK pathway | |
| - nuclear factor-kappa B | |
| RAB11A | - Notch signaling pathway |
| - Thromboxane A2 receptor signaling | |
| - Arf6 downstream pathway | |
| - IL8- and CXCR2-mediated signaling events | |
| FGG | - IL6-mediated signaling events |
| - Beta1, beta2, and beta3 integrin cell surface interactions | |
| - Urokinase-type plasminogen activator (uPA) and uPAR-mediated signaling | |
| FGA | - Beta1, beta2, and beta3 integrin cell surface interactions |
| - Urokinase-type plasminogen activator (uPA) and uPAR-mediated signaling | |
| SERPINF2 | - fibrinolysis pathway |
| APOC2 | - metabolism of lipids and lipoproteins |
Candidate proteins were uploaded into The Pathway Interaction Database of the National Cancer Institute after manually filtering for protein groups as described.
DISCUSSION
We report the use of shotgun proteomics to identify proteins that might be part of the induction of a signature inflammatory cytokine response in PBC. The cells most closely similar to the true target of disease are normal HiBECs from unaffected individuals. These are the most abundant available source of cells that would mimic biliary cells at the time of induction of disease. Importantly it is apoptotic bodies derived from these normal HiBECs that stimulate a pro-inflammatory response (8, 12), but there may be aspects of their proteome that differ from those found in apoptotic bodies isolated from HiBECs of patients at the commencement of disease. Such patients are difficult to identify and there are ethical issues with obtaining sufficient tissue for experimentation. However as an alternative we also examined apoptotic bodies present in the residual biliary epithelial cells that have survived immune destruction in PBC patients. These patients are many years after the disease induction event and their liver has been under sustained immunological attack for decades. Thus these HiBECs may now be somewhat different from such cells at the time of disease initiation. Further, unfortunately the extent of biliary destruction in PBC patients is quite severe and the low number of such surviving cells makes it technically impossible to derive sufficient apoptotic blebs to determine if such late stage cells are capable of inducing a pro-inflammatory response, but there are enough cells to provide a sample for proteomic analysis. The fact that proteomic analysis of apoptotic bodies from these cells suggest that they differ from apoptotic bodies of normal human HiBECs is noteworthy.
Immunological attack appears to have affected the repertoire of proteins expressed. Indeed, these cells may be thought of as having lived in an environment that would continuously select against production of molecules that would be stimulatory to the immune response. This almost “Darwinian” selection may explain why such cells do not produce the same set of unique proteins present in the apoptotic bodies of normal HiBECs. The unique proteins they do produce may be candidates for proteins that down regulate or ameliorate immune attack in such a way as to allow survival of these cells. We chose bile salts for apoptosis induction to mirror hepatic physiology. Bile salts accumulating in the liver during cholestatis trigger liver injury and subsequent fibrosis. A constituent of the hydrophobic bile salt, GCDC, induces apoptosis of hepatocytes at a concentration of 50 µM (30). GCDC has also an apoptotic effect in cholangiocytes (24). Future studies should take advantage of a variety of other epithelial cells and including comparisons of large versus small bile ducts from explants of PBC and non-PBC liver diseases.
Through a rigorous and sensitive comparison of the proteome of apoptotic bodies from HiBEC and non-stimulatory epithelial apoptotic bodies, we detected 11 proteins specifically located within apoptotic bodies from HiBEC. These proteins are largely involved in immune (and in the right context, autoimmune) reactions including NF-kB activation, ERK pathway, Notch signaling pathway, and IL8- and CXCR2-mediated signaling events (31–34). Of note, four proteins out of these eleven proteins were detected in all three intact cells (HiBEC, HRPTEpiC and BrEPC) and one protein was detected in only intact HiBEC. This finding reinforces our thesis that apoptotic bodies play a pathogenic role in biliary inflammation (35). Interestingly we were not able with this technique to detect PDC-E2 within HiBEC apoptotic bodies. It may be that the signature peptides that lead to the identification of PDC-E2 are not soluble, or are degraded/eliminated by the protocol utilized since there is limited knowledge on the susceptibility of peptides to such procedures.
