Abstract
Xbp1, a key mediator of the unfolded protein response (UPR), is activated by IRE1α-mediated splicing, which results in a frameshift to encode a protein with transcriptional activity. However, the direct function of Xbp1 in epithelial cells during mammary gland development is unknown. Here we report that the loss of Xbp1 in the mammary epithelium through targeted deletion leads to poor branching morphogenesis, impaired terminal end bud formation, and spontaneous stromal fibrosis during the adult virgin period. Additionally, epithelial Xbp1 deletion induces endoplasmic reticulum (ER) stress in the epithelium and dramatically inhibits epithelial proliferation and differentiation during lactation. The synthesis of milk and its major components, α/β-casein and whey acidic protein (WAP), is significantly reduced due to decreased prolactin receptor (Prlr) and ErbB4 expression in Xbp1-deficient mammary epithelium. Reduction of Prlr and ErbB4 expression and their diminished availability at the cell surface lead to reduced phosphorylated Stat5, an essential regulator of cell proliferation and differentiation during lactation. As a result, lactating mammary glands in these mice produce less milk protein, leading to poor pup growth and postnatal death. These findings suggest that the loss of Xbp1 induces a terminal UPR which blocks proliferation and differentiation during mammary gland development.
INTRODUCTION
The primary function of the mammary gland is to provide nutrition for newborns through production of milk protein and lipids (1). These milk proteins are synthesized in the endoplasmic reticulum (ER) and are secreted into the mammary duct as classical secretory proteins (2). The mammary gland undergoes dramatic, continual developmental changes throughout adulthood and provides a valuable model through which to track the interplay between secretory pathway competence and epithelial cell maturation during postnatal development (3). Development of the mammary gland is governed by hormonal stimuli, which include the prolactin/ErbB4/Stat5 signaling axis (4–8). During pregnancy the mammary epithelium grows and branches until midpregnancy and differentiates functionally during late pregnancy and the early postpartum period. This epithelial differentiation is accompanied by the expression of milk protein genes, such as whey acidic protein (WAP) and α/β-casein, and by the production of milk droplets (6).
The ER has a crucial role in quality control during the folding and secretion of secretory proteins. The accumulation of misfolded proteins in the ER provokes ER stress by increasing the demand for energy, chaperones, and other proteins that are needed to fold client proteins or to degrade unfoldable secretory cargo. This stress activates a signaling network called the unfolded protein response (UPR). The UPR increases the folding capacity of the secretory pathway through the transcription and the upregulation of ER chaperones and foldases and the ER quality control machinery. Xbp1 is one master regulator of the UPR. It is produced as an RNA that is regulated by IRE1α-mediated cytoplasmic splicing of Xbp1 mRNA, resulting in a frameshift that then creates an mRNA that encodes a transcriptionally active protein (spliced Xbp1 [sXbp1]). Spliced Xbp1 regulates the transcription of a number of ER quality control genes and is essential for the development, survival, and function of intestinal epithelial cells, immune cells, and hepatocytes (9) and of adipocytes (10) as well as for the development of professional secretory cells, such as B cells, hepatocytes, and pancreatic β cells (11, 12). Two recent studies have characterized the contribution of the UPR to lactation. The first has implicated the PERK arm of the UPR in regulating lipogenesis in the mammary epithelium during lactation (13). In the second study, adipocyte Xbp1 was proven to be essential for the activity of the lactating gland, and Xbp1 splicing in adipocytes was induced by the lactogenic hormone prolactin. However, these changes in Xbp1 splicing in adipocytes did not alter the milk composition, mammary lipogenic activity, or mammary function, as assessed by determination of the level of Stat5 phosphorylation or expression of prolactin or the ErbB4 receptor (10). Accordingly, the contribution of mammary epithelium Xbp1 to breast epithelial cell function and development is unknown.
We have recently reported that ER stress induces fibrogenic activity in hepatic stellate cells, which are the key fibrogenic cells in the liver, through activation of IRE1α/Xbp1 signaling (14). This observation raised the possibility that there are pathophysiological levels of chronic ER stress associated with tissue fibrosis. During our study to explore the role of Xbp1 in mesenchymal cells using the Cre-loxP genetic recombination system using human glial fibrillary acid protein (hGFAP)-Cre mice (15–18) crossed to Xbp1flox/flox mice, we discovered that in Xbp1flox/flox;hGFAP-Cre transgenic mice, Xbp1 effectively targeted mammary epithelial cells rather than mesenchymal cells. In these animals, we observed that chronic ER stress caused by the loss of Xbp1 in mammary epithelial cells induces alterations in the ductal epithelium and spontaneous stromal fibrosis in the mammary glands and that these changes disrupt mammary gland development, with impaired epithelial cell proliferation and differentiation. Thus, serendipitously, the present study reveals a previously unrecognized function for Xbp1 in the regulation of mammary gland development.
MATERIALS AND METHODS
Generation and breeding of Xbp1flox/flox; GFAP-Cre mice.
Xbp1flox/flox mice were a gift from Laurie H. Glimcher (Weill Cornell Medical College). Xbp1flox/flox mice, which had the C57BL/6 background and which were described previously (19), were crossed with a transgenic FVB line expressing Cre recombinase under the control of the hGFAP promoter (GFAP-Cre) (15) to generate Xbp1flox/flox; GFAP-Cre mice. For cell fate studies, hGFAP-Cre mice were crossed with B6.129(Cg)-Gt(ROSA)26Sortm4(ACTB-tdTomato,-EGFP)Luo/J reporter mice (20). A breeding strategy that ensured that control Xbp1flox/flox and experimental mice Xbp1flox/flox; GFAP-Cre mice were always littermates was followed. Xbp1flox/flox and Xbp1flox/flox; GFAP-Cre dams that delivered a litter within 24 h of each other were used in cross-fostering experiments. The litters born to Xbp1flox/flox; GFAP-Cre and Xbp1flox/flox dams were swapped as described previously (21). All experimental mice were anesthetized (ketamine at 100 mg/kg of body weight, xylazine at 20 mg/kg) and humanely euthanized prior to collection of tissue; the conduct of our experiments has complied with the highest international criteria for studies with animals. All studies were approved by the Institutional Animal Care and Use Committee of the Icahn School of Medicine at Mount Sinai and followed the National Institutes of Health guidelines for animal care.
Antibodies.
The antibodies used in this study were as follows: anti-GFP antibody (1:1,000; catalog number ab6556; Abcam), anti-keratin 8/18 (anti-K8/18) antibody (1:200; Progen Biotechnik), anti-smooth muscle actin α (anti-α-SMA) conjugated with Cy3 antibody (1:500; catalog number C6198; Sigma, St. Louis, MO), anti-Xbp1 antibody (1:200; catalog number M-186; Santa Cruz), anti-protein disulfide isomerase (anti-PDI) antibody (catalog number 3501; Cell Signaling), anti-Bip antibody (1:1,000; catalog number 3177; Cell Signaling), anti-CHOP antibody (1:1,000; catalog number 2895; Cell Signaling), anti-IRE1α antibody (1:1,000; catalog number 3294; Cell Signaling), anti-GAPDH (anti-glyceraldehyde-3-phosphate dehydrogenase) antibody (1:5,000; catalog number ab9482; Abcam), anti-Ki67 antibody (1:2,500; catalog number ab15580; Abcam), anti-phospho-histone H3 (Ser10) antibody (1:300; catalog number 9701; Cell Signaling), anti-cleaved caspase 3 (Asp175) antibody (1:300; catalog number 9661; Cell Signaling), anti-phospho-Stat5 (Tyr694; C71E5) antibody (1:400 for immunohistochemistry and 1:1,000 for Western blotting; catalog number 9314; Cell Signaling), anti-Stat5 antibody (1:1,000; catalog number 9363; Cell Signaling), and anti-mouse milk-specific protein antibody (1:5,000; Accurate Chemical).
Preparation of tissue protein and immunoblot analysis.
For total protein extraction, mouse mammary gland tissues and cells were lysed in radioimmunoprecipitation assay buffer (50 mM Tris-HCl, pH 8.0, 150 mM NaCl, 1% NP-40, 0.2% SDS) containing protease and phosphatase inhibitors (Roche). Protein content was measured by use of the Bio-Rad protein assay dye reagent (catalog number 500-0006). Proteins were resolved by the use of NuPAGE 4 to 12% bis-Tris gels (Invitrogen) and transferred to polyvinylidene difluoride membranes. Blots were probed with the primary antibody for overnight at 4°C, washed, and incubated with horseradish peroxidase-linked secondary anti-rabbit or anti-mouse IgG antibody (catalog numbers 7074 and 7076, respectively; Cell Signaling) for 1 h at room temperature. Blots were developed using the HyGlo quick spray reagent (Denville).
Genotyping PCR.
