Pre–B cell leukemia homeobox 1 (Pbx1)-d is a dominant-negative splice isoform of the gene Pbx1 that corresponds to the NZM2410 lupus susceptibility locus Sle1a1. Pbx1 is required to maintain stem cell self-renewal, including that of mesenchymal stem cells (MSCs). MSCs have immunosuppressive functions that require stem cell maintenance. We tested the hypothesis that the expression of Pbx1-d favors MSC differentiation and impairs their immunosuppressive functions. We demonstrate that Sle1a1 MSCs express high levels of Pbx1-d as compared with congenic C57BL/6J (B6) MSCs. Sle1a1 MSCs grew faster and differentiated significantly more rapidly into osteoblasts than did B6 MSCs. This corresponded to a significant decrease in the expression of genes associated with stemness and an increase in the expression of genes associated with differentiation. Additionally, Sle1a1 MSCs express a gene expression profile associated with an enhanced innate immunity and inflammation. Suppression of Ig production from TLR-activated B6 B cells and IL-2 secretion from activated B6 CD4+ T cells was significantly impaired in Sle1a1 MSCs as compared with B6 MSCs. B6.Sle1a1 MSCs showed intermediate activity in suppressing lupus immunophenotypes in three different mouse models. Taken together, these data suggest that the expression of the lupus susceptibility allele Pbx1-d isoform impairs MSC functions, which may contribute to lupus pathogenesis both through a defective immunosuppression and the promotion of a proinflammatory environment.

This article is featured in In This Issue, p.1

Systemic lupus erythematosus (SLE) is a complex disease with defects in multiple cell types that are mediated through a large number of susceptibility genes whose identity is starting to be revealed (1). Using the NZM2410 lupus-prone mouse, we have identified a number of susceptibility genes and have produced a series of congenic strains that represent a powerful tool to characterize the function of the lupus susceptibility genes singly and in combination (2). This has led to the production of the B6.Sle1.Sle2.Sle3 (TC) strain that coexpresses on a C57BL/6J (B6) background the three Sle susceptibility loci that are necessary and sufficient to develop clinical lupus (3). Among the NZM2410 susceptibility genes, we have identified the pre–B cell leukemia homeobox 1 (Pbx1) gene as the gene corresponding to Sle1a1 (4). Sle1a1 is a contributor to the Sle1 global locus, which controls the loss of tolerance to chromatin (57), and is necessary for disease initiation (8, 9). NZM2410-derived B6.Sle1a1 (Sle1a1) congenic mice as well as TC mice, which carry Sle1a1, present an elevated number of activated autoreactive CD4+ T cell that provide help to B cells producing anti-chromatin IgG (6, 7).

Pbx1, the only gene located in the Sle1a1 interval, is a well-characterized member of the TALE family of homeodomain proteins that functions as a transcriptional regulator through the modulation of the DNA-binding function of Hox proteins (10). The NZM2410 allele is a splice isoform (Pbx1-d) in which DNA-binding exon 6 and Hox-binding exon 7 are missing and thus induces a dominant-negative function (11). Importantly, the Pbx1 amino acid sequence is identical between mice and humans. PBX1-d was found more frequently in SLE patients than in healthy controls, and its expression corresponded to a decreased naive/central memory CD4+ T cell ratio, which is characteristic of SLE T cells (4). Prior to this newly discovered role in regulating T cell functions, full-length Pbx1-b was known to be necessary to block lineage-specific differentiation and maintains self-renewal of hematopoietic stem cells (12) as well as mesenchymal stem cells (MSCs) (13). We have shown using an MSC line that the overexpression of Pbx1-d was functionally equivalent to knocking down Pbx1-b, resulting in an accelerated differentiation into osteoblasts (11).

MSCs serve as a reservoir for tissue regeneration through their multipotent differentiation into chondrocytes, osteoblasts, or adipocytes (14). Additionally, MSCs play a role in tissue and immune homeostasis through regulation of apoptosis and immune modulation (15). MSCs suppress T cell proliferation and dendritic cell maturation, and either promote or suppress B cell proliferation and Ab production. Several cases of successful treatment of refractory SLE with allogeneic MSCs have been reported, but treatment of two patients with autologous MSCs was not successful (16, 17). Additionally, MSCs from lupus have defective functions, with an impaired differentiation into osteoblasts (18), enhanced senescence and apoptosis, as well as impaired self-renewal (19, 20), providing potential mechanisms for the observed failure of autologous MSC therapies. One study, however, reported that MSCs from autoimmune patients presented immunosuppressive properties similar to those of healthy controls (21). Mixed results have also been obtained with mouse models of lupus (17). MRL/lpr mice have been successfully treated with both allogeneic and syngeneic MSCs in several studies (2225). In the (NZB × NZW)F1 (BWF1) strain, which has the same Pbx1 allele as Sle1a1/NZM2410, allogeneic bone marrow (BM)-derived MSC treatments accelerated (26) or attenuated (27) disease, whereas human adipose–derived MSCs (28), or young syngeneic MSCs (24), induced complete reversal. However, MSCs derived from diseased BWF1 mice were defective, suggesting that disease-induced factors, such as inflammation and possibility age-related intrinsic defects, affect their functions (24).

Based on this body of work, we hypothesized that Pbx1-d expression favoring lineage-specific differentiation should impair MSC immune suppression, which is dependent on the maintenance of pluripotential status (29). We show that the major Pbx1 isoform in Sle1a1 MSCs is Pbx1-d, and that Sle1a1 MSCs display a transcriptional signature as well as surface markers that are consistent with enhanced lineage differentiation. In vitro, Sle1a1 MSCs differentiated into osteoblasts significantly faster than did congenic B6 MSCs, and they showed an impaired suppression of TLR-induced Ab secretion by B cells, as well as IL-2 secretion in stimulated CD4+ T cells. In vivo, Sle1a1 MSCs showed an intermediate immunodulatory capacity in three different models of lupus as compared with B6 MSCs. Overall, our results showed that the lupus susceptibility allele Pbx1-d impairs MSC functions, and they suggest that this may not only contribute to pathogenesis through a direct effect in immune cell functions but also impair allogeneic MSC immunotherapeutic potential.

The B6.Sle1a1 (Sle1a1) and TC congenic strains have been previously described (3, 4). B6, BWF1, and MRL/lpr mice were obtained from The Jackson laboratory (Bar Harbor, ME). Sle1a1, TC, and B6 mice were bred at the University of Florida. Only females were used in this study. The experiments were conducted according to protocols approved by the University of Florida Institutional Animal Care and Use Committee.

MSCs were obtained from pooled BM of four 2-mo-old Sle1a1 or B6 mice. Specifically, whole BM was isolated from the femur and tibia, passed through a 70-μm filter (Fisher Scientific), and plated at a 1 × 105/ml concentration in MesenCult proliferation media (StemCell Technologies). Cells were incubated at 37°C with 5% CO2 until 80% confluent. Cells were split 1:3 until they reached passage 9. All experiments were performed using cells at passages 2 or 3. The number of days of culture prior to reaching 80% confluence and thus passage was recorded to evaluate growth rate of MSCs. Additionally, the MSC phenotype was characterized by flow cytometry following trypsinization.

Trypsinized MSCs were suspended in osteogenic differentiation medium (MODM, ScienCell) and plated at a concentration of 5 × 104/ml. In some experiments, osteoblastic differentiation was visualized using Alizarin red. Specifically at days 0–7 of culture, cells were fixed in 4% paraformaldehyde, washed three times with distilled H2O, and stained with 2% Alizarin red S stain solution (ScienCell) for 30 min, after which the cells were washed five times with distilled H2O and placed in 1 ml distilled H2O to keep hydrated. Data are reported as the percentage of the culture that is Alizarin red+. In additional experiments protein was harvested at days 0 and 6 of culture, and Bmp4 expression was assessed by Western blot. Cell lysates prepared from B6 and Sle1a1 MSC passage 3 in Nonidet P-40 lysis buffer were probed with goat polyclonal BMP-4 Ab (A-20) (sc-6267, Santa Cruz Biotechnology). Mouse monoclonal anti–β-actin Ab (clone AC-74, Sigma-Aldrich) was used as control, and secondary Abs were goat anti-rabbit IgG-HRP (sc-2004, Santa Cruz Biotechnology) and goat anti-mouse IgG (H+L)-HRP (1031-05, SouthernBiotech). Protein transfer was carried out on Trans-Blot Turbo transfer system (Bio-Rad), and the membranes were developed by Immobilon Western chemiluminescent HRP substrate (WBKLS0500, EMD Millipore).