In this context, among the proteins located within HiBEC apoptotic bodies, GPC6 looks particularly interesting as it is a secreted protein that has marked functional effects in the CNS (38). Six members of the glypicans family have been identified in mammals (GPC1-GPC6). Glypicans act as regulators of various cytokines, including Wnts, Hedgehogs, and bone morphogenetic proteins (39–41). No functional information on GPC6 is yet available. Data available from public databases (http://www.proteinatlas.org/index.php) note moderate immunostaining in hepatocytes, but prominent staining in the epithelial lining of the gall bladder (ie. cholangiocytes). Hedgehog signaling pathways are well known to be involved in the fibroproliferative response upon cholestatic biliary epithelial cell injury in PBC (43).
It is interesting to note and discuss LRP1 and its potential role in the pathogenesis of PBC. Firstly, we should emphasize that PBC reoccurs following liver transplants and therefore there does not appear to be a PBC-specific biliary phenotype. Although this intense proteomic approach provides significant new data on biliary biology, we cannot specifically address etiology, including the potential role of either environmental agents or infections. However, lipoprotein receptors from the low density lipoprotein receptor (LRP) family are membrane proteins with multifunctional scavenger and signaling receptor functions (44). The largest member of this family, the LDL receptor-related protein 1 (LRP1), also known as cluster of differentiation 91 (CD91), serves as a multifunctional endocytic receptor with a broad range of ligands. CD91 is pivotal in the regulation of heat shock proteins (HSPS) mediated immune co-stimulation (45). CD91 and members of the scavenger receptor family efficiently direct Hsp70:peptide complexes into the MHC class II presentation pathway and thus enhance antigen-specific CD4+ T cell responses (46).
RAB11A is part of the small GTPase superfamily, Rab family. RAB11A is involved in both constitutive and regulated secretory pathways, and takes part in protein transport. RAB11A is a well-known marker for protein trafficking, sorting, and recycling in the endosomal pathway. RAB11A is also an important regulator of membrane delivery during cytokinesis. Importantly, the ability of toll-like receptors (TLRs) to activate innate immunity depends on their transport to pathogen-containing organelles.
Although a specific factor in apoptotic bodies of HiBECs may be the factor responsible for the innate immune response in PBC patients (8), it is possible that it is the loss or absence of down modulating proteins found in the other tissues that may be part of the pathophysiology. Therefore, we also investigated the presence of proteins unique to apoptotic bodies from BrEPC and HRPTEpiC, but absent in those from HiBEC. Among these molecules, IPR3, which regulates the release of Ca(2+) from intracellular stores, might play an important role in the immune response and uptake of apoptotic bodies. Furthermore, it is also involved in both naïve CD4 T cells cytokine release (51) and FcγR-mediated phagocytosis (52). Other findings of interest in this set of proteins are cytokeratines 19 and 7, as well as laminin subunit gamma-2, which are known to be located in epithelial cells (including BrEPC and HRPTEpiC) in high concentration. An accumulation of the linker-histone (histone 1) as well as the core-histones (histone 2A, histone 2B, histone 3, histone 4) have been demonstrated to be widely located in apoptotic bodies (53), so their absence from HiBECs is unexpected. Finally, we note that Annexin A3 is enriched in the apoptotic bodies of renal tubules and bronchial epithelium, but it is annexin A6 that is enriched in biliary epithelial cells.
Our data reflect the results of a detailed differential analysis of the proteome of apoptotic bodies of HiBEC that has led to the identification of proteins uniquely expressed by HiBECs with a potential pathogenic role in PBC. Indeed this data highlights the role of the innate immune system activation in PBC, in which NF-kB pathway involvement has been strengthened in recent GWAS studies (54–56). Further, these results also suggest that the protein content within apoptotic bodies from HiBEC could be involved in biliary proliferation. After liver injury, Notch signaling pathway activation leads to biliary regeneration and proliferation through bipotent hepatic progenitor cells polarization (57). Finally, IL-8 and CXCR2 activation may lead to inflammatory cell recruitment (58).