Xbp1flox/flox; hGFAP-Cre and ROSA26mT/mG; hGFAP-Cre mice were genotyped via PCR analysis, as previously described (19). The presence of the floxed Xbp1 allele was detected via PCR using mouse tail DNA as a template. A PCR product specific for the floxed Xbp1 allele was amplified by using the following primers: 5′-ACTTGCACCAACACTTGCCATTTC-3′ (forward) and 5′-CAAGGTGGTTCACTGCCTGTAATG-3′ (reverse). The PCR conditions were as follows: 94°C for 3 min, followed by 35 cycles at 94°C for 30 s, 56°C for 30 s, and 72°C for 30 s and then 72°C for 5 min (the PCR product was 141 bp for wild-type [WT× mice and 183 bp for floxed mice). The presence of the hGFAP-Cre transgene was detected by using the following PCR primers: 5′-CCTGGAAAATGCTTCTGTCCG-3′ (forward) and 5′-CAGGGTGTTATAAGCAATCCC-3′ (reverse). The PCR conditions were as follows: 94°C for 4 min, followed by 40 cycles at 94°C for 30 s, 60°C for 30 s, and 72°C for 1.5 min and then 72°C for 10 min (size of the PCR product, 400 bp).
Real-time PCR.
PCR primers for real-time PCR are summarized in Table S1 in the supplemental material. Fresh isolated mammary tissues were frozen and stored at −80°C until specimens were homogenized by bead milling using a TissueLyser LT apparatus (Qiagen, Hilden, Germany), which rapidly disrupts up to 12 samples simultaneously via high-speed vertical shaking (oscillation frequency, 50 Hz) with a bead (7 mm) in a sealed tube for 3 min. Then, mammary tissue RNA was extracted using Qiagen minicolumns and RNeasy minikits (Qiagen, Germantown, MD) with an on-column DNase treatment. One microgram of RNA was reverse transcribed using an RT Complete Double PrePrimed kit (Clontech, Mountain View, CA). FastStart SYBR green master mix (Roche, Indianapolis, IN) was used for PCR. The samples were analyzed in triplicate, and the data were analyzed by the use of Microsoft Excel software (Microsoft Corp., Redmond, WA), with the levels of gene expression being normalized to the level of β-actin gene expression.
Whole-mount analysis and histological analysis.
Whole-mount analysis of mouse mammary glands was performed as described previously (22). Mammary glands were excised and spread on microscope slides. The tissues were fixed in 10% formalin at 4°C, defatted in Carnoy's fixative, washed in 70% ethanol, hydrated by passage through decreasing ethanol concentrations, and stained with carmine alum stain (0.2% carmine, 0.5% aluminum potassium sulfate) overnight at room temperature. For histological analysis, mammary gland specimens were fixed overnight in 10% formalin at 4°C, dehydrated, and embedded in paraffin. Tissue blocks were sectioned into 5-μm-thick slices and stained with hematoxylin and eosin (H&E). Sirius red (Sigma) was used to determine collagen deposition. Sections were stained with Sirius red solution (saturated picric acid containing 0.1% Direct Red 80 and 0.1% fast green) to visualize collagen deposition. The relative fibrosis area was assessed on the basis of the findings for 20 fields of Sirius red-stained mammary gland sections from each animal. Whole-field areas were from the mammary luminal area, and each field was acquired at a ×400 field magnification and then analyzed using a computerized Bioquant morphometry system (R & M Biometrics, Nashville, TN). To evaluate the relative fibrosis area, the measured collagen area was divided by the net field area.
Immunohistochemistry and immunofluorescence.
Tissues were immediately fixed in 10% neutral buffered formalin for 24 h. After fixation, the tissue was immersed in 70% ethanol until processing. All tissues were processed simultaneously. The fixed tissues were dehydrated in ethanol, cleared in xylene, and embedded in paraffin blocks, which were cooled before sectioning. The mammary gland tissues were sectioned into 4-μm-thick slices, and the sections were mounted on silane-coated slides. For antigen activation, the slides were incubated in target retrieval solution (Dako) and heated for 30 min in a microwave oven. The slides were allowed to cool. Then, sections were used for immunohistochemistry with a Dako EnVision detection system using the immunoperoxidase method according to the Dako EnVision kit manual.
All samples subjected to immunohistochemistry staining were probed with the primary antibody overnight at 4°C. The slides were then washed three times in phosphate-buffered saline and incubated with secondary anti-rabbit or anti-mouse antibody for 1 h at room temperature. For immunofluorescence analysis, Alexa Fluor 488–donkey anti-rabbit IgG (H+L) antibody (1:500; Invitrogen) and Alexa Fluor 647–goat anti-guinea pig IgG (H+L) antibody (1:500; Life Technologies) were used as secondary antibodies. An isotype control was used to assess nonspecific binding. The slides were mounted with DAPI (4′,6-diamidino-2-phenylindole) ProLong Gold antifade reagent (catalog number P36931; Life Technologies, Carlsbad, CA) and examined in a Zeiss Axiophot photomicroscope (Carl Zeiss, Oberkochen, Germany). The cells in the stained sections that by diaminobenzidine staining were positive for Ki67, phosphorylated histone H3, and cleaved caspase 3 were counted under a microscope at a ×200 or ×400 field magnification, using 20 randomly selected microscopic fields per section.
Isolation of mouse mammary epithelial cells.
Mouse mammary epithelial cells were isolated as previously described, with minor modifications (23). Inguinal glands from ROSA26mT/mG; hGFAP-Cre and Xbp1flox/flox; hGFAP-Cre mice were removed aseptically, minced, and digested with collagenase at 37°C for 45 min. The digested glands were subsequently centrifuged at 1,000 rpm for 5 min, and the fat layer and supernatant were removed. The pellet was washed and incubated in red blood cell lysis buffer (catalog number R7757; Sigma, St. Louis, MO) for 2 min before centrifugation. The cells were then plated in Dulbecco modified Eagle medium (DMEM) with 10% fetal bovine serum and 1% penicillin-streptomycin and incubated for 30 min at 37°C to allow the selective attachment of fibroblasts. The supernatant was transferred into bovine serum albumin-coated tubes and washed once before resuspension in MCF10A medium (DMEM–F-12 medium supplemented with antibiotics, epidermal growth factor [20 ng/ml], hydrocortisone [0.5 μg/ml], insulin [10 μg/ml], cholera toxin [0.1 μg/ml], and 5% heat-inactivated horse serum) on a 10-cm dish coated with Matrigel (BD, San Jose, CA).
Mammary gland transplantation.
Transplantation experiments were performed as described previously (24). Pubescent 3- to 4-week old donor and host female mice were anesthetized, and midsagittal and oblique cuts were made in order to expose the 4th inguinal mammary gland. The fat pad in the mammary gland was cleared from the host epithelium by removing the tissue anterior to the lymph node. Epithelial fragments (2 mm3) of the donor mice were transplanted into the remaining cleared fat pad of the host (the fat pad from Xbp1flox/flox; GFAP-Cre mice was transplanted into Xbp1flox/flox mice and the fat pad of Xbp1flox/flox mice was transplanted into Xbp1flox/flox; GFAP-Cre mice) and the skin was closed. After 6 weeks, pregnant females that had received the transplants were sacrificed at day 7 of lactation. The transplanted glands were excised and processed for whole-mount analysis as described above.
Evaluation of serum prolactin level.
Xbp1flox/flox; GFAP-Cre (n = 3) and Xbp1flox/flox (n = 5) littermate mice were mated after mammary gland transplantation and sacrificed at day 7 of lactation. Blood was collected via inferior vena cava puncture and allowed to clot overnight at 4°C. Samples were centrifuged at 4,000 × g for 15 min, and serum was collected and stored at −80°C. Prolactin concentrations were assessed by prolactin mouse enzyme-linked immunosorbent assay (ELISA) by use of an ELISA kit (Abcam).
RT-PCR for Xbp1 splicing assay.
Reverse transcription-PCR (RT-PCR) analysis was performed using a 2× PCR master mix (Thermo). A PCR product specific for the spliced Xbp1 was amplified by using the following primers: 5′-ACACGCTTGGGAATGGACAC-3′ (forward) and 5′-CCATGGGAAGATGTTCTGGG-3′ (reverse). For spliced Xbp1 and cyclophilin detection by standard RT-PCR, the following program was used: (i) 94°C for 3 min, (ii) 30 cycles of 94°C for 30 s, 58°C for 30 s, and 72°C for 30 s, and (iii) 72°C for 10 min. The PCR products were separated by agarose gel electrophoresis to resolve the 171-bp (unspliced) and 145-bp (spliced) amplicons. RT-PCR was performed with cyclophilin mRNA as a loading control to validate cDNA synthesis.
Evaluation of serum cholesterol and triglyceride concentration.
Quantitative determination of serum cholesterol and triglyceride levels was performed by use of a kit from Pointe Scientific, Inc. (Canton, MI) and spectrophotometer analysis.
Statistical analysis.
Statistical analysis was performed using a commercially available software package (Prism, version 5.0; GraphPad Software). Data were tested by Student's t test. Differences were considered statistically significant at a P value of <0.05. All results were derived from at least 3 independent experiments.
RESULTS
Pups suckling on Xbp1flox/flox; GFAP-Cre dams fail to thrive.