MSCs were stained with Ab against CD90.2 (53-2.1), CD73 (ebioTY/11.8), CD105 (MJ7/18), CD14 (Sa2-8), CD34 (RAM34), CD45 (30-F11), and MHC class II (M5/114.15.2). To evaluate immune cell subsets following MSC therapy, single-cell suspensions were prepared from spleens by lysing RBCs with 0.83% NH4Cl, blocked with saturating amounts of anti-CD16/CD32 (2.4G2), and then stained with Abs against CD4 (RM4-5), CD44 (IM7), CD62L (MEL-14), CD19 (1D3), and IgM (IgH6). The Abs were all purchased from BD Pharmingen or eBioscience. Acquisition was performed on either a LSRFortessa or FACSCalibur cytometer (BD Biosciences) with at least 100,000 events per sample collected. Analysis was performed using FCS Express (De Novo Software). Live cells were gated based on forward and side scatter characteristics.

RNA was isolated using a Qiagen kit from MSC lines at passage 2. Pbx1 isoform expression was quantified with the primers listed in Supplemental Table I and SYBR Green incorporation. Additionally, Pbx1-d was quantified using a custom-made TaqMan probe (Supplemental Table I). Pbx1 protein expression was analyzed by Western blot with rabbit polyclonal Pbx1 Ab (PA5-17223; Thermo Scientific) and goat anti-rabbit IgG-HRP (sc-2004, Santa Cruz Biotechnology). Mouse monoclonal anti–GAPDH-peroxidase (clone GAPDH-71.1, Sigma-Aldrich) was used as control. HEK293 cells were grown to 70–80% confluence and transfected with plasmids (pTYF-Pbx1d and pCDNA3-Pbx1b) and Lipofectamine (Invitrogen) according to the manufacturer’s protocol. These cells were used as controls for RNA and protein expression. A MSC PCR array (PAMM-0827, Qiagen) was used, according to the manufacturer’s instructions, on RNA purified from three MSC lines each from Sle1a1 and B6 mice. Quantitative RT-PCR (qRT-PCR) was performed for validation with the primers listed in Supplemental Table I, by incorporation of SYBR Green using a LightCycler thermocycler (Roche Applied Science). Expression of each gene tested with qRT-PCR was normalized against Gapdh for each sample and then expressed as the fold ratio to the average for B6 samples. The level of IL-6, IL-10, and IL-33 produced by MSCs was measured by ELISA (IL-6 and IL-10 kits from BD Biosciences, IL-33 kit from eBioscience) supernatants diluted 1:10.

RNA was isolated from three B6 and three Sle1a1 MSC lines at passage 2 with a Qiagen kit. Libraries were constructed with a TruSeq RNA sample preparation kit (Illumina) and sequencing was performed on the HiSequation 2000 platform (Illumina) by the University of Florida Interdisciplinary Center for Biotechnology Research Genomics core. Raw sequencing data were processed with the cutadapt program and scripts developed in-house at the Interdisciplinary Center for Biotechnology Research of the University of Florida to trim off sequencing adaptors, primers, and low-quality bases with respect to a quality value cutoff of 20 (Phred-like score). Accordingly, reads that were shorter than 40 bases or that contained >10 bases with quality <20 were discarded. After quality control assessment, sequencing reads were mapped independently for each sample to the GRCm38 reference genome using TopHat (2.0.1) with a maximum of three mismatches for each read. The mapping results were processed with in-house scripts to remove potential PCR duplicates. The uniquely mapped reads were selected for quantifying gene expression. The mapped reads for each individual gene were counted using the HTSeq package and scripts developed in-house. The counting results were used to quantify gene expression by the DEB application (30). Genes with a significant differential expression were selected using an adjusted p value of <0.05 and expressed as log2-fold change of Sle1a1 over B6 MSC expression. For robustness, we also repeated the differential expression analysis using the Cufflinks/Cuffdiff pipeline (http://cufflinks.cbcb.umd.edu/), comparing expression between the two strains. This identified 1471 genes showing differential expression at the 5% significance level, of which 1004 were also identified by the previous analysis. We analyzed this list of 1004 genes using the ToppFun program, part of the ToppGene suite (https://toppgene.cchmc.org/), to identify functional categories showing enrichment for the genes in our list. Note that the RNA-Seq data set can be accessed at http://www.ncbi.nlm.nih.gov/bioproject?LinkName=sra_bioproject&from_uid=1073997 (accession no. PRJNA264078).

MSC function was characterized by suppression assays. Following trypsinization, indicated numbers of MSCs were primed with 5 ng/ml IFN-γ (Sigma-Aldrich) overnight, after which primed MSCs were cocultured with either 6 × 104 B6 B cells or 6 × 104 B6 CD4+ T cells purified by negative selection with magnetic beads (Miltenyi Biotec). B cells were stimulated with 1 μg/ml LPS or CpG (Invivogen). T cells were stimulated with 6 × 104 anti-CD3/CD28 beads (Life Technologies). Cultures were stimulated for 3 d, after which supernatants were probed for either total Ig (B cells) (eBioscience) or IL-2 (T cells) (BD Biosciences) by ELISA. A standard curve and five parameter logistics were used to determine concentrations.

We have adapted a previously published protocol (24) to compare the immunosuppressive capacity of Sle1a1 and B6 MSCs to reverse lupus in three different mouse models. Cohorts of age-matched anti-dsDNA IgG+ mice were injected i.v. with 106 BM-derived B6 or Sle1a1 MSCs, or left untreated. We treated TC mice at 25–31 wk of age, BWF1 mice at 21 wk old, and MRL/lpr mice at 8 wk old. Serum was collected before treatment and every 2 wk after treatment for autoantibody measurements. Proteinuria was measured weekly with Albustix strips (Bayer) on a 0–4 scale corresponding to <100, 300, 1000, 3000, and >3000 μg/ml of protein. All mice were sacrificed 8 wk after injection, except for two mice that presented with terminal disease and were sacrificed early in the treatment. Immune cell subset distribution and activation were analyzed by flow cytometry of splenocytes. Renal pathology was scored from H&E and periodic acid–Schiff stains as previously described (31). Briefly, glomerulonephritis (GN) was scored on a semiquantitative 0–4 scale, and qualified as mesangial or proliferative. Additionally, the samples within each strain were ranked according to increased severity by a pathologist (B.C.) who did not have knowledge of the treatment status. The glomerular deposition of C3 and IgG2a immune complexes was performed on frozen kidney sections and scored as previously described (31). Glomerular size and the extent of C3 deposition were measured from anti-C3–stained sections averaging four to six glomeruli per sample using the Metamorph 7.5 image analysis software (Molecular Devices, Downingtown, PA). Microalbuminuria was measured on terminal urine samples with a kit according to the manufacturer’s instructions (Kamiya Biomedical).

Anti-dsDNA and anti-chromatin IgG ELISA assays were carried out as previously described (3). Test sera were incubated in duplicate at 1:100 dilutions. Relative units were standardized using a pool of TC sera, arbitrarily setting reactivity at a 1:100 dilution to 100 U. Antinuclear autoantibodies (ANAs) were measured from sera diluted 1:40 applied to Hep-2 cell slides (INOVA Diagnositics) followed by a FITC-conjugated anti-mouse IgG (SouthernBiotech) diluted 1:50. ANA staining was quantified with the Metamorph 7.5 image analysis software as a percentage of the image scoring above a set intensity. For each strain of recipient mice, ANA staining was performed for as a single batch for all samples. The number of Hep-2 cells was similar between slides within each batch.