The current results therefore extend our knowledge about the immunological properties of HiBECs, which indicate that they are more than an innocent victim in the pathogenesis of PBC. However, it is important to emphasize that proteomic pathway analysis is primarily a tool to generate a hypothesis on the mechanisms and protein involvement in the local inflammatory response and will require further testing. Future work will address whether the activity of these proteins in apoptotic bodies and their ability to directly or indirectly activate and/or modulate macrophage function.
Acknowledgments
Financial support: Funding supported by National Institutes of Health grant, DK39588.
Abbreviations
- PBC
Primary biliary cirrhosis
- PDC-E2
E2 subunit of the pyruvate dehydrogenase complex
- BECs
biliary epithelial cells
- HiBECs
human intrahepatic biliary cells
- LC-MS/MS
liquid chromatography-tandem mass spectrometry
- HRPTEpiC
Human Renal Proximal Tubular Epithelial cells
- BrEPC
and human bronchial epithelial cells
- FBS
fetal bovine serum
- GLM
Generalized Linear Modeling
- FDR
False Discovery Rate
- LRP
low density lipoprotein receptor
- LRP1
LDL receptor-related protein 1
- CD91
cluster of differentiation 91
- HSPS
heat shock proteins
- TLRs
Toll-like receptors
Contributor Information
Ana Lleo, Email: ana.lleo@humanitas.it.
Weici Zhang, Email: ddzhang@ucdavis.edu.
W. Hayes McDonald, Email: hayes.mcdonald@Vanderbilt.edu.
Erin H. Seeley, Email: erin.h.seeley@Vanderbilt.edu.
Patrick S.C. Leung, Email: psleung@ucdavis.edu.
Ross L. Coppel, Email: ross.coppel@monash.edu.
Aftab A. Ansari, Email: pathaaa@emory.edu.
David H. Adams, Email: d.h.adams@bham.ac.uk.
Simon Afford, Email: s.c.afford@bham.ac.uk.
Pietro Invernizzi, Email: pietro.invernizzi@humanitas.it.
M. Eric Gershwin, Email: megershwin@ucdavis.edu.
REFERENCES
- 1.Chen RC, Naiyanetr P, Shu SA, Wang J, Yang GX, Kenny TP, Guggenheim KC, et al. Antimitochondrial antibody heterogeneity and the xenobiotic etiology of primary biliary cirrhosis. Hepatology. 2013;57:1498–1508. doi: 10.1002/hep.26157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Gershwin ME, Coppel RL, Bearer E, Peterson MG, Sturgess A, Mackay IR. Molecular cloning of the liver-specific rat F antigen. J Immunol. 1987;139:3828–3833. [PubMed] [Google Scholar]
- 3.Gershwin ME, Mackay IR, Sturgess A, Coppel RL. Identification and specificity of a cDNA encoding the 70 kd mitochondrial antigen recognized in primary biliary cirrhosis. J Immunol. 1987;138:3525–3531. [PubMed] [Google Scholar]
- 4.Gershwin ME, Van de Water J. Cholangiocytes and primary biliary cirrhosis: prediction and predication. J Clin Invest. 2001;108:187–188. doi: 10.1172/JCI13583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Jin Q, Moritoki Y, Lleo A, Tsuneyama K, Invernizzi P, Moritoki H, Kikuchi K, et al. Comparative analysis of portal cell infiltrates in antimitochondrial autoantibody-positive versus antimitochondrial autoantibody-negative primary biliary cirrhosis. Hepatology. 2012;55:1495–1506. doi: 10.1002/hep.25511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Kaplan MM, Gershwin ME. Primary biliary cirrhosis. N Engl J Med. 2005;353:1261–1273. doi: 10.1056/NEJMra043898. [DOI] [PubMed] [Google Scholar]
- 7.Watt FE, James OF, Jones DE. Patterns of autoimmunity in primary biliary cirrhosis patients and their families: a population-based cohort study. QJM. 2004;97:397–406. doi: 10.1093/qjmed/hch078. [DOI] [PubMed] [Google Scholar]