In the process of breeding and expanding the Xbp1flox/flox; GFAP-Cre colonies to generate mesenchymal cell-specific Xbp1-knockout mice, we observed an unexpected postnatal loss of several pups. Xbp1flox/flox; GFAP-Cre mice were distinguished from control littermates by PCR with Cre and Xbp1 genotyping primers (Fig. 1A) and were born at Mendelian ratios (data not shown). However, in pups of Xbp1flox/flox; GFAP-Cre dams, the body size and weight were clearly decreased at day 10 compared to those of the pups of Xbp1flox/flox dams (Fig. 1B). Xbp1flox/flox; GFAP-Cre female mice had normal pregnancies and gave birth to pups of the same number and sex as those of Xbp1flox/flox control female mice (Table 1).
FIG 1.
Pups suckling on Xbp1flox/flox; GFAP-Cre dams fail to thrive. (A) Genotyping of Xbp1flox/flox; GFAP-Cre mice by PCR. The 183-bp loxP allele-specific PCR product was amplified from Xbp1flox/flox mice, while only the 141-bp WT allele-specific PCR product was amplified from Xbp1wt/wt mice. Both the 421-bp and the 371-bp PCR products were detected in Xbp1flox/wt mice. The 400-bp Cre-specific PCR product was detected only in GFAP-Cre mice. A schematic representation of the gene targeting strategy is shown in Fig. 3G. (B) Images of pups born to and suckled on Xbp1flox/flox or Xbp1flox/flox; GFAP-Cre dams on postnatal day 10. All litters were mixtures of pup genotypes (Xbp1flox/flox and Xbp1flox/flox; GFAP-Cre; n = 6 to 12 litters per genotype). Weight gain is dependent on the genotype of the dam and not that of the pups. (C) Postpartum growth curve of Xbp1flox/flox (n = 4) and Xbp1flox/flox; GFAP-Cre (n = 4) mice. (D) Body weights at day 1, day 2, and day 3 of the pups of Xbp1flox/flox mothers (n = 37 pups from 4 mothers) and Xbp1flox/flox; GFAP-Cre mothers (n = 38 pups from 4 mothers). The statistical significance of differences between Xbp1flox/flox and Xbp1flox/flox; GFAP-Cre mice was determined: ****, P < 0.0001; n.s., no significant difference. (E) Proportions of surviving pups of Xbp1flox/flox mothers (n = 4) and Xbp1flox/flox; GFAP-Cre mothers (n = 4). (F) Images of cross-fostered Xbp1flox/flox or Xbp1flox/flox; GFAP-Cre pups on postnatal day 7. All litters were mixtures of pup genotypes. Weight gain was dependent on the genotype of the fostering dam. (G) Postpartum growth curve for pups cross fostered by Xbp1flox/flox (n = 3) or Xbp1flox/flox; GFAP-Cre (n = 3) dams starting at postnatal day 1.
TABLE 1.
Average number of pups, by genotype and sex, from the pregnancies of Xbp1flox/flox and Xbp1flox/flox; GFAP-Cre damsa
| Pup sex | Avg no. (%) of pups | 
|
|---|---|---|
| Xbp1flox/flox (n = 5) | Xbp1flox/flox; GFAP-Cre (n = 5) | |
| Female | 3.8 (25.7) | 3.8 (25.7) | 
| Male | 4.0 (28.3) | 3.0 (20.3) | 
Data are for five Xbp1flox/flox dams and five Xbp1flox/flox; GFAP-Cre dams.
To investigate pup growth in detail during nursing, we recorded pup weights daily from the time of birth to day 20 (Fig. 1C). The weights of the pups from Xbp1flox/flox; GFAP-Cre dams were significantly decreased from postnatal 3 day compared to those of the pups from Xbp1flox/flox dams (Fig. 1C and D), which began to die at day 12 in the initial pregnancies, and all pups were dead by day 20 (Fig. 1E). Overall, these data indicate that pups born to Xbp1flox/flox; GFAP-Cre dams failed to thrive.
Next, we confirmed that the failure to grow was due to the dam phenotype, because pups of Xbp1flox/flox; GFAP-Cre dams were rescued by cross fostering by Xbp1flox/flox dams. In contrast, the pups of Xbp1flox/flox dams had impaired growth when fostered by Xbp1flox/flox; GFAP-Cre dams (Fig. 1F and G). These data suggest that Xbp1flox/flox; GFAP-Cre dams were unable to sufficiently nourish the newborn pups, arguing for a potential mammary gland phenotype.
GFAP-Cre labels mammary epithelial cells in the mammary gland.
Based on the observations described above, we sought to localize the specific Cre-expressing cells in female mammary glands. We generated ROSA26mT/mG; GFAP-Cre mice for lineage tracing using Cre reporter mice expressing double-fluorescent membrane-targeting tdTomato fluorescent protein (mT) and membrane-targeting green fluorescent protein (GFP) (mG) crossed with GFAP-Cre mice. This reporter mouse ubiquitously expresses mT prior to Cre activation and mG upon Cre activation (Fig. 2A). We first visualized Cre-mediated recombination in situ in whole-mount mammary gland tissue without fixation at the virgin stage and on day 7 of lactation. Although all cells, including mammary epithelium and adipocytes, expressed dtTomato red in ROSA26mT/mG control mice, Cre expression was observed in mammary branches only in ROSA26mT/mG; GFAP-Cre mice at the virgin stage and on day 7 of lactation, as indicated by GFP expression (Fig. 2B).
FIG 2.
GFAP-Cre labels mammary epithelium in ROSA26mT/mG; GFAP-Cre mice. (A) Schematic view of the strategy used to assess the degree of epithelium-specific recombination. GFAP-Cre mice were crossed to B6.129(Cg)-Gt(ROSA)26Sortm4(ACTB-tdTomato,-EGFP)Luo/J reporter mice. These reporter mice ubiquitously express membrane Tomato red (mT) prior to Cre activation and membrane GFP green (mG) upon Cre activation. (B) Images of whole-mount preparations of mouse mammary glands expressing endogenous fluorescent mT/mG proteins from female ROSA26mT/mG and ROSA26mT/mG; GFAP-Cre mice at the virgin stage (left) and lactation day 7 (right). (Inset at lower left) High-magnification image of Cre expression within branching mammary morphology in virgin ROSA26mT/mG; GFAP-Cre mice. (C) Immunofluorescence images of mammary glands from female ROSA26mT/mG; GFAP-Cre mice for membrane-targeting GFP and K8/18, mammary myoepithelial cell marker α-SMA, and DAPI at the virgin stage (left) and lactation day 7 (right) using paraffin sections. Membrane-targeting GFP expression was detected by use of a GFP antibody. (D) Immunofluorescence images of mammary epithelial cells isolated from virgin female ROSA26mT/mG; GFAP-Cre mice (12 weeks) for detection of membrane-targeting GFP and K8/18 and DAPI, and α-SMA (a mammary myoepithelial cell marker). Membrane-targeting GFP expression was detected by use of a GFP antibody.
Furthermore, to determine the Cre-expressing cell type(s) in mammary branches in ROSA26mT/mG; GFAP-Cre mice, we analyzed the colocalization of membrane-targeting GFP, the epithelial cell marker keratin 8/18 (K8/18), and the myoepithelial cell marker α-SMA in mammary gland sections and isolated primary epithelial cells by multiple-labeling fluorescent immunohistochemistry. Surprisingly, GFP expression was almost completely colocalized with K8/18 but not with α-SMA in these virgin and lactating female ROSA26mT/mG; GFAP-Cre mice (Fig. 2C and D). These data indicate that in these animals GFAP-Cre is expressed specifically in epithelial cells, most likely of luminal origin, in the mammary gland.
Xbp1 loss impairs mammary gland development and induces abnormal stromal fibrosis.
Next, to determine the effects of Xbp1 depletion on mammary morphogenesis, we performed whole-mount analysis and H&E staining of paraffin-embedded sections of mammary glands from Xbp1flox/flox; GFAP-Cre mice at the virgin stage, during early pregnancy (pregnancy day 9 [P9]) and late pregnancy (pregnancy day 16 [P16]), and during lactation (lactation day 7 [L7]). The mammary glands of Xbp1flox/flox; GFAP-Cre mice showed poor branching morphogenesis, impaired terminal end bud (TEB) formation at the virgin stage, and strongly impaired lobuloalveolar development during the early lactation period (Fig. 3A and B). Interestingly, during pregnancy there were no morphological differences between control and Xbp1flox/flox; GFAP-Cre mice (Fig. 3A and B). These data suggest that the loss of Xbp1 in mammary epithelial cells during lactation renders the gland unable to respond to the pups' suckling. Double immunofluorescence staining for K8/18 (red) and α-SMA (green) also demonstrated a lack of epithelial cell cohesiveness, with mammary epithelial cells being delocalized from the basement membrane and decreased cell numbers being noted in the luminal layer in virgin Xbp1flox/flox; GFAP-Cre mice (Fig. 3C, arrowheads). Furthermore, the stromal border of the ducts exhibited increased matrix deposition in Xbp1flox/flox; GFAP-Cre mice (Fig. 3D, top, arrowheads). To confirm collagen accumulation in the mammary glands of Xbp1flox/flox; GFAP-Cre mice, Sirius red staining, which specifically identifies collagen, was performed on sections of mammary gland from these animals at 12 weeks. Stromal fibrosis thickness was significantly higher in Xbp1flox/flox; GFAP-Cre mice than in control mice at the virgin stage (Fig. 3D, bottom, and E). These data suggest that epithelial Xbp1 deletion in the epithelium induces abnormal stromal fibrosis in the mammary gland during adulthood. To investigate whether macrophage infiltration is altered as part of an aberrant immune response in the abnormal stromal fibrosis of Xbp1flox/flox; GFAP-Cre mice, we performed F4/80 staining of mammary gland sections from Xbp1fl/fl; GFAP-Cre mice. Overall, the percentage of cells staining positive for F4/80 was low (<0.5% in each section), and their frequency and expression levels of F4/80 were not different between in Xbp1flox/flox control and Xbp1flox/flox; GFAP-Cre mice (data not shown).