Statistical analyses were performed using GraphPad Prism 6.0. Unless specified, the graphs show means and SEM. Comparisons were performed with either two-tailed t tests or Mann–Whitney U tests, depending on the normality of the data distribution. Multiple comparison corrections were used when appropriate. Time-course graphs were compared with two-way ANOVA.

We compared Pbx1 isoform expression in MSCs by semiquantitative RT-PCR using the set of primers that amplifies all isoforms (Fig. 1A) that we previously used for CD4+ T cells (4). The main isoform expressed by B6 MSCs was Pbx1-b, which has been shown by others to be the major isoform expressed in normal MSCs (13), whereas the main isoform expressed by Sle1a1 MSCs was Pbx1-d (Fig. 1B). To quantify this differential expression, we designed isoform-specific qRT-PCR primers, as well as a TaqMan probe for Pbx1-d (Fig. 1A, Supplemental Table I). There was no difference of expression for total Pbx1, Pbx1-a, or Pbx1-b between the two strains, but Sle1a1 MSCs expressed significantly more Pbx1-d than did B6 MSCs (Fig. 1C). This finding was confirmed using a TaqMan probe with six other Pbx1-d–specific primer pairs (Fig. 1D and data not shown). Moreover, the relative ratios of Pbx1-d over total Pbx1, Pbx1-a, or Pbx1-b expression were significantly higher in Sle1a1 than in B6 MSCs (Fig. 1E). Although Western blot analysis of B6 and Sle1a1 MSCs showed a similar amount of total Pbx1 in the two strains, confirming the qRT-PCR results, the isoform results could not be validated at the protein level with currently available Pbx1 Abs, as they bind equally to both isoforms (Fig. 1F). The predicted size of each isoform is 3 aa apart (Pbx1-b, 38 kDa; Pbx1-d, 37 kDa) and thus cannot be distinguished by electrophoresis as shown by the transfection of HEK293 cells with expression plasmids (Fig. 1F).

FIGURE 1.

Pbx1-d is expressed at a higher level in Sle1a1 MSCs than in B6 MSCs. (A) Map of the three Pbx1 isoforms and location of the primers, indicated by arrows, used in this study. In addition to the three isoform-specific primer pairs, we used a primer pair in exons 5 and 8 that amplifies each of the three isoforms, as well as a pair in exons 2 and 3 that amplifies total Pbx1. Pbx1-d was amplified with a forward primer that spans the exon 5–8 junction with a reverse primer in exon 9. The star indicates the position of the Pbx1-d TaqMan probe in exon 8. (B) Pbx1 expression in B6 and Sle1a1 MSCs, assessed with the primers that amplify all three isoforms. Amplification of the specific isoforms from HEK293 cells transfected with cloned cDNA is shown on the left as reference. (C) Expression of total Pbx-1, Pbx1-a, Pbx1-b, and Pbx1-d as measured by SYBR Green incorporation. (D) Pbx1-d expression measured with a TaqMan probe. Results were obtained from three lines per strain, first normalized to Gapdh expression and then expressed relative to the B6 average. (E) Pbx1-d relative expression to total Pbx1, Pbx1-a, or Pbx1-b. (F) Pbx1 protein expression in B6 and Sle1a1 MSCs relative to Gapdh. The blot on the left shows Pbx1 expression measured in HEK293 cells transfected with either Pbx1-b or two different Pbx1-d cDNAs. Results are representative of three experiments. *p < 0.05.

FIGURE 1.

Pbx1-d is expressed at a higher level in Sle1a1 MSCs than in B6 MSCs. (A) Map of the three Pbx1 isoforms and location of the primers, indicated by arrows, used in this study. In addition to the three isoform-specific primer pairs, we used a primer pair in exons 5 and 8 that amplifies each of the three isoforms, as well as a pair in exons 2 and 3 that amplifies total Pbx1. Pbx1-d was amplified with a forward primer that spans the exon 5–8 junction with a reverse primer in exon 9. The star indicates the position of the Pbx1-d TaqMan probe in exon 8. (B) Pbx1 expression in B6 and Sle1a1 MSCs, assessed with the primers that amplify all three isoforms. Amplification of the specific isoforms from HEK293 cells transfected with cloned cDNA is shown on the left as reference. (C) Expression of total Pbx-1, Pbx1-a, Pbx1-b, and Pbx1-d as measured by SYBR Green incorporation. (D) Pbx1-d expression measured with a TaqMan probe. Results were obtained from three lines per strain, first normalized to Gapdh expression and then expressed relative to the B6 average. (E) Pbx1-d relative expression to total Pbx1, Pbx1-a, or Pbx1-b. (F) Pbx1 protein expression in B6 and Sle1a1 MSCs relative to Gapdh. The blot on the left shows Pbx1 expression measured in HEK293 cells transfected with either Pbx1-b or two different Pbx1-d cDNAs. Results are representative of three experiments. *p < 0.05.

Close modal

We examined Pbx1 expression using an RNA-Seq data set that compared Sle1a1 and B6 MSC RNA expression (Supplemental Table II). The total number of Pbx1 reads was significantly lower in Sle1a1 than in B6 MSCs. The analysis of the individual Pbx1 isoforms detected four isoforms above background, with Pbx1-a being the most abundant and the only one showing a significant difference between strains, with the Sle1a1 MSCs expressing less (Supplemental Table II). Taken together, these data support the fact that Pbx1 transactivates itself, and that Pbx1-d has a dominant-negative function. As shown above, we and others have shown by RT-PCR that Pbx1-b is the most abundant isoform in normal MSCs. Pbx1-d was not found in B6 MSCs, and it was not formally detected above background in Sle1a1 MSCs, which prevented statistical analyses. Isoform detection through RNA-Seq is quite difficult, especially when a gene has several isoforms, and when expression is low, as is this case for Pbx1. We therefore think that our RT-PCR is more reliable, and thus we have demonstrated that Pbx1-d is the major Pbx1 isoform expressed in Sle1a1 MSCs.

BM-derived cultures from both strains resulted in similar frequency of CD90+CD73+ cells; however, the level of expression of surface markers was significantly different (Fig. 2A). Specifically, Sle1a1 MSCs expressed significantly lower levels of markers characteristic of the pluripotent capacity or “stemness” of these cells, such as CD90 and CD105 (Fig. 2B), while expressing higher levels of markers found on differentiated leukocytes, such as MCH class II, CD34, and CD45 (Fig. 2A, 2C). Additionally, Sle1a1 MSCs expanded at a significantly higher rate (Fig. 2D) and differentiated more rapidly into osteoblasts upon induction than did B6 MSCs (Fig. 2E). To this end, protein levels of a marker of osteoblast differentiation, Bmp4, were significantly higher in Sle1a1 than in B6 MSCs and increased to a much higher level in Sle1a1 MSCs upon osteogenic induction (Fig. 2F). Overall, these results demonstrate that Pbx1-d expression in MSCs is associated with a more differentiated and less stemness phenotype.

FIGURE 2.

Sle1a1 MSCs differentiate faster than do B6 MSCs. (A) Representative bright field microscopy and flow cytometry comparison of MSCs between the two strains. Original magnification ×60. The number in the upper right corner indicates the percentage of CD90+CD73+ cells. The histogram overlays on the right show the expression of three surface makers associated with MSC stemness (CD105 [dotted line], CD90, and CD73) and three markers associated with leukocyte differentiation (MHC class II [blue], CD34 [red], and CD45 [green]). (B and C) Mean fluorescence intensity (MFI) quantitation of the stemness (B) and leukocyte differentiation (C) markers shown in (A). Statistical significance is shown for t tests between strains for each marker (D). Growth rate measured as the number of days necessary to reach each passage. (E) Osteoblast differentiation time course measured as the percentage of Alizarin red+ cells. For (D) and (E), statistical significance compared the two strains with two-way ANNOVA. (F) Bmp4 protein expression in Sle1a1 and B6 MSCs before (day 0) and 6 d after osteogenic differentiation. β-Actin expression is shown as internal control. ***p < 0.001.