- 8.Lleo A, Bowlus CL, Yang GX, Invernizzi P, Podda M, Van de Water J, Ansari AA, et al. Biliary apotopes and anti-mitochondrial antibodies activate innate immune responses in primary biliary cirrhosis. Hepatology. 2010;52:987–998. doi: 10.1002/hep.23783. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Lleo A, Selmi C, Invernizzi P, Podda M, Coppel RL, Mackay IR, Gores GJ, et al. Apotopes and the biliary specificity of primary biliary cirrhosis. Hepatology. 2009;49:871–879. doi: 10.1002/hep.22736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Lleo A, Selmi C, Invernizzi P, Podda M, Gershwin ME. The consequences of apoptosis in autoimmunity. J Autoimmun. 2008;31:257–262. doi: 10.1016/j.jaut.2008.04.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Lleo A, Shimoda S, Ishibashi H, Gershwin ME. Primary biliary cirrhosis and autoimmune hepatitis: apotopes and epitopes. J Gastroenterol. 2011;46(Suppl 1):29–38. doi: 10.1007/s00535-010-0303-8. [DOI] [PubMed] [Google Scholar]
- 12.Rong G, Zhong R, Lleo A, Leung PS, Bowlus CL, Yang GX, Yang CY, et al. Epithelial cell specificity and apotope recognition by serum autoantibodies in primary biliary cirrhosis. Hepatology. 2011;54:196–203. doi: 10.1002/hep.24355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Selmi C, Lleo A, Pasini S, Zuin M, Gershwin ME. Innate immunity and primary biliary cirrhosis. Curr Mol Med. 2009;9:45–51. doi: 10.2174/156652409787314525. [DOI] [PubMed] [Google Scholar]
- 14.Balan V, Ruppert K, Demetris AJ, Ledneva T, Duquesnoy RJ, Detre KM, Wei YL, et al. Long-term outcome of human leukocyte antigen mismatching in liver transplantation: results of the National Institute of Diabetes and Digestive and Kidney Diseases Liver Transplantation Database. Hepatology. 2008;48:878–888. doi: 10.1002/hep.22435. [DOI] [PubMed] [Google Scholar]
- 15.Manousou P, Arvaniti V, Tsochatzis E, Isgro G, Jones K, Shirling G, Dhillon AP, et al. Primary biliary cirrhosis after liver transplantation: influence of immunosuppression and human leukocyte antigen locus disparity. Liver Transpl. 2010;16:64–73. doi: 10.1002/lt.21960. [DOI] [PubMed] [Google Scholar]
- 16.McDonald WH, Yates JR., 3rd Shotgun proteomics and biomarker discovery. Dis Markers. 2002;18:99–105. doi: 10.1155/2002/505397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Wu CC, Yates JR., 3rd The application of mass spectrometry to membrane proteomics. Nat Biotechnol. 2003;21:262–267. doi: 10.1038/nbt0303-262. [DOI] [PubMed] [Google Scholar]
- 18.Boja ES, Rodriguez H. Mass spectrometry-based targeted quantitative proteomics: achieving sensitive and reproducible detection of proteins. Proteomics. 2012;12:1093–1110. doi: 10.1002/pmic.201100387. [DOI] [PubMed] [Google Scholar]
- 19.Li M, Gray W, Zhang H, Chung CH, Billheimer D, Yarbrough WG, Liebler DC, et al. Comparative shotgun proteomics using spectral count data and quasi-likelihood modeling. J Proteome Res. 2010;9:4295–4305. doi: 10.1021/pr100527g. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Picotti P, Aebersold R. Selected reaction monitoring-based proteomics: workflows, potential, pitfalls and future directions. Nat Methods. 2012;9:555–566. doi: 10.1038/nmeth.2015. [DOI] [PubMed] [Google Scholar]