FIG 3.
Mammary epithelial Xbp1 loss impairs mammary lobuloalveolar development during lactation. (A to C) Carmine alum-stained whole-mount mammary gland tissues (A), hematoxylin and eosin staining (B), and immunofluorescence images of mammary glands for detection of the K8/18 (red) mammary epithelial cell marker, the α-SMA (green) mammary myoepithelial cell marker, and DAPI (blue) (C) at the virgin stage (12 weeks [12w]), during pregnancy day 9 (early pregnancy) and pregnancy day 16 (late pregnancy), and during lactation day 7 from female Xbp1flox/flox and Xbp1flox/flox; GFAP-Cre mice using whole-mount tissues and paraffin sections. (D) Hematoxylin and eosin staining (HE) and Sirius red staining of paraffin sections of mammary gland tissue from Xbp1flox/flox and Xbp1flox/flox; GFAP-Cre mice at the virgin stage. (E) Quantification of the Sirius red-positive area. The statistical significance of the differences between Xbp1flox/flox (n = 5) and Xbp1flox/flox; GFAP-Cre (n = 4) mice was determined. *, P < 0.05. (F and I) Immunoblots of sXbp1 proteins using whole mammary gland extracts (F) and isolated primary mammary epithelial cells (I) from Xbp1flox/flox (n = 3) or Xbp1flox/flox; GFAP-Cre (n = 3) mice. GAPDH expression was used as a loading control. (G) Targeting strategy used for Xbp1 gene interruption. The diagram shows the wild-type Xbp1 locus, the targeting construct, and the targeted locus before and after homologous recombination. Exon 2 was deleted by mating the floxed mice with GFAP-Cre recombinase mice. Excision of Xbp1 was determined by the use of locus-specific primers. The primers (WT-205F and WT-272R) that detect the WT bind to a region in floxed exon 2, while the primers delta-F and delta-R, which detect the mutant with the Xbp1 deletion (the ΔXbp1 mutant) bind to a sequence present only in the recombined allele with deletion of exon 2 (combined red and green lines). These primers were used for qPCR to confirm the effect of Cre recombination of Xbp1. The primer sets are shown as arrows in the diagram. (H) The levels of expression of WT Xbp1 and mutant ΔXbp1 in mammary gland tissues from lactation day 7 samples were measured by quantitative real-time PCR, with β-actin mRNA used as a control. Values represent the amounts of each mRNA relative to those in control Xbp1flox/flox mice. The statistical significance of differences between Xbp1flox/flox (n = 3) and Xbp1flox/flox; GFAP-Cre (n = 3) mice was determined. **, P < 0.01; ****, P < 0.0001.
During lactation, Xbp1flox/flox; GFAP-Cre dams also dramatically lacked expansion of the mammary epithelial compartment (Fig. 3B). Although between pregnancy and parturition the large cytoplasmic lipid droplets (CLDs) are usually replaced by small lipid droplets outside the cells on the luminal surface (Fig. 3B) (25), CLDs remained in the luminal alveolar cells in the Xbp1flox/flox; GFAP-Cre mouse mammary glands (Fig. 3B). These data suggest that the physiological response to mammary epithelium differentiation may be impaired by the loss of Xbp1.
The sXbp1 protein is highly induced during lactation in mammary epithelium.
Next, we investigated whether sXbp1 is expressed in mammary glands in Xbp1flox/flox and Xbp1flox/flox; GFAP-Cre mice during adult mammary gland development by assessing whole mammary gland proteins at progressive developmental stages (at the virgin stage, during early pregnancy [P9] and late pregnancy [P16], and on L7). Interestingly, sXbp1 was detectable only during lactation and was almost completely absent in Xbp1flox/flox; GFAP-Cre mice (Fig. 3F). To confirm the activity of Cre recombinase on Xbp1 by GFAP-Cre in mammary glands, we also performed quantitative PCR (qPCR) using primers that detect either wild-type (WT) Xbp1 or the Xbp1 deletion (the ΔXbp1 mutant) only. To quantify WT Xbp1 mRNA, we used a reverse primer that binds to a region in the floxed exon 2, and to detect the ΔXbp1 deletion, we used a primer that binds to a sequence present only in mice with theΔXbp1 deletion. Relative to the level of expression of Xbp1 mRNA in whole mammary gland tissues from control, WT mice on lactation day 7, in Xbp1flox/flox; GFAP-Cre mice, the WT Xbp1 mRNA level was decreased 99% and the level of expression of the allele with the Xbp1 mRNA deletion was increased 1,000-fold compared to that in WT mice (Fig. 3G and H).
We further performed Western blotting of isolated mammary epithelial cell proteins from Xbp1flox/flox and Xbp1flox/flox; GFAP-Cre virgin and lactating mice (Fig. 3I). sXbp1 was completely absent in mammary epithelial cells isolated from lactating Xbp1flox/flox; GFAP-Cre mice (Fig. 3I). These data confirm adequate Cre recombination in the mammary epithelium, consistent with the findings for GFAP-Cre reporter mice.
Impaired lobuloalveolar development during lactation occurs only when Xbp1 is absent in mammary epithelial cells.
We used a mammary gland transplantation technique to determine whether the observed lactation defect results directly from epithelial Xbp1 loss or is an indirect, systemic effect due to Xbp1 loss in the brain or other cells of Xbp1flox/flox; GFAP-Cre mice. Transplantation was performed, in which gland tissue fragments from which Xbp1 was deleted were placed into cleared fat pads of Xbp1flox/flox recipients and Xbp1flox/flox gland tissue fragments were placed into Xbp1flox/flox; GFAP-Cre recipients to establish that the lactation defect is present only when the mammary gland lacks Xbp1 (Fig. 4A). At postnatal day 7, the weights of pups of Xbp1flox/flox; GFAP-Cre dams into which the Xbp1flox/flox pad was transplanted were significantly decreased compared to those of pups of Xbp1flox/flox dams into which the Xbp1flox/flox; GFAP-Cre pad was transplanted (Fig. 4B). Transplanted Xbp1flox/flox mammary glands developed normal alveologenesis during lactation in Xbp1flox/flox; GFAP-Cre mice (Fig. 4C). However, the mammary glands transplanted from Xbp1flox/flox; GFAP-Cre mice into Xbp1flox/flox mice demonstrated impaired alveologenesis during lactation (Fig. 4C). These data suggest that the lactation defect is present only when the mammary gland lacks Xbp1.
FIG 4.
Impaired lobuloalveolar development during lactation is present only when the mammary epithelium lacks Xbp1. (A) Schematic view of the mammary gland transplantation strategy using Xbp1flox/flox and Xbp1flox/flox; GFAP-Cre mice. KO, knockout. (B) Postpartum growth in the pups of Xbp1flox/flox (n = 5) and Xbp1flox/flox; GFAP-Cre (n = 3) dams transplanted with fat pads from mice of the other genotype. (C) Carmine alum-stained whole-mount mammary gland tissues from Xbp1flox/flox (n = 5) and Xbp1flox/flox; GFAP-Cre (n = 3) dams transplanted with fat pads from mice of the other genotype at lactation day 7. Native mammary glands and transplanted mammary glands are shown. (D) Serum prolactin hormone levels in Xbp1flox/flox (n = 5) and Xbp1flox/flox; GFAP-Cre (n = 3) dams transplanted with fat pads from mice of the other genotype at lactation day 7. ****, P < 0.0001; ns, no significant difference.
We also measured by ELISA serum prolactin levels in transplanted mice on lactation day 7. There were no significant differences in prolactin levels between Xbp1flox/flox and Xbp1flox/flox; GFAP-Cre mice (Fig. 4D). These data suggest that the phenotype did not result from the altered prolactin levels resulting from the loss of Xbp1 in the central nervous system.
Epithelial Xbp1 deficiency leads to impaired mammary alveologenesis via reduced proliferation.