FIGURE 2.

Sle1a1 MSCs differentiate faster than do B6 MSCs. (A) Representative bright field microscopy and flow cytometry comparison of MSCs between the two strains. Original magnification ×60. The number in the upper right corner indicates the percentage of CD90+CD73+ cells. The histogram overlays on the right show the expression of three surface makers associated with MSC stemness (CD105 [dotted line], CD90, and CD73) and three markers associated with leukocyte differentiation (MHC class II [blue], CD34 [red], and CD45 [green]). (B and C) Mean fluorescence intensity (MFI) quantitation of the stemness (B) and leukocyte differentiation (C) markers shown in (A). Statistical significance is shown for t tests between strains for each marker (D). Growth rate measured as the number of days necessary to reach each passage. (E) Osteoblast differentiation time course measured as the percentage of Alizarin red+ cells. For (D) and (E), statistical significance compared the two strains with two-way ANNOVA. (F) Bmp4 protein expression in Sle1a1 and B6 MSCs before (day 0) and 6 d after osteogenic differentiation. β-Actin expression is shown as internal control. ***p < 0.001.

Close modal

Pbx1 is a transcription factor, and the expression of its dominant-negative allele is predicted to alter gene expression. To test this hypothesis, we used a PCR array that contains MSC-related genes to compare the gene profile of Sle1a1 and B6 MSCs. In accord with our phenotypic data, most genes that were underexpressed in Sle1a1 MSCs, such as Thy1 or Pdgfr, were genes associated with a stem cell phenotype, whereas the genes that were overexpressed in Sle1a1 MSCs were associated with lineage-specific differentiation, such as Gdf7 for chondrogenesis or Bmp4 and Fgf10 for osteogenesis (Fig. 3A). Most of these results were validated by RNA-Seq (Fig. 3B) and qRT-PCR (Fig. 3C) analyses of an independent set of MSC cultures. Additionally, we have validated the increased expression of Bmp4 protein by Sle1a1 MSCs (Fig. 2F).

FIGURE 3.

Sle1a1 MSCs express a different gene signature than do B6 MSCs. (A) Expression of MSC-related genes in Sle1a1 MSCs relative to B6 MSCs measured with a PCR array. (B) Expression of the same genes as in (A) detected by RNA-Seq in a new set of MSC lines for each strain with a p value of <0.05. (C) qRT-PCR validation of the same genes as in (A) showing a significant difference between the two strains with a p value of <0.05. Open bars show MSC-specific genes, gray bars show MSC-associated genes, and black bars show genes associated with lineage-specific differentiation. The arrows indicate genes mentioned in the text (n = 3 lines/strain/experiment). Genes in the “GO: stem cell proliferation” (D) and “GO: cytokine production” (E) pathways that were differentially expressed between B6 and Sle1a1 MSCs are shown. The threshold for differential expression was set at −0.5 > log2(fold change) > 0.5 for (D) or −1 > log2(fold change) > 1 for (E), and p < 0.001 for both. (F) qRT-PCR validation of selected genes in the cytokine production pathway. Means ± SEM of n = 3/strain are shown. *p < 0.05 for one-tailed t tests; **p < 0.01, ***p < 0.001 for two-tailed t tests. (G) IL-10 and IL-6 production in MSC supernatant. Means ± SEM of n = 4/strain are shown. *p < 0.05 for two-tailed t tests.

FIGURE 3.

Sle1a1 MSCs express a different gene signature than do B6 MSCs. (A) Expression of MSC-related genes in Sle1a1 MSCs relative to B6 MSCs measured with a PCR array. (B) Expression of the same genes as in (A) detected by RNA-Seq in a new set of MSC lines for each strain with a p value of <0.05. (C) qRT-PCR validation of the same genes as in (A) showing a significant difference between the two strains with a p value of <0.05. Open bars show MSC-specific genes, gray bars show MSC-associated genes, and black bars show genes associated with lineage-specific differentiation. The arrows indicate genes mentioned in the text (n = 3 lines/strain/experiment). Genes in the “GO: stem cell proliferation” (D) and “GO: cytokine production” (E) pathways that were differentially expressed between B6 and Sle1a1 MSCs are shown. The threshold for differential expression was set at −0.5 > log2(fold change) > 0.5 for (D) or −1 > log2(fold change) > 1 for (E), and p < 0.001 for both. (F) qRT-PCR validation of selected genes in the cytokine production pathway. Means ± SEM of n = 3/strain are shown. *p < 0.05 for one-tailed t tests; **p < 0.01, ***p < 0.001 for two-tailed t tests. (G) IL-10 and IL-6 production in MSC supernatant. Means ± SEM of n = 4/strain are shown. *p < 0.05 for two-tailed t tests.

Close modal

We identified 12 biological process classes from gene ontology (GO) and three mouse phenotypes (MP) that are either related to stem cells or immune processes (Table I). In particular, the three MP pathways, that is, abnormal innate immunity, increased inflammatory response, and abnormal inflammatory response, showed very strong evidence of enrichment. Examination of the individual genes in the stem cell proliferation pathway (Fig. 3D) indicated, similar to the gene array and phenotypic analysis, a decreased expression in genes associated with multipotent lineage such as Tbx18, Bdnf, and Zeb31 and, alternatively, an increased expression of genes associated with osteoblast differentiation (Wnt10b, Bmp4), as well as suppression of Wnt signaling by Sfrp2, which leads to decreased self-renewal (32). Examination of the cytokine production pathway indicated an increased expression by Sle1a1 MSCs of cytokines found at higher levels in SLE patients such as IL-10 (33) and IL-33 (34), as well as a very high expression of Camk4, a protein kinase that is highly expressed in the T cells from SLE patients and that is required for IL-17 production (35) (Fig. 3E). The increased message expression of Camk4, Il33, and Il10 in Sle1a1 MSCs was confirmed by qRT-PCR (Fig. 3F). On the other end of the differentially expressed genes in the cytokine production pathway, we confirmed the decreased expression of Il1rl2 in Sle1a1 MSCs. The level of IL-10 in MSC supernatants was highly variable, and the difference between strains did not reach statistical significance (Fig. 3G). The amount of IL-33 in the same supernatants was below detection level (data not shown), suggesting that MSCs produce the newly discovered nuclear full-length form of the cytokine that activates the MyD88 pathway and induces Ig secretion (36). Finally, IL-6 production was significantly reduced in Sle1a1 MSC supernatants (Fig. 3G), although we did not detect a significant difference at the message level (data not shown). Overall, our results suggest that Pbx1-d expression in MSCs is associated with a unique gene expression profile, and they confirm a cellular programming geared toward lineage differentiation and commitment rather than a stem cell state.