- 21.Strain A, Wallace L, Joplin R, Daikuhara Y, Ishii T, Kelly D, Neuberger J. Characterization of biliary epithelial cells isolated from needle biopsies of human liver in the presence of hepatocyte growth factor. The American journal of pathology. 1995;146:537. [PMC free article] [PubMed] [Google Scholar]
- 22.Yang Y, Raper S, Cohn J, Engelhardt J, Wilson J. An approach for treating the hepatobiliary disease of cystic fibrosis by somatic gene transfer. Proceedings of the National Academy of Sciences of the United States of America. 1993;90:4601. doi: 10.1073/pnas.90.10.4601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Joplin R, Strain AJ, Neuberger JM. Immuno-isolation and culture of biliary epithelial cells from normal human liver. In Vitro Cell Dev Biol. 1989;25:1189–1192. doi: 10.1007/BF02621273. [DOI] [PubMed] [Google Scholar]
- 24.Drudi Metalli V, Mancino MG, Mancino A, Torrice A, Gatto M, Attili AF, Alpini G, et al. Bile salts regulate proliferation and apoptosis of liver cells by modulating the IGF1 system. Dig Liver Dis. 2007;39:654–662. doi: 10.1016/j.dld.2007.03.008. [DOI] [PubMed] [Google Scholar]
- 25.MacCoss MJ, McDonald WH, Saraf A, Sadygov R, Clark JM, Tasto JJ, Gould KL, et al. Shotgun identification of protein modifications from protein complexes and lens tissue. Proc Natl Acad Sci U S A. 2002;99:7900–7905. doi: 10.1073/pnas.122231399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Tabb DL, Fernando CG, Chambers MC. MyriMatch: highly accurate tandem mass spectral peptide identification by multivariate hypergeometric analysis. J Proteome Res. 2007;6:654–661. doi: 10.1021/pr0604054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc Ser B. 1995;57(2):289–300. [Google Scholar]
- 28.Schaefer CF, Anthony K, Krupa S, Buchoff J, Day M, Hannay T, Buetow KH. PID: the Pathway Interaction Database. Nucleic Acids Res. 2009;37:D674–D679. doi: 10.1093/nar/gkn653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Franceschini A, Szklarczyk D, Frankild S, Kuhn M, Simonovic M, Roth A, Lin J, et al. STRING v9.1: protein-protein interaction networks, with increased coverage and integration. Nucleic Acids Res. 2012 doi: 10.1093/nar/gks1094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Guicciardi ME, Gores GJ. Bile acid-mediated hepatocyte apoptosis and cholestatic liver disease. Dig Liver Dis. 2002;34:387–392. doi: 10.1016/s1590-8658(02)80033-0. [DOI] [PubMed] [Google Scholar]
- 31.Kikuchi K, Hsu W, Hosoya N, Moritoki Y, Kajiyama Y, Kawai T, Takai A, et al. Ursodeoxycholic acid reduces CpG-induced IgM production in patients with primary biliary cirrhosis. Hepatol Res. 2009;39:448–454. doi: 10.1111/j.1872-034X.2008.00474.x. [DOI] [PubMed] [Google Scholar]
- 32.Singh K, Deshpande P, Li G, Yu M, Pryshchep S, Cavanagh M, Weyand CM, et al. K-RAS GTPase- and B-RAF kinase-mediated T-cell tolerance defects in rheumatoid arthritis. Proc Natl Acad Sci U S A. 2012;109:E1629–E1637. doi: 10.1073/pnas.1117640109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Billiard F, Lobry C, Darrasse-Jeze G, Waite J, Liu X, Mouquet H, DaNave A, et al. Dll4-Notch signaling in Flt3-independent dendritic cell development and autoimmunity in mice. J Exp Med. 2012;209:1011–1028. doi: 10.1084/jem.20111615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Liu L, Belkadi A, Darnall L, Hu T, Drescher C, Cotleur AC, Padovani-Claudio D, et al. CXCR2-positive neutrophils are essential for cuprizone-induced demyelination: relevance to multiple sclerosis. Nat Neurosci. 2010;13:319–326. doi: 10.1038/nn.2491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Nagata S, Hanayama R, Kawane K. Autoimmunity and the clearance of dead cells. Cell. 2010;140:619–630. doi: 10.1016/j.cell.2010.02.014. [DOI] [PubMed] [Google Scholar]