Next, we investigated whether Xbp1 loss in luminal epithelial cells affected their proliferation by immunostaining for Ki67 on lactation day 7. There was a significant reduction in the number of Ki67-positive cells in Xbp1flox/flox; GFAP-Cre mice (Fig. 5A and D). The Ki67 mRNA level was also significantly reduced in Xbp1flox/flox; GFAP-Cre mice compared to that in the controls, as assessed by quantitative PCR (Fig. 5E). Furthermore, the number of phosphorylated histone H3 protein-positive cells in mammary glands was decreased in Xbp1flox/flox; GFAP-Cre mice (Fig. 5B and F). These findings indicate that the delay in mammary gland development in Xbp1-deficient mice during early lactation is caused by the reduced proliferation of epithelial cells.
FIG 5.
Xbp1 deficiency impairs mammary alveologenesis during lactation. (A to C) Immunostaining for Ki67 (A), phosphorylated histone H3 (p-histone H3) protein (B), and cleaved caspase 3 (C) in mammary glands from Xbp1flox/flox and Xbp1flox/flox; GFAP-Cre mice on lactation day 7. (D and F) The proliferation index of the glands from Xbp1flox/flox and Xbp1flox/flox; GFAP-Cre mice was determined by examining the nuclear expression of the Ki67 protein and phosphorylated histone H3 protein. A two-sample test for equality of proportions was used as the statistical test for comparison of the two proportions. (E) Quantitative RT-PCR for determination of the level of Ki67 mRNA expression using mammary gland tissues from samples collected on lactation day 7. (G) The apoptosis indexes of the glands from Xbp1flox/flox and Xbp1flox/flox; GFAP-Cre mice were determined by examining the nuclear expression of cleaved caspase 3. A two-sample test for the equality of proportions was used as the statistical test for comparison of the two proportions. The statistical significance of the differences between Xbp1flox/flox (n = 3) and Xbp1flox/flox; GFAP-Cre (n = 3) mice was determined. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ns, no significant difference.
To examine whether the hypomorphic state of mammary gland tissue was linked to an increase in apoptosis, we performed cleaved caspase 3 staining on day 7 of lactation. The percentage of cleaved caspase 3-positive cells was low (<0.5% in each section). They were commonly luminal, and their frequency and expression levels were not different between Xbp1flox/flox control mice and Xbp1flox/flox; GFAP-Cre mice (Fig. 5C and G). During normal lactation, only a small proportion of apoptotic cells are present in the mammary gland (26). Taken together, our data provide evidence that the loss of mammary gland tissue upon Xbp1 deletion is due to reduced proliferation and not increased apoptosis. This is surprising, as the loss of Xbp1 in the developing liver leads to apoptosis (27), which is likely due to the persistent ER stress that occurs in cells with a high secretory output when this important UPR regulator is missing.
ER stress in the mammary epithelium is provoked by Xbp1 deficiency.
In mammals, activation of IRE1α results in splicing of the Xbp1 mRNA to generate an active form of Xbp1 (spliced Xbp1 [sXbp1]) that initiates an arm of the UPR program (19). Therefore, to investigate the effect of Xbp1 loss on the UPR pathway, we analyzed the expression of UPR mRNAs and the IRE1α protein in the mammary glands of Xbp1flox/flox; GFAP-Cre mice by qPCR and immunoblotting, respectively. The expression level of the Xbp1 target chaperone protein protein disulfide isomerase (PDI) was significantly decreased in Xbp1flox/flox; GFAP-Cre mice, as were the mRNA levels of Edem1 and Hrd1, which are Xbp1 target genes in the endoplasmic reticulum-associated degradation (ERAD) pathway (Fig. 6A and B).
FIG 6.
Xbp1 deficiency provokes ER stress in mouse mammary glands during lactation. (A) Expression of IRE1α and Xbp1 target ERAD genes, such as Edem1 and Hrd1, in mammary gland tissues from samples collected on lactation day 7 was measured by quantitative real-time PCR with β-actin mRNA as a control. Values represent the amounts of each mRNA relative to those in control Xbp1flox/flox mice. (B) Immunoblot of UPR proteins IRE1α, BiP, CHOP, and PDI in whole mammary gland extracts from Xbp1flox/flox (n = 3) and Xbp1flox/flox; GFAP-Cre (n = 3) mice. GAPDH expression was used as a loading control. (C) Immunostaining for IRE1α in paraffin sections of mammary glands collected from Xbp1flox/flox and Xbp1flox/flox; GFAP-Cre mice on lactation day 7. (D) Levels of Xbp1 mRNA splicing from RNA of mammary gland tissues from Xbp1flox/flox (n = 3) or Xbp1flox/flox; GFAP-Cre (n = 3) mice collected on lactation day 7 were measured by reverse transcription-PCR. Statistically significant differences between Xbp1flox/flox (n = 3) and Xbp1flox/flox; GFAP-Cre (n = 3) mice are shown: *, P < 0.05; **, P < 0.01; ***, P < 0.001; ns, no significant difference.
In contrast, the ER stress marker proteins BiP, CHOP, and IRE1α were significantly induced in Xbp1flox/flox; GFAP-Cre mice (Fig. 6B). Although the IRE1α protein was induced in Xbp1flox/flox; GFAP-Cre mice, as previously reported in other systems (19), the Ire1α mRNA expression level was not changed between control and Xbp1flox/flox; GFAP-Cre mice (Fig. 6A), which is not surprising, given that this gene is not regulated at the transcriptional level. To identify the localization of IRE1α induction, we performed immunostaining for IRE1α on lactating mammary glands from Xbp1flox/flox; GFAP-Cre mice. IRE1α was highly expressed in the cells of the mammary gland ducts and alveoli during lactation (Fig. 6C). In addition, we confirmed by RT-PCR ΔXbp1 mRNA mutant splicing in Xbp1flox/flox; GFAP-Cre mice using extracts from whole mammary gland tissue (Fig. 6D). The splicing of the mutant ΔXbp1 mRNA was detected in Xbp1flox/flox; GFAP-Cre mice (Fig. 6D). These data suggest that epithelial Xbp1 loss leads to chronic ER stress and constitutive activation of its upstream activator IRE1α in the mammary epithelium. The fact that BiP and CHOP were also significantly upregulated suggests that ATF6 and PERK signaling might be induced. The induction of CHOP and possibly of PERK could explain the strong growth arrest phenotype, as both pathways can induce arrest at G0/G1 phase (28, 29).
Reduced quality and quantity of milk in the Xbp1flox/flox;GFAP-Cre mouse mammary gland during lactation.
The maturation and secretion of essential protein constituents of milk are dependent on the capacity of the ER to properly function, and Xbp1 is known to be required for the secretory ability of exocrine glands (11, 12). Thus, we measured the level of milk in isolated primary epithelial cells during lactation using an anti-mouse milk-specific antibody. The mouse milk protein production was significantly decreased in Xbp1flox/flox; GFAP-Cre mice (Fig. 7A). Furthermore, casein, which is a major component of mouse milk, has a significant effect on maturation of milk secretion and growth of the offspring (30). Therefore, we assessed the expression of genes for mouse milk constitutive proteins in whole mammary lactating gland tissue by qPCR. Interestingly, mRNA levels for major components of mouse milk, α/β-casein and whey acidic protein (WAP), were decreased by 75% in Xbp1flox/flox; GFAP-Cre mice (Fig. 7B). These data suggest that Xbp1 deficiency impairs the functional differentiation involved in the control of the quality and quantity of mouse milk.
FIG 7.
Reduced quality and quantity of mouse milk in the Xbp1flox/flox; GFAP-Cre mouse mammary gland during lactation. (A) Immunoblot of detection of mouse milk-specific protein (46 kDa) by anti-rabbit mouse milk-specific protein antibody using primary mammary epithelial cells isolated from the mammary glands of Xbp1flox/flox and Xbp1flox/flox; GFAP-Cre mice on lactation day 7. (B and C) Expression of α/β-casein and Wap genes for the milk component (B) and expression of Atf6, Acc2, Scd1, Srebp1, Chrebp, Ppar-γ, and Adrp for lipogenic genes (C) in whole mammary glands collected from Xbp1flox/flox (n = 3) or Xbp1flox/flox; GFAP-Cre (n = 3) mice on lactation day 7 were measured by quantitative real-time PCR with β-actin mRNA as a control. Values represent the amounts of each mRNA relative to those in control Xbp1flox/flox mice. The statistical significance of differences between Xbp1flox/flox (n = 3) and Xbp1flox/flox; GFAP-Cre (n = 3) mice was determined. *, P < 0.05; **, P < 0.01; ns, no significant difference. (D to G) Body weights (D), isolated inguinal fat pad weights (E), serum cholesterol levels (F), and triglyceride levels (G) of Xbp1flox/flox (n = 4) and Xbp1flox/flox; GFAP-Cre (n = 4) dams on lactation day 7. Body weights, isolated inguinal fat pad weights, and serum cholesterol and triglyceride levels were not significantly different between Xbp1flox/flox; GFAP-Cre and control mice. TG, triglyceride; BW, body weight; MG, mammary gland.
sXbp1 regulates the expression of genes encoding lipogenic enzymes, including Acc2 and Scd1, which are direct sXbp1 targets. The levels of Acc2 and Scd1 expression were significantly decreased in Xbp1flox/flox; GFAP-Cre mice (Fig. 7C). However, the expression of upstream master factors that control lipid metabolism, Srebp-1, Chrebp, Ppar-γ, and Adrp/adipophilin, were not altered (Fig. 7C). In addition, there was no compensatory induction of Atf6 mRNA, which is involved in de novo lipogenesis (31–33) and which is involved in another branch of UPR (Fig. 7C); however, it is possible that cleavage and activation of the Atf6 protein were altered by Xbp1 deletion. There were no significant differences between Xbp1flox/flox; GFAP-Cre and control mice in total body weight and isolated inguinal fat pad weight (Fig. 7D and E) or in serum cholesterol and triglyceride levels at lactation day 7 (Fig. 7F and G). Taken together, these data suggest that epithelial Xbp1 is also essential for the expression of genes involved in the maturation of milk production but not basal metabolic parameters.