Table I.
Stem cell and immune-related GO pathways differentially expressed between Sle1a1 and B6 MSCs as determined by RNA-Seq
IDNamep Valueq Value (Bonferroni)aq Value (B&H)aHit Counts in Query ListbHit Counts in Genomec
GO: 0072089 Stem cell proliferation 9.57 × 10−7 6.07 × 10−3 2.55 × 10−5 22 129 
GO: 0006954 Inflammatory response 9.79 × 10−13 6.21 × 10−9 1.35 × 10−10 73 566 
GO: 0002682 Regulation of immune system process 4.58 × 10−11 2.91 × 10−7 3.64 × 10−9 113 1146 
GO: 0006955 Immune response 6.84 × 10−11 4.34 × 10−7 4.99 × 10−9 127 1353 
GO: 0001816 Cytokine production 1.45 × 10−10 9.18 × 10−7 1.03 × 10−8 66 541 
GO: 0002684 Positive regulation of immune system process 6.80 × 10−9 4.32 × 10−5 3.22 × 10−7 74 699 
GO: 0002520 Immune system development 8.17 × 10−9 5.18 × 10−5 3.76 × 10−7 74 702 
MP: 0002419 Abnormal innate immunity 1.29 × 10−7 6.77 × 10−4 6.15 × 10−5 76 521 
GO: 0050776 Regulation of immune response 2.35 × 10−7 1.49 × 10−3 7.20 × 10−6 75 776 
GO: 0045321 Leukocyte activation 3.00 × 10−7 1.91 × 10−3 8.90 × 10−6 65 642 
GO: 0002764 Immune response–regulating signaling pathway 5.50 × 10−7 3.49 × 10−3 1.56 × 10−5 52 478 
GO: 0002757 Immune response–activating signal transduction 8.63 × 10−7 5.47 × 10−3 2.36 × 10−5 43 369 
MP: 0001846 Increased inflammatory response 1.08 × 10−6 5.65 × 10−3 3.53 × 10−4 103 816 
GO: 0002253 Activation of immune response 9.30 × 10−7 5.90 × 10−3 2.50 × 10−5 47 421 
MP: 0001845 Abnormal inflammatory response 1.31 × 10−6 6.88 × 10−3 4.05 × 10−4 118 974 
IDNamep Valueq Value (Bonferroni)aq Value (B&H)aHit Counts in Query ListbHit Counts in Genomec
GO: 0072089 Stem cell proliferation 9.57 × 10−7 6.07 × 10−3 2.55 × 10−5 22 129 
GO: 0006954 Inflammatory response 9.79 × 10−13 6.21 × 10−9 1.35 × 10−10 73 566 
GO: 0002682 Regulation of immune system process 4.58 × 10−11 2.91 × 10−7 3.64 × 10−9 113 1146 
GO: 0006955 Immune response 6.84 × 10−11 4.34 × 10−7 4.99 × 10−9 127 1353 
GO: 0001816 Cytokine production 1.45 × 10−10 9.18 × 10−7 1.03 × 10−8 66 541 
GO: 0002684 Positive regulation of immune system process 6.80 × 10−9 4.32 × 10−5 3.22 × 10−7 74 699 
GO: 0002520 Immune system development 8.17 × 10−9 5.18 × 10−5 3.76 × 10−7 74 702 
MP: 0002419 Abnormal innate immunity 1.29 × 10−7 6.77 × 10−4 6.15 × 10−5 76 521 
GO: 0050776 Regulation of immune response 2.35 × 10−7 1.49 × 10−3 7.20 × 10−6 75 776 
GO: 0045321 Leukocyte activation 3.00 × 10−7 1.91 × 10−3 8.90 × 10−6 65 642 
GO: 0002764 Immune response–regulating signaling pathway 5.50 × 10−7 3.49 × 10−3 1.56 × 10−5 52 478 
GO: 0002757 Immune response–activating signal transduction 8.63 × 10−7 5.47 × 10−3 2.36 × 10−5 43 369 
MP: 0001846 Increased inflammatory response 1.08 × 10−6 5.65 × 10−3 3.53 × 10−4 103 816 
GO: 0002253 Activation of immune response 9.30 × 10−7 5.90 × 10−3 2.50 × 10−5 47 421 
MP: 0001845 Abnormal inflammatory response 1.31 × 10−6 6.88 × 10−3 4.05 × 10−4 118 974 
a

False discovery rate q values were evaluated based on the Bonferroni correction and on the Benjamini–Hochberg (B&H) method.

b

Number of genes in the pathway that are differentiatlly expressed between the two strains.

c

Number of genes in the pathway.

MSCs inhibit lymphocyte functions in vitro, and this immunosuppressive capacity is enhanced in the presence of IFN-γ. We have evaluated the ability of Sle1a1 MSCs as compared with B6 MSCs to suppress Ig secretion from B6 B cells stimulated with either LPS or CpG (Fig. 4A–D), as well as IL-2 secretion from B6 CD4+ T cells stimulated with anti-CD3 and anti-CD28 (Fig. 4E, 4F). We confirmed that MSC immunosuppression of Ig production is greatly enhanced by IFN-γ (compare Fig. 4A and 4C to Fig. 4B and 4D). MSC suppression of IL-2 production by T cells was less sensitive to enhancement by exogenous IFN-γ (compare Fig. 4E to Fig. 4F). In each condition, however, Sle1a1 MSCs were less suppressive than were B6 MSCs. Specifically, in the presence of IFN-γ, B6 MSCs significantly suppressed Ig production induced by either LPS or CpG up to a 3:1 B cell/MSC ratio, whereas Sle1a1 MSCs were suppressive at only a 1:1 B cell/MSC ratio. Without IFN-γ, B6 MSCs significantly suppressed Ig production induced at a 1:1 B cell/MSC ratio, but no suppression was observed with the Sle1a1 MSCs (Fig. 4A–D). Similarly, B6 MSCs significantly suppressed IL-2 production up to a 3:1 CD4+ T cells/MSC ratio whereas Sle1a1 MSCs were immunosuppressive only at the 1:1 ratio, either in the presence or absence of IFN-γ (Fig. 4E, 4F). Overall, our results demonstrate that Pbx1-d expression in MSCs is associated with a decreased suppressive function.

FIGURE 4.

Sle1a1 MSCs have a lower immunosuppressive capacity than do B6 MSCs in vitro. (AD) B6 B cells were cocultured with increasing ratios (10:1 to 0:1 B cells/MSCs) of B6 (filled bars) or Sle1a1 (open bars) MSCs and stimulated with LPS (A and B) or CpG (C and D) in the presence (A and C) or absence (B and D) of IFN-γ. Total Ig was measured in the supernatants. (E and F) B6 CD4+ T cells were cocultured with increasing ratios (10:1 to 0:1 CD4+ T cells/MSCs) of B6 or Sle1a1 MSCs and stimulated with anti-CD3/anti-CD28 Ab in the presence (E) or absence (F) of IFN-γ. The secretion of IL-2 was measured in the supernatants. The graphs show means ± SEM of triplicates. The graphs are representative of two independent assays. *p < 0.05 for two-tailed t tests comparing the value obtained for each B cell/MSC or T cell/MSC ratio to the value obtained in the absence of MSCs for each condition.

FIGURE 4.

Sle1a1 MSCs have a lower immunosuppressive capacity than do B6 MSCs in vitro. (AD) B6 B cells were cocultured with increasing ratios (10:1 to 0:1 B cells/MSCs) of B6 (filled bars) or Sle1a1 (open bars) MSCs and stimulated with LPS (A and B) or CpG (C and D) in the presence (A and C) or absence (B and D) of IFN-γ. Total Ig was measured in the supernatants. (E and F) B6 CD4+ T cells were cocultured with increasing ratios (10:1 to 0:1 CD4+ T cells/MSCs) of B6 or Sle1a1 MSCs and stimulated with anti-CD3/anti-CD28 Ab in the presence (E) or absence (F) of IFN-γ. The secretion of IL-2 was measured in the supernatants. The graphs show means ± SEM of triplicates. The graphs are representative of two independent assays. *p < 0.05 for two-tailed t tests comparing the value obtained for each B cell/MSC or T cell/MSC ratio to the value obtained in the absence of MSCs for each condition.

Close modal

We compared the ability of Sle1a1 MSCs to suppress autoimmune pathology in three models of lupus, that is, BWF1, TC, and MRL/lpr mice. All mice were positive for serum anti-dsDNA IgG at the time of treatment, indicating that disease had been initiated. All mice were sacrificed 8 wk after treatment, except for two TC mice and one MRL/lpr mouse treated with Sle1a1 MSCs that showed terminal clinical disease and were sacrificed at weeks 2, 3, and 6 after treatment, respectively.