- 36.Zelenay S, Keller AM, Whitney PG, Schraml BU, Deddouche S, Rogers NC, Schulz O, et al. The dendritic cell receptor DNGR-1 controls endocytic handling of necrotic cell antigens to favor cross-priming of CTLs in virus-infected mice. J Clin Invest. 2012;122:1615–1627. doi: 10.1172/JCI60644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Iborra S, Izquierdo HM, Martinez-Lopez M, Blanco-Menendez N, Reis e Sousa C, Sancho D. The DC receptor DNGR-1 mediates cross-priming of CTLs during vaccinia virus infection in mice. J Clin Invest. 2012;122:1628–1643. doi: 10.1172/JCI60660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Allen NJ, Bennett ML, Foo LC, Wang GX, Chakraborty C, Smith SJ, Barres BA. Astrocyte glypicans 4 and 6 promote formation of excitatory synapses via GluA1 AMPA receptors. Nature. 2012;486:410–414. doi: 10.1038/nature11059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Avanesov A, Honeyager SM, Malicki J, Blair SS. The role of glypicans in Wnt inhibitory factor-1 activity and the structural basis of Wif1’s effects on Wnt and Hedgehog signaling. PLoS Genet. 2012;8:e1002503. doi: 10.1371/journal.pgen.1002503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Kim MS, Saunders AM, Hamaoka BY, Beachy PA, Leahy DJ. Structure of the protein core of the glypican Dally-like and localization of a region important for hedgehog signaling. Proc Natl Acad Sci U S A. 2011;108:13112–13117. doi: 10.1073/pnas.1109877108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Dejima K, Kanai MI, Akiyama T, Levings DC, Nakato H. Novel contact-dependent bone morphogenetic protein (BMP) signaling mediated by heparan sulfate proteoglycans. J Biol Chem. 2011;286:17103–17111. doi: 10.1074/jbc.M110.208082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Karlsen TH, Franke A, Melum E, Kaser A, Hov JR, Balschun T, Lie BA, et al. Genome-wide association analysis in primary sclerosing cholangitis. Gastroenterology. 2010;138:1102–1111. doi: 10.1053/j.gastro.2009.11.046. [DOI] [PubMed] [Google Scholar]
- 43.Jung Y, McCall SJ, Li YX, Diehl AM. Bile ductules and stromal cells express hedgehog ligands and/or hedgehog target genes in primary biliary cirrhosis. Hepatology. 2007;45:1091–1096. doi: 10.1002/hep.21660. [DOI] [PubMed] [Google Scholar]
- 44.Herz J, Strickland DK. LRP: a multifunctional scavenger and signaling receptor. J Clin Invest. 2001;108:779–784. doi: 10.1172/JCI13992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Binder RJZY, Messmer MN, Pawaria S. CD91-Dependent Modulation of Immune Responses by Heat Shock Proteins: A Role in Autoimmunity. Autoimmune Diseases. 2012 doi: 10.1155/2012/863041. 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Fischer N, Haug M, Kwok WW, Kalbacher H, Wernet D, Dannecker GE, Holzer U. Involvement of CD91 and scavenger receptors in Hsp70-facilitated activation of human antigen-specific CD4+ memory T cells. Eur J Immunol. 2010;40:986–997. doi: 10.1002/eji.200939738. [DOI] [PubMed] [Google Scholar]