Xbp1 loss disrupts Stat5 phosphorylation due to reduced prolactin and ErbB4 receptor expression.
Next, we studied the mechanism by which Xbp1 could be affecting mouse milk synthesis. We focused on the prolactin-Stat5 pathway, which regulates mouse milk synthesis, because Prlr (prolactin receptor) (34), ErbB4 (a tyrosine-protein kinase receptor) (35), and Stat5 (36) are essential for the development of secretory mammary epithelial cells during lactation, proliferation, and milk production (37). When we analyzed whole mammary gland tissue for the expression of the genes for Prlr, ErbB4, and Cav1 (caveolin-1, which is involved in JAK2 activation), the levels of the transcripts of these genes were significantly decreased in Xbp1flox/flox; GFAP-Cre mice; there was no difference in the level of Cav1 gene expression (Fig. 8A). These data show an association between Xbp1 loss and the disruption of Prlr/ErbB4 gene expression.
FIG 8.
Xbp1 loss disrupts Stat5 phosphorylation due to reduced prolactin and ErbB4 receptor expression. (A and D) Quantitative RT-PCR for determination of levels of Prlr, ErbB4, and Cav1 mRNA expression (A) and Stat5a and Stat5b mRNA expression (D) in samples of mammary gland tissues collected on lactation day 7. β-Actin mRNA was used as a control. Values represent the amounts of each mRNA relative to that in the control. (B) Immunostaining of phospho-Stat5 in mammary gland sections from Xbp1flox/flox and Xbp1flox/flox; GFAP-Cre mice collected on lactation day 7. (C) Immunoblot for phospho-Stat5 and total Stat5 from protein lysates in mammary glands from Xbp1flox/flox and Xbp1flox/flox; GFAP-Cre mice collected on lactation day 7. ns, no significant difference.
Because Prlr and ErbB4 regulate Stat5 tyrosine phosphorylation (pStat5) and nuclear translocation in secretory mammary epithelial cells during lactation, we compared Stat5 tyrosine phosphorylation levels in mammary gland sections from Xbp1flox/flox and Xbp1flox/flox; GFAP-Cre animals by immunostaining and immunoblot analysis (Fig. 8B and C). Interestingly, epithelial pStat5 was dramatically decreased on lactation day 7 according to both the number of nuclei stained and the protein levels obtained by immunoblotting (Fig. 8B and C). There were no differences in the expression of total Stat5 protein and Stat5a/b mRNA (Fig. 8C and D). These data suggest that Xbp1 loss disrupts Stat5 phosphorylation due to reduced prolactin and ErbB4 receptor expression in the mammary epithelium.
DISCUSSION
This work reveals a requirement for Xbp1 in mammary gland development and demonstrates that Xbp1 in mammary epithelial cells is essential for the maintenance of the luminal epithelium in the adult mammary gland (Fig. 9). Lactation represents one of the most highly regulated and productive functions of the secretory pathway, and here we show that adequate milk production requires an intact UPR. Without Xbp1, it is likely that the mammary epithelial cells can neither manage the secretory demand nor proliferate and expand, underscoring the vital role of UPR to support epithelial cell proliferation in conjunction with the growing demands of the ER client protein load as milk maturation proceeds during lactation.
FIG 9.
Proposed model highlighting the function of Xbp1 in the mouse lactating mammary gland.
Expansion and differentiation of the mouse mammary alveolar compartment during pregnancy and lactation are controlled through the prolactin receptor (38, 39), Jak2 (40), ErbB4 (41, 42), and Stat5 (43, 44). Activation of Stat5a and Stat5b by tyrosine phosphorylation is tightly linked to mammary gland differentiation to support milk protein and lipid production and requires signaling through prolactin- and ErbB4-dependent mechanisms. Our findings reveal that Xbp1 is involved in Stat5 phosphorylation and prolactin and ErbB4 receptor expression during epithelium differentiation in lactation. Xbp1 regulates not only the expression of key milk components, α/β-casein and WAP, which are required for milk production, but also the expression of key metabolic enzymes, ACC2 and SCD1, which are essential for lipid production during early lactation. In contrast, conditional deletion of PERK in mouse mammary epithelium affects milk lipid but not its protein composition (13). Thus, each UPR branch (e.g., PERK and IRE1-Xbp1) may contribute differently to the nutritional components of milk to optimize its quality. The direct link to the expression of these genes is not entirely clear, but it may involve direct regulation of these transcripts by Xbp1, or the changes in gene expression may result from alterations in the epithelium due to acute ER stress. Nevertheless, our study reveals a previously unrecognized role of Xbp1 and UPR signaling homeostasis of the secretory mammary epithelium during morphogenesis and lactation.
In addition to the alterations in UPR and milk production, two other aspects of our work are of interest. One is the stromal fibrosis associated with Xbp1 and the profound reduction in proliferation of the mammary epithelium in the absence of apoptosis also when Xbp1 is lost. Interaction between the epithelium and the extracellular matrix (ECM) plays a major role in controlling mammary gland branching morphogenesis, and this process can be altered when ductal invasion is disrupted upon the loss of factors that regulate the ECM (45). Proper side branching also requires that the ECM and the cellular microenvironment surrounding the ductal epithelium be maintained (45). Several studies suggest a prominent role of ER stress and activation of the UPR in fibrosis of the lungs, liver, gastrointestinal tract, kidney, and heart (46). ER stress may enhance the susceptibility of structural cells, in most cases, the epithelium, to profibrotic stimuli (46). Although the role of Xbp1 in organ fibrosis is still unclear, our data indicate that epithelial Xbp1 normally attenuates fibrosis within mammary stroma at puberty, as its deletion leads to increased stromal fibrosis. Our data also argue that since Xbp1 deletion is restricted to luminal cells, the myoepithelium or the stromal fibroblasts respond to the hypomorphic luminal epithelium by mounting a fibrotic response similar to that observed during wounding. It is possible that this stromal fibrosis contributes to impede proper branching morphogenesis within the mammary microenvironment.
The loss of Xbp1 primarily affected mammary epithelial cell proliferation (46), primarily by provoking arrest of the cell cycle at G0 phase. The lack of apoptosis in the ducts suggests that the changes in gene expression caused by Xbp1 loss were not activating a proapoptotic arm of the UPR but, rather, were suppressing growth, although in some other tissues (e.g., liver) the loss of Xbp1 during development leads to apoptosis (27). Thus, UPR induction may yield a different outcome depending on the context and inducing stimuli (47). In Xbp1-deficient epithelium, reduced proliferation could be due to the loss of prolactin and ErbB4 signaling (35, 48). However, ER stress can also directly inhibit cell proliferation through induction of the CDK inhibitor p21Cip1, leading to cell cycle arrest, for example, in chondrocytes (49) and prostate cancer cells (50). Specifically, PERK signaling can downregulate cyclin D1 concomitantly with p21Cip1 induction, causing arrest in G0/G1 phase. In mammary epithelium, PERK signaling reportedly limits the proliferation of mammary epithelial cells, and its loss contributes to premalignant cancer lesions (28), which also could lead to cancer initiation in mice deficient for PERK (51). While we did not measure the phosphorylation of PERK or the α subunit of eukaryotic initiation factor 2 (eIF2α) in Xbp1-deficient tissues, which is required for the growth arrest phenotypes cited above, because CHOP was upregulated upon Xbp1 loss and this transcription factor is a target of PERK signaling and can induce growth arrest (29), it is likely that the CHOP pathway contributes to growth arrest (52). However, other pathways could be involved, as CHOP can be induced by other stress signals (53).
The proproliferative function of Xbp1 might also have consequences in the long-term homeostasis of mammary tissue (26, 54, 55). Recent studies indicate that Xbp1 plays a critical role in the pathogenesis of triple-negative breast cancer by controlling the HIF1α pathway, which drives tumor cell survival, adaptation, and tumorigenesis (56). These vital clinical data underscore the importance of understanding how Xbp1 controls normal breast development and maturation. Our work provides the first mechanistic basis as to how during mammary gland life cycles Xbp1 signaling, if deregulated, might contribute to cancer development by maintaining high levels of growth factor and hormone signaling. However, future studies in the context of oncogene signaling will be needed to understand how Xbp1 and oncogenic pathways cooperate to induce breast cancer.