We compared the effect of B6-derived MSC and Sle1a1-derived MSC treatments on the production of serum anti-dsDNA IgG as well as ANAs. As previously reported (24), MSC treatments had little effect on the existing production of autoantibodies regardless of the source of MSCs (Fig. 5A–C). A time course analysis of biweekly serum anti-dsDNA IgG revealed that both B6 and Sle1a1 MSC treatments of BWF1 mice resulted in the maintenance of anti-dsDNA IgG levels for the first 4 wk (data not shown), an effect that was not sustained. However, at termination of the experiment, untreated BWF1 mice had significantly higher levels of anti-dsDNA IgG than did B6 MSC-treated mice, with an intermediate level of anti-dsDNA IgG observed in mice treated with Sle1a1 MSCs (Fig. 5A). There was a trend for a decrease in anti-dsDNA IgG levels in MSC-treated TC mice (Fig. 5B) and no difference in MRL/lpr mice whether they were treated with MSCs derived from either strain or were left untreated (Fig. 5C). The analysis of terminal ANA serum levels by Hep-2 cell immunofluorescence staining confirmed the results obtained by ELISA for anti-dsDNA IgG. The serum from BWF1 mice treated with B6 MSCs, but not those treated with Sle1a1 MSCs, demonstrated a significant reduction in staining intensity as compared with controls (Fig. 5D, 5E). No consistent difference in ANA reactivity was found in the TC (Fig. 5F, 5G) and MRL/lpr (Fig. 5H, 5I) cohorts. Additionally, the staining pattern, nuclear versus cytoplasmic, was not significantly affected by either MSC treatment in any of the models tested. Taken together, these data suggest that MSC treatments were at best able prevent a further increase of the autoantibody production (anti-dsDNA IgG or ANA) in BWF1 mice, but never to reverse or significantly decrease this process. Importantly, B6 MSCs tended to be more effective than those derived from Sle1a1 mice.

FIGURE 5.

Effect of MSC treatment on autoantibody production. (AC) Relative change between the terminal and initial serum anti-dsDNA IgG levels for each of the three cohorts. (DI) ANA from terminal sera detected with FITC-conjugated anti-IgG (original magnification ×40). Two representative samples are shown per treatment and per strain. The graphs show the quantitation expressed as the average staining intensity (set with the same threshold for all samples within each cohort). The percentage of ANAhi pixels showed the same results (data not shown). *p < 0.05 for two-tailed t tests.

FIGURE 5.

Effect of MSC treatment on autoantibody production. (AC) Relative change between the terminal and initial serum anti-dsDNA IgG levels for each of the three cohorts. (DI) ANA from terminal sera detected with FITC-conjugated anti-IgG (original magnification ×40). Two representative samples are shown per treatment and per strain. The graphs show the quantitation expressed as the average staining intensity (set with the same threshold for all samples within each cohort). The percentage of ANAhi pixels showed the same results (data not shown). *p < 0.05 for two-tailed t tests.

Close modal

We also compared the renal pathology between treatments within each of the three cohorts using a variety of biomarkers. Treatment with either B6 or Sle1a1 MSCs tended to reduce the size of glomeruli in both BWF1 and TC mice, but the difference was significant only upon treatment with B6 MSCs (Fig. 6A). The glomerular size of MRL/lpr mice was not affected by either treatment. Glomerular immune complex deposition is a hallmark of lupus nephritis. Treatment with B6 MSCs significantly decreased the amount of C3 found in the glomeruli of BWF1 and MRL/lpr mice, whereas Sle1a1 MSC treatment had a significant effect only on the amount of C3 found in MRL/lpr mice (Fig. 6B, 6C). The MSC treatments had no significant effect on C3 deposits in the kidneys of TC mice. Similar results were obtained for IgG2a glomerular deposits (data not shown). To account for differences of kidney pathology between strains, as well as the different time points in disease development at which the treatment was initiated, we compared the effect of the treatments by ranking blinded samples in order of increasing severity within a cohort. BWF1 mice treated with B6 MSCs showed a significantly reduced pathology as compared with control mice (Fig. 6D). Although there was a trend in the same direction for Sle1a1 MSCs, the difference was not significant. TC mice treated with Sle1a1 MSCs showed a more severe pathology than did mice treated with B6 MSCs and equivalent to that of untreated mice (Fig. 6E). There was no effect of either treatment in MRL/lpr mice (Fig. 6F). Finally, we measured proteinuria on a weekly basis with semiquantitative strips (data not shown) as well as with quantitative microalbuminuria in the terminal samples (Fig. 6G–I). In BWF1 and TC mice, treatment with B6 MSCs significantly decreased proteinuria as compared with controls, whereas Sle1a1 MSCs had an intermediate effect in BWF1 mice and no effect in TC mice (Fig. 6G, 6H). Finally, proteinuria was generally low in our MRL/lpr cohort, with a trend in mice treated with Sle1a1 MSCs presenting higher levels of proteinuria (Fig. 6I). Body weight loss is an outward biomarker of clinical disease in both BWF1 and TC mice. BWF1 mice treated with B6 MSCs lost significantly less terminal body weight than did controls, whereas there was no difference between mice treated with Sle1a1 MSCs and controls (Fig. 6K). Similarly, TC mice treated with Sle1a1 MSCs lost significantly more terminal body weight than did mice treated with B6 MSCs (Fig. 6K). MRL/lpr mice develop an extensive lymphadenopathy that contributes greatly to an increased body weight. This gain in body weight was significantly reduced by treatment with B6 but not Sle1a1 MSCs (Fig. 6L). Overall, the Sle1a1 MSCs had a reduced ability to reduce clinical pathology in three different models of lupus as compared with B6 MSCs.

FIGURE 6.

Effect of MSC treatment on clinical disease. (A) Glomerular size in the BWF1, TC, and MRL/lpr cohorts treated with B6 or Sle1a1 MSCs or untreated controls (none). (B) Glomerular C3 deposition in the same cohorts. The two graphs show means ± SEM, and the statistical significance (*p < 0.05) corresponds to two-tailed t tests with the control group within each cohort. (C) Representative FITC-conjugated anti-C3 IgG staining of kidney sections from BWF1 and MRL/lpr mice treated with B6 or Sle1a1 MSCs or untreated controls. Original magnification ×10. (DF) Glomerulonephritis severity ranked within each cohort. (GI) Terminal albuminuria. (JL) Percentage body change 8 wk after MSC treatment. *p < 0.05, **p < 0.01 for two-tailed t or Mann–Whitney tests.

FIGURE 6.

Effect of MSC treatment on clinical disease. (A) Glomerular size in the BWF1, TC, and MRL/lpr cohorts treated with B6 or Sle1a1 MSCs or untreated controls (none). (B) Glomerular C3 deposition in the same cohorts. The two graphs show means ± SEM, and the statistical significance (*p < 0.05) corresponds to two-tailed t tests with the control group within each cohort. (C) Representative FITC-conjugated anti-C3 IgG staining of kidney sections from BWF1 and MRL/lpr mice treated with B6 or Sle1a1 MSCs or untreated controls. Original magnification ×10. (DF) Glomerulonephritis severity ranked within each cohort. (GI) Terminal albuminuria. (JL) Percentage body change 8 wk after MSC treatment. *p < 0.05, **p < 0.01 for two-tailed t or Mann–Whitney tests.