- 47.Jehle AW, Gardai SJ, Li S, Linsel-Nitschke P, Morimoto K, Janssen WJ, Vandivier RW, et al. ATP-binding cassette transporter A7 enhances phagocytosis of apoptotic cells and associated ERK signaling in macrophages. J Cell Biol. 2006;174:547–556. doi: 10.1083/jcb.200601030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Pawaria S, Binder RJ. CD91-dependent programming of T-helper cell responses following heat shock protein immunization. Nat Commun. 2011;2:521. doi: 10.1038/ncomms1524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Husebye H, Aune MH, Stenvik J, Samstad E, Skjeldal F, Halaas O, Nilsen NJ, et al. The Rab11a GTPase controls Toll-like receptor 4-induced activation of interferon regulatory factor-3 on phagosomes. Immunity. 2010;33:583–596. doi: 10.1016/j.immuni.2010.09.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Mao TK, Lian ZX, Selmi C, Ichiki Y, Ashwood P, Ansari AA, Coppel RL, et al. Altered monocyte responses to defined TLR ligands in patients with primary biliary cirrhosis. Hepatology. 2005;42:802–808. doi: 10.1002/hep.20859. [DOI] [PubMed] [Google Scholar]
- 51.Nagaleekar VK, Diehl SA, Juncadella I, Charland C, Muthusamy N, Eaton S, Haynes L, et al. IP3 receptor-mediated Ca2+ release in naive CD4 T cells dictates their cytokine program. J Immunol. 2008;181:8315–8322. doi: 10.4049/jimmunol.181.12.8315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Steinckwich N, Schenten V, Melchior C, Brechard S, Tschirhart EJ. An essential role of STIM1, Orai1, and S100A8-A9 proteins for Ca2+ signaling and FcgammaR-mediated phagosomal oxidative activity. J Immunol. 2011;186:2182–2191. doi: 10.4049/jimmunol.1001338. [DOI] [PubMed] [Google Scholar]
- 53.Schiller M, Bekeredjian-Ding I, Heyder P, Blank N, Ho AD, Lorenz HM. Autoantigens are translocated into small apoptotic bodies during early stages of apoptosis. Cell Death Differ. 2008;15:183–191. doi: 10.1038/sj.cdd.4402239. [DOI] [PubMed] [Google Scholar]
- 54.Kar SP, Seldin MF, Chen W, Lu E, Hirschfield GM, Invernizzi P, Heathcote J, et al. Pathway-based analysis of primary biliary cirrhosis genome-wide association studies. Genes Immun. 2013;14:179–186. doi: 10.1038/gene.2013.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Lleo A, Gershwin ME, Mantovani A, Invernizzi P. Towards common denominators in primary biliary cirrhosis: the role of IL-12. J Hepatol. 2012;56:731–733. doi: 10.1016/j.jhep.2011.05.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Mells GF, Floyd JA, Morley KI, Cordell HJ, Franklin CS, Shin SY, Heneghan MA, et al. Genome-wide association study identifies 12 new susceptibility loci for primary biliary cirrhosis. Nat Genet. 2011;43:329–332. doi: 10.1038/ng.789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Boulter L, Govaere O, Bird TG, Radulescu S, Ramachandran P, Pellicoro A, Ridgway RA, et al. Macrophage-derived Wnt opposes Notch signaling to specify hepatic progenitor cell fate in chronic liver disease. Nat Med. 2012;18:572–579. doi: 10.1038/nm.2667. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Zimmermann HW, Seidler S, Gassler N, Nattermann J, Luedde T, Trautwein C, Tacke F. Interleukin-8 is activated in patients with chronic liver diseases and associated with hepatic macrophage accumulation in human liver fibrosis. PLoS One. 2011;6:e21381. doi: 10.1371/journal.pone.0021381. [DOI] [PMC free article] [PubMed] [Google Scholar]