In summary, we have demonstrated that Xbp1 is essential for mammary epithelial differentiation and proliferation during lactation in the postnatal period by affecting mammary epithelial cell intrinsic pathways as well as microenvironmental mechanisms that affect proper mammary tissue homeostasis and morphogenesis.
Supplementary Material
ACKNOWLEDGMENTS
We are grateful to Laurie H. Glimcher for the helpful gift of Xbp1flox/flox mice, Kirsten C. Sadler for critically reading the manuscript, and Rosa Kim, Luke Noon, and Nina Linde for helpful discussions and technical assistance.
This work was supported by U.S. National Institutes of Health grants DK56621, AA020709, P20AA017067, and 1K05AA018408 to S.L.F., NIH/National Cancer Institute grant CA109182 and support from the Samuel Waxman Cancer Research Foundation to J.A.A.-G., and fellowships from the Astellas Foundation for Research on Metabolic Disorders to D.H.
The funders had no role in study design, data collection and analysis, the decision to publish, or preparation of the manuscript.
Footnotes
Supplemental material for this article may be found at http://dx.doi.org/10.1128/MCB.00136-15.
REFERENCES
- 1.Watson CJ, Khaled WT. 2008. Mammary development in the embryo and adult: a journey of morphogenesis and commitment. Development 135:995–1003. doi: 10.1242/dev.005439. [DOI] [PubMed] [Google Scholar]
 - 2.Le Parc A, Leonil J, Chanat E. 2010. AlphaS1-casein, which is essential for efficient ER-to-Golgi casein transport, is also present in a tightly membrane-associated form. BMC Cell Biol 11:65. doi: 10.1186/1471-2121-11-65. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 3.Bussard KM, Smith GH. 2011. The mammary gland microenvironment directs progenitor cell fate in vivo. Int J Cell Biol 2011:451676. doi: 10.1155/2011/451676. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 4.Topper YJ, Freeman CS. 1980. Multiple hormone interactions in the developmental biology of the mammary gland. Physiol Rev 60:1049–1106. [DOI] [PubMed] [Google Scholar]
 - 5.Forsyth IA. 1986. Variation among species in the endocrine control of mammary growth and function: the roles of prolactin, growth hormone, and placental lactogen. J Dairy Sci 69:886–903. doi: 10.3168/jds.S0022-0302(86)80479-9. [DOI] [PubMed] [Google Scholar]
 - 6.Robinson GW, McKnight RA, Smith GH, Hennighausen L. 1995. Mammary epithelial cells undergo secretory differentiation in cycling virgins but require pregnancy for the establishment of terminal differentiation. Development 121:2079–2090. [DOI] [PubMed] [Google Scholar]
 - 7.Hennighausen L, Robinson GW. 1998. Think globally, act locally: the making of a mouse mammary gland. Genes Dev 12:449–455. doi: 10.1101/gad.12.4.449. [DOI] [PubMed] [Google Scholar]
 - 8.Hennighausen L, Robinson GW. 2005. Information networks in the mammary gland. Nat Rev Mol Cell Biol 6:715–725. doi: 10.1038/nrm1714. [DOI] [PubMed] [Google Scholar]
 - 9.Glimcher LH. 2010. XBP1: the last two decades. Ann Rheum Dis 69(Suppl 1):i67–i71. doi: 10.1136/ard.2009.119388. [DOI] [PubMed] [Google Scholar]
 - 10.Gregor MF, Misch ES, Yang L, Hummasti S, Inouye KE, Lee AH, Bierie B, Hotamisligil GS. 2013. The role of adipocyte XBP1 in metabolic regulation during lactation. Cell Rep 3:1430–1439. doi: 10.1016/j.celrep.2013.03.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 11.Lee AH, Chu GC, Iwakoshi NN, Glimcher LH. 2005. XBP-1 is required for biogenesis of cellular secretory machinery of exocrine glands. EMBO J 24:4368–4380. doi: 10.1038/sj.emboj.7600903. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 12.Lee AH, Heidtman K, Hotamisligil GS, Glimcher LH. 2011. Dual and opposing roles of the unfolded protein response regulated by IRE1alpha and XBP1 in proinsulin processing and insulin secretion. Proc Natl Acad Sci U S A 108:8885–8890. doi: 10.1073/pnas.1105564108. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 13.Bobrovnikova-Marjon E, Hatzivassiliou G, Grigoriadou C, Romero M, Cavener DR, Thompson CB, Diehl JA. 2008. PERK-dependent regulation of lipogenesis during mouse mammary gland development and adipocyte differentiation. Proc Natl Acad Sci U S A 105:16314–16319. doi: 10.1073/pnas.0808517105. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 14.Hernandez-Gea V, Hilscher M, Rozenfeld R, Lim MP, Nieto N, Werner S, Devi LA, Friedman SL. 2013. Endoplasmic reticulum stress induces fibrogenic activity in hepatic stellate cells through autophagy. J Hepatol 59:98–104. doi: 10.1016/j.jhep.2013.02.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 15.Yang L, Jung Y, Omenetti A, Witek RP, Choi S, Vandongen HM, Huang J, Alpini GD, Diehl AM. 2008. Fate-mapping evidence that hepatic stellate cells are epithelial progenitors in adult mouse livers. Stem Cells 26:2104–2113. doi: 10.1634/stemcells.2008-0115. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 16.Scholten D, Osterreicher CH, Scholten A, Iwaisako K, Gu G, Brenner DA, Kisseleva T. 2010. Genetic labeling does not detect epithelial-to-mesenchymal transition of cholangiocytes in liver fibrosis in mice. Gastroenterology 139:987–998. doi: 10.1053/j.gastro.2010.05.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 17.Hernandez-Gea V, Ghiassi-Nejad Z, Rozenfeld R, Gordon R, Fiel MI, Yue Z, Czaja MJ, Friedman SL. 2012. Autophagy releases lipid that promotes fibrogenesis by activated hepatic stellate cells in mice and in human tissues. Gastroenterology 142:938–946. doi: 10.1053/j.gastro.2011.12.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 18.Lujambio A, Akkari L, Simon J, Grace D, Tschaharganeh DF, Bolden JE, Zhao Z, Thapar V, Joyce JA, Krizhanovsky V, Lowe SW. 2013. Non-cell-autonomous tumor suppression by p53. Cell 153:449–460. doi: 10.1016/j.cell.2013.03.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 19.Lee AH, Scapa EF, Cohen DE, Glimcher LH. 2008. Regulation of hepatic lipogenesis by the transcription factor XBP1. Science 320:1492–1496. doi: 10.1126/science.1158042. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 20.Wessels A, van den Hoff MJ, Adamo RF, Phelps AL, Lockhart MM, Sauls K, Briggs LE, Norris RA, van Wijk B, Perez-Pomares JM, Dettman RW, Burch JB. 2012. Epicardially derived fibroblasts preferentially contribute to the parietal leaflets of the atrioventricular valves in the murine heart. Dev Biol 366:111–124. doi: 10.1016/j.ydbio.2012.04.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 21.Chen F, Capecchi MR. 1999. Paralogous mouse Hox genes, Hoxa9, Hoxb9, and Hoxd9, function together to control development of the mammary gland in response to pregnancy. Proc Natl Acad Sci U S A 96:541–546. doi: 10.1073/pnas.96.2.541. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 22.Wen HC, Avivar-Valderas A, Sosa MS, Girnius N, Farias EF, Davis RJ, Aguirre-Ghiso JA. 2011. p38alpha signaling induces anoikis and lumen formation during mammary morphogenesis. Sci Signal 4:ra34. doi: 10.1126/scisignal.2001684. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 23.Smalley MJ. 2010. Isolation, culture and analysis of mouse mammary epithelial cells. Methods Mol Biol 633:139–170. doi: 10.1007/978-1-59745-019-5_11. [DOI] [PubMed] [Google Scholar]
 - 24.Dunphy KA, Tao L, Jerry DJ. 2010. Mammary epithelial transplant procedure. J Vis Exp 2010:1849. doi: 10.3791/1849. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 25.Anderson SM, Rudolph MC, McManaman JL, Neville MC. 2007. Key stages in mammary gland development. Secretory activation in the mammary gland: it's not just about milk protein synthesis! Breast Cancer Res 9:204. doi: 10.1186/bcr1653. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 26.Boutinaud M, Guinard-Flamenta J, Jammes H. 2004. The number and activity of mammary epithelial cells, determining factors for milk production. Reprod Nutr Dev 44:499–508. doi: 10.1051/rnd:2004054. [DOI] [PubMed] [Google Scholar]