Close modal

Finally, we evaluated the immune phenotypes in the spleens of each cohort 8 wk after treatment. Neither MSC treatment resulted in significantly different numbers or percentages of CD4+ T cells, CD8+ T cells, or B cells (data not shown). No difference was observed either for the numbers or percentages of germinal center or class-switched B cells (data not shown). CD4+ T cell activation was, however, affected by MSC treatment. BWF1 mice treated with B6 but not Sle1a1 MSCs showed a significant increase of the CD62L+CD44 naive CD4+ T cells relative to the CD62LCD44+ effector memory population (Fig. 7A). This variable was not affected by either MSC treatment in the two other strains. CD25 expression on CD4+ T cells was significantly decreased by B6 but not Sle1a1 MSC treatment in all three cohorts (Fig. 7B). There was no difference, however, in the numbers and percentages of Foxp3+CD4+ T cells (data not shown). Finally, B6 MSC but not Sle1a1 MSC treatment reduced the percentage of CD4+ and CD8+ T cell blasts in the three cohorts (Fig. 7C, 7D), as well as the percentage of B cell blasts in BWF1 mice (Fig. 7E). Overall, the Sle1a1 MSCs were not as effective in reducing the immunopathology in three different models of lupus as compared with B6 MSCs.

FIGURE 7.

Effect of MSC treatment on lymphocytes. Splenic T and B cell phenotypes were compared in each cohort 8 wk after treatment. (A) Ratio of naive CD62L+CD44 over effector memory CD62LCD44+ CD4+ T cells. (B) Percentage of CD4+ T cells expressing CD25. (C) Percentage of CD4+ T cells blasts measured as forward light scatter (FSC)hi. (D) Percentage of CD8+ T cells blasts measured as FSChi. (E) Percentage of CD19+ B cells blasts measured as FSChi. *p < 0.05, **p < 0.01 for two-tailed t tests.

FIGURE 7.

Effect of MSC treatment on lymphocytes. Splenic T and B cell phenotypes were compared in each cohort 8 wk after treatment. (A) Ratio of naive CD62L+CD44 over effector memory CD62LCD44+ CD4+ T cells. (B) Percentage of CD4+ T cells expressing CD25. (C) Percentage of CD4+ T cells blasts measured as forward light scatter (FSC)hi. (D) Percentage of CD8+ T cells blasts measured as FSChi. (E) Percentage of CD19+ B cells blasts measured as FSChi. *p < 0.05, **p < 0.01 for two-tailed t tests.

Close modal

There are three checkpoints to overcome in lupus pathogenesis (37). Checkpoint 1 involves immune tolerance in the adaptive arm of the immune system, ensuring that autoreactive B and T cells are censored. Checkpoint 2 involves both the adaptive and the innate arms of the immune system to prevent peripheral amplification of the autoimmune response leading to the generation of pathogenic autoantibodies and effector lymphocytes. Checkpoint 3 involves protection of the end organs from pathogenic autoantibodies and effector immune cells. The coordinated activation of disease susceptibility genes at all three checkpoints appears to be necessary for full-blown disease in murine models and humans. The data presented in the present study and in our previous work suggest that Pbx1 is one such susceptibility gene involved in this coordinated activation. We have previously shown that Sle1a1, the locus where Pbx1 is located, regulates the production of autoreactive CD4+ T cells, as well as the homeostasis of the regulatory T cell compartment (4). We now show that Pbx1 regulates MSC immunoregulatory functions.

Pbx1 is an essential gene to maintain stem cell functions, as shown both for hematopoietic stem cells and MSCs (12, 13). MSCs serve as a reservoir for tissue regeneration through their multipotent differentiation (14) as well as a regulator of tissue and immune homeostasis (15). Specifically, MSCs suppress T cell proliferation and dendritic cell maturation, and they have pleiotropic effects on B cells, including either promotion or suppression of B cell proliferation and induction of Ab production. Therefore, MSCs have the potential to regulate checkpoints 1 and 3 in SLE disease progression, making them an attractive therapeutic.

Based on the dominant-negative function of the NZM2410-derived Pbx1-d lupus susceptibility allele (11), we hypothesized that MSCs expressing Pbx1-d would present a defective maintenance of stem cell functions with an increase in lineage-specific differentiation and thus a poor suppression of inflammation. In this study, we demonstrate that MSCs produced from the congenic B6.Sle1a1 strain that carries the NZM2410 allele of Pbx1 on a B6 background express Pbx1-d as their major isoform, whereas Pbx1-b is the major isoform in B6 MSCs. The RNA-Seq results did not support this conclusion. We think, however, that a number of technical issues, namely the relatively large number of splice isoforms, each of which with a low expression, an insufficient number of sequencing reads, and the short length of the reads, prevented the RNA-Seq to provide informative results on this issue. Please note that we obtained a high level of concordance between RNA-Seq and qRT-PCR results for other genes (Fig. 3 and data not shown). As predicted, Sle1a1 MSC surface marker phenotype, proliferation, osteoblast differentiation, as well as gene expression profiles indicate a decreased stemness and an increased lineage-specific differentiation, including the expression of hematopoietic-derived lineage markers. This was accompanied by a decreased suppressive function.

In vivo, MSCs from aged lupus-prone mice have been reported to not be as effective as those from either aged non–autoimmune mice or young predisease lupus-prone mice, with the suggestion that the inflammatory environment from which they were derived render them less effective (24). Our in vitro and in vivo studies were conducted on MSCs derived from young Sle1a1 mice, whereby this strain does not develop clinical disease (7). This excludes the possible contribution of a lupus-related inflammatory milieu to the phenotype and altered function of Sle1a1 MSCs. Many soluble factors have been proposed as the mediators of MSC immunosuppression (38). Interestingly, MSCs need to be “licensed” by the inflammatory milieu, largely through IFN-γ signaling (39). SLE is an inflammatory disease in which IFN-γ is found in large quantities (40), and therefore one could assume a robust licensing for MSC immunosuppression. IFN-γ–primed Sle1a1 MSCs provided a level of immunosuppression of B cells equivalent to that of unprimed B6 MSCs, whereas unprimed Sle1a1 MSCs lost their immunosuppressive capacity. It has been shown that TLR ligands can obstruct MSC immunosuppression (41), which may counteract the licensing effect of IFN-γ; therefore, a differential response to TLR ligands could be involved in the reduced immunosuppressive function of Sle1a1 MSCs. However, Sle1a1 MSCs were also defective in suppressing IL-2 production by CD4+ T cells, independent of TLR stimulation. Taken together, this suggests a generalized loss of immunosuppressive functions that may affect multiple pathways as the cells differentiate away from their pluripotent state.

The mechanisms by which MSC treatment ameliorates lupus severity in either patients or mice are not clear, with variable effects reported in different studies and models. Not only strain differences but also differences in the extent of MSC immunosuppressive activity have been observed: MSCs ameliorate renal disease and lymphocyte activation in all lupus models examined, but some studies have reported significant autoantibody suppression, including anti-dsDNA IgG (25), whereas others found no change (24). We compared three lupus-prone mouse models and found the most complete therapeutic effect of B6 MSC treatment in BWF1 mice and NZM2410-derived TC mice, with more variable effects in MRL/lpr mice. BWF1 and TC mice share a common origin, although they differ by several disease mechanisms (42). It is not clear at this time why these two strains were more responsive to MSC treatment than were MRL/lpr mice in our study. The cohort that we used was relatively young (8 wk old), whereas we also treated an older cohort at 14 wk of age, in which disease was too advanced to be reversed or delayed (data not shown).

Note that for all the phenotypes for which B6 MSCs showed a significant effect, Sle1a1 MSCs showed either no or an intermediate effect as compared with untreated mice. B6 but not Sle1a1 MSC treatment decreased CD25 expression on CD4+ T cells and the number of CD4+ blasts in all three models, as well as the ratio of effector memory cells relative to naive CD4+ T cells in the BWF1 mice, whereas treatment did not induce any change in the regulatory T cell populations in any of the three models, consistent with previously reported results (24). An increase in peripheral regulatory T cells has been observed in successful (18) but also failed (43) MSC transplants in SLE patients, which makes the contribution of regulatory T cells to the MSC therapeutic effect in lupus inconclusive, whereas suppression or inhibition of the inflammatory subset of activated CD4+CD25+Foxp3 cells reported in murine (44) and human (45) lupus is effective in ameliorating some symptoms of disease. Allogeneic BALB/c MSC transfer has been reported to protected MRL/lpr mice from lupus nephritis by inhibiting B cell activation (46). To the contrary, we observed minimal effect of B6 MSC transfer on B cell activation or autoantibody production in our in vivo study, with the exception that B6 and not Sle1a1 MSCs decreased autoantibody production and the formation of B cell blasts in BWF1 mice. Overall, the strongest immunosuppression induced in vivo by B6 MSCs was on CD4+ T cells, whereby the same level of suppression was not achieved by Sle1a1 MSCs.