 - 27.Reimold AM, Etkin A, Clauss I, Perkins A, Friend DS, Zhang J, Horton HF, Scott A, Orkin SH, Byrne MC, Grusby MJ, Glimcher LH. 2000. An essential role in liver development for transcription factor XBP-1. Genes Dev 14:152–157. doi: 10.1101/gad.14.2.152. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 28.Sequeira SJ, Ranganathan AC, Adam AP, Iglesias BV, Farias EF, Aguirre-Ghiso JA. 2007. Inhibition of proliferation by PERK regulates mammary acinar morphogenesis and tumor formation. PLoS One 2:e615. doi: 10.1371/journal.pone.0000615. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 29.Wang XZ, Ron D. 1996. Stress-induced phosphorylation and activation of the transcription factor CHOP (GADD153) by p38 MAP kinase. Science 272:1347–1349. [DOI] [PubMed] [Google Scholar]
 - 30.Kolb AF, Huber RC, Lillico SG, Carlisle A, Robinson CJ, Neil C, Petrie L, Sorensen DB, Olsson IA, Whitelaw CB. 2011. Milk lacking alpha-casein leads to permanent reduction in body size in mice. PLoS One 6:e21775. doi: 10.1371/journal.pone.0021775. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 31.Howarth DL, Lindtner C, Vacaru AM, Sachidanandam R, Tsedensodnom O, Vasilkova T, Buettner C, Sadler KC. 2014. Activating transcription factor 6 is necessary and sufficient for alcoholic fatty liver disease in zebrafish. PLoS Genet 10:e1004335. doi: 10.1371/journal.pgen.1004335. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 32.Basseri S, Austin RC. 2012. Endoplasmic reticulum stress and lipid metabolism: mechanisms and therapeutic potential. Biochem Res Int 2012:841362. doi: 10.1155/2012/841362. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 33.Volmer R, Ron D. 2014. Lipid-dependent regulation of the unfolded protein response. Curr Opin Cell Biol 33C:67–73. doi: 10.1016/j.ceb.2014.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 34.Ali S, Ali S. 1998. Prolactin receptor regulates Stat5 tyrosine phosphorylation and nuclear translocation by two separate pathways. J Biol Chem 273:7709–7716. [DOI] [PubMed] [Google Scholar]
 - 35.Long W, Wagner KU, Lloyd KC, Binart N, Shillingford JM, Hennighausen L, Jones FE. 2003. Impaired differentiation and lactational failure of Erbb4-deficient mammary glands identify ERBB4 as an obligate mediator of STAT5. Development 130:5257–5268. doi: 10.1242/dev.00715. [DOI] [PubMed] [Google Scholar]
 - 36.Cui Y, Riedlinger G, Miyoshi K, Tang W, Li C, Deng CX, Robinson GW, Hennighausen L. 2004. Inactivation of Stat5 in mouse mammary epithelium during pregnancy reveals distinct functions in cell proliferation, survival, and differentiation. Mol Cell Biol 24:8037–8047. doi: 10.1128/MCB.24.18.8037-8047.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 37.Hennighausen L, Robinson GW. 2008. Interpretation of cytokine signaling through the transcription factors STAT5A and STAT5B. Genes Dev 22:711–721. doi: 10.1101/gad.1643908. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 38.Ormandy CJ, Binart N, Kelly PA. 1997. Mammary gland development in prolactin receptor knockout mice. J Mammary Gland Biol Neoplasia 2:355–364. [DOI] [PubMed] [Google Scholar]
 - 39.Ormandy CJ, Camus A, Barra J, Damotte D, Lucas B, Buteau H, Edery M, Brousse N, Babinet C, Binart N, Kelly PA. 1997. Null mutation of the prolactin receptor gene produces multiple reproductive defects in the mouse. Genes Dev 11:167–178. [DOI] [PubMed] [Google Scholar]
 - 40.Shillingford JM, Miyoshi K, Robinson GW, Grimm SL, Rosen JM, Neubauer H, Pfeffer K, Hennighausen L. 2002. Jak2 is an essential tyrosine kinase involved in pregnancy-mediated development of mammary secretory epithelium. Mol Endocrinol 16:563–570. doi: 10.1210/mend.16.3.0805. [DOI] [PubMed] [Google Scholar]
 - 41.Jones FE, Welte T, Fu XY, Stern DF. 1999. ErbB4 signaling in the mammary gland is required for lobuloalveolar development and Stat5 activation during lactation. J Cell Biol 147:77–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 42.Tidcombe H, Jackson-Fisher A, Mathers K, Stern DF, Gassmann M, Golding JP. 2003. Neural and mammary gland defects in ErbB4 knockout mice genetically rescued from embryonic lethality. Proc Natl Acad Sci U S A 100:8281–8286. doi: 10.1073/pnas.1436402100. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 43.Liu X, Robinson GW, Wagner KU, Garrett L, Wynshaw-Boris A, Hennighausen L. 1997. Stat5a is mandatory for adult mammary gland development and lactogenesis. Genes Dev 11:179–186. [DOI] [PubMed] [Google Scholar]
 - 44.Miyoshi K, Shillingford JM, Smith GH, Grimm SL, Wagner KU, Oka T, Rosen JM, Robinson GW, Hennighausen L. 2001. Signal transducer and activator of transcription (Stat) 5 controls the proliferation and differentiation of mammary alveolar epithelium. J Cell Biol 155:531–542. doi: 10.1083/jcb.200107065. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 45.Wiseman BS, Werb Z. 2002. Stromal effects on mammary gland development and breast cancer. Science 296:1046–1049. doi: 10.1126/science.1067431. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 46.Tanjore H, Lawson WE, Blackwell TS. 2013. Endoplasmic reticulum stress as a pro-fibrotic stimulus. Biochim Biophys Acta 1832:940–947. doi: 10.1016/j.bbadis.2012.11.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 47.Vacaru AM, Di Narzo AF, Howarth DL, Tsedensodnom O, Imrie D, Cinaroglu A, Amin S, Hao K, Sadler KC. 2014. Molecularly defined unfolded protein response subclasses have distinct correlations with fatty liver disease in zebrafish. Dis Model Mech 7:823–835. doi: 10.1242/dmm.014472. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 48.Naylor MJ, Lockefeer JA, Horseman ND, Ormandy CJ. 2003. Prolactin regulates mammary epithelial cell proliferation via autocrine/paracrine mechanism. Endocrine 20:111–114. doi: 10.1385/ENDO:20:1-2:111. [DOI] [PubMed] [Google Scholar]
 - 49.Yang L, Carlson SG, McBurney D, Horton WE Jr. 2005. Multiple signals induce endoplasmic reticulum stress in both primary and immortalized chondrocytes resulting in loss of differentiation, impaired cell growth, and apoptosis. J Biol Chem 280:31156–31165. doi: 10.1074/jbc.M501069200. [DOI] [PubMed] [Google Scholar]
 - 50.Zu K, Bihani T, Lin A, Park YM, Mori K, Ip C. 2006. Enhanced selenium effect on growth arrest by BiP/GRP78 knockdown in p53-null human prostate cancer cells. Oncogene 25:546–554. doi: 10.1038/sj.onc.1209071. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 51.Bobrovnikova-Marjon E, Grigoriadou C, Pytel D, Zhang F, Ye J, Koumenis C, Cavener D, Diehl JA. 2010. PERK promotes cancer cell proliferation and tumor growth by limiting oxidative DNA damage. Oncogene 29:3881–3895. doi: 10.1038/onc.2010.153. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 52.Xu T, Yang L, Yan C, Wang X, Huang P, Zhao F, Zhao L, Zhang M, Jia W, Wang X, Liu Y. 2014. The IRE1alpha-XBP1 pathway regulates metabolic stress-induced compensatory proliferation of pancreatic beta-cells. Cell Res 24:1137–1140. doi: 10.1038/cr.2014.55. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 53.Tabas I, Ron D. 2011. Integrating the mechanisms of apoptosis induced by endoplasmic reticulum stress. Nat Cell Biol 13:184–190. doi: 10.1038/ncb0311-184. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 54.Schroder M, Kaufman RJ. 2005. The mammalian unfolded protein response. Annu Rev Biochem 74:739–789. doi: 10.1146/annurev.biochem.73.011303.074134. [DOI] [PubMed] [Google Scholar]
 - 55.Zeng L, Zampetaki A, Margariti A, Pepe AE, Alam S, Martin D, Xiao Q, Wang W, Jin ZG, Cockerill G, Mori K, Li YS, Hu Y, Chien S, Xu Q. 2009. Sustained activation of XBP1 splicing leads to endothelial apoptosis and atherosclerosis development in response to disturbed flow. Proc Natl Acad Sci U S A 106:8326–8331. doi: 10.1073/pnas.0903197106. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 56.Chen X, Iliopoulos D, Zhang Q, Tang Q, Greenblatt MB, Hatziapostolou M, Lim E, Tam WL, Ni M, Chen Y, Mai J, Shen H, Hu DZ, Adoro S, Hu B, Song M, Tan C, Landis MD, Ferrari M, Shin SJ, Brown M, Chang JC, Liu XS, Glimcher LH. 2014. XBP1 promotes triple-negative breast cancer by controlling the HIF1alpha pathway. Nature 508:103–107. doi: 10.1038/nature13119. [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.