In this study, we have focused on the defective immunosuppression of Sle1a1 MSCs. Gene expression profiles revealed a pattern of immune-related genes that were overexpressed by Sle1a1 MSCs, predicting an enhanced innate immunity and inflammation in these cells as compared with B6 controls. Some of these genes have been associated with lupus, such as IL-10 (33), IL-33 (34), and Camk4 (35). High IL-1 serum levels correlate with lupus severity (47), and polymorphisms in the IL-1A and in the IL-1 receptor antagonist genes are associated with SLE (48). Additionally, Sle1a1 MSCs secrete a reduced amount of IL-6, a cytokine that has been associated with their immune suppression in multiple studies (49, 50). We also observed that Sle1a1 MSCs express immune surface markers, which suggests that they may also differentiate into innate immune cells. It is therefore possible that MSCs expressing Pbx1-d contribute to lupus pathogenesis not only by a lack of immunosuppression, but also (or possibly mainly) by creating an inflammatory milieu. This hypothesis is supported by our in vivo suppression studies, in which we observed overall worse outcomes, such as death before the study end point, in mice treated with Sle1a1 MSCs.

In conclusion, this study showed that the lupus susceptibility gene Pbx1 not only contributes to autoimmune pathology by impairing T cell tolerance, but also impairs the function of MSCs, which may also contribute to the disease directly or indirectly. Our results also suggest that MSC therapeutic transplants could be less successful with cells collected from donors expressing Pbx1-d, which should be considered as MSC therapies are considered in an increasing number of conditions, including SLE.

We thank Dr. Lijun Yang for helpful discussions and Nathalie Kanda for excellent mouse care.

This work was supported by a University of Florida Opportunity Fund grant (to S.W.) and a Lupus Foundation of America grant (to L.M.).

The online version of this article contains supplemental material.

Abbreviations used in this article:

ANA

anti-nuclear autoantibody

B6

C57BL/6J

BM

bone marrow

BWF1

(NZB × NZW)F1

GO

gene ontology

MP

mouse phenotype

MSC

mesenchymal stem cell

Pbx1

pre–B cell leukemia homeobox 1

qRT-PCR

quantitative RT-PCR

SLE

systemic lupus erythematosus

TC

B6.Sle1.Sle2.Sle3.

1
Morel
L.
2010
.
Genetics of SLE: evidence from mouse models.
Nat. Rev. Rheumatol.
6
:
348
357
.
2
Morel
L.
2012
.
Mapping lupus susceptibility genes in the NZM2410 mouse model.
Adv. Immunol.
115
:
113
139
.
3
Morel
L.
,
Croker
B. P.
,
Blenman
K. R.
,
Mohan
C.
,
Huang
G.
,
Gilkeson
G.
,
Wakeland
E. K.
.
2000
.
Genetic reconstitution of systemic lupus erythematosus immunopathology with polycongenic murine strains.
Proc. Natl. Acad. Sci. USA
97
:
6670
6675
.
4
Cuda
C. M.
,
Li
S.
,
Liang
S.
,
Yin
Y.
,
Potula
H. H.
,
Xu
Z.
,
Sengupta
M.
,
Chen
Y.
,
Butfiloski
E.
,
Baker
H.
, et al
.
2012
.
Pre-B cell leukemia homeobox 1 is associated with lupus susceptibility in mice and humans.
J. Immunol.
188
:
604
614
.
5
Mohan
C.
,
Alas
E.
,
Morel
L.
,
Yang
P.
,
Wakeland
E. K.
.
1998
.
Genetic dissection of SLE pathogenesis. Sle1 on murine chromosome 1 leads to a selective loss of tolerance to H2A/H2B/DNA subnucleosomes.
J. Clin. Invest.
101
:
1362
1372
.
6
Chen
Y.
,
Cuda
C.
,
Morel
L.
.
2005
.
Genetic determination of T cell help in loss of tolerance to nuclear antigens.
J. Immunol.
174
:
7692
7702
.
7
Cuda
C. M.
,
Zeumer
L.
,
Sobel
E. S.
,
Croker
B. P.
,
Morel
L.
.
2010
.
Murine lupus susceptibility locus Sle1a requires the expression of two sub-loci to induce inflammatory T cells.
Genes Immun.
11
:
542
553
.
8
Morel
L.
,
Tian
X. H.
,
Croker
B. P.
,
Wakeland
E. K.
.
1999
.
Epistatic modifiers of autoimmunity in a murine model of lupus nephritis.
Immunity
11
:
131
139
.
9
Morel
L.
,
Blenman
K. R.
,
Croker
B. P.
,
Wakeland
E. K.
.
2001
.
The major murine systemic lupus erythematosus susceptibility locus, Sle1, is a cluster of functionally related genes.
Proc. Natl. Acad. Sci. USA
98
:
1787
1792
.
10
Mann
R. S.
,
Chan
S. K.
.
1996
.
Extra specificity from extradenticle: the partnership between HOX and PBX/EXD homeodomain proteins.
Trends Genet.
12
:
258
262
.
11
Sengupta
M.
,
Liang
S.
,
Potula
H. H. S.
,
Chang
L. J.
,
Morel
L.
.
2012
.
The SLE-associated Pbx1-d isoform acts as a dominant-negative transcriptional regulator.
Genes Immun.
13
:
653
657
.
12
Ficara
F.
,
Murphy
M. J.
,
Lin
M.
,
Cleary
M. L.
.
2008
.
Pbx1 regulates self-renewal of long-term hematopoietic stem cells by maintaining their quiescence.
Cell Stem Cell
2
:
484
496
.
13
Gordon
J. A.
,
Hassan
M. Q.
,
Saini
S.
,
Montecino
M.
,
van Wijnen
A. J.
,
Stein
G. S.
,
Stein
J. L.
,
Lian
J. B.
.
2010
.
Pbx1 represses osteoblastogenesis by blocking Hoxa10-mediated recruitment of chromatin remodeling factors.
Mol. Cell. Biol.
30
:
3531
3541
.
14
Djouad
F.
,
Bouffi
C.
,
Ghannam
S.
,
Noël
D.
,
Jorgensen
C.
.
2009
.
Mesenchymal stem cells: innovative therapeutic tools for rheumatic diseases.
Nat. Rev. Rheumatol.
5
:
392
399
.
15
Ghannam
S.
,
Bouffi
C.
,
Djouad
F.
,
Jorgensen
C.
,
Noël
D.
.
2010
.
Immunosuppression by mesenchymal stem cells: mechanisms and clinical applications.
Stem Cell Res Ther
1
:
2
.
16
Collins
E.
,
Gilkeson
G.
.
2013
.
Hematopoetic and mesenchymal stem cell transplantation in the treatment of refractory systemic lupus erythematosus—where are we now?
Clin. Immunol.
148
:
328
334
.
17
MacDonald
G. I. A.
,
Augello
A.
,
De Bari
C.
.
2011
.
Role of mesenchymal stem cells in reestablishing immunologic tolerance in autoimmune rheumatic diseases.
Arthritis Rheum.
63
:
2547
2557
.
18
Sun
L.
,
Wang
D.
,
Liang
J.
,
Zhang
H.
,
Feng
X.
,
Wang
H.
,
Hua
B.
,
Liu
B.
,
Ye
S.
,
Hu
X.
, et al
.
2010
.
Umbilical cord mesenchymal stem cell transplantation in severe and refractory systemic lupus erythematosus.
Arthritis Rheum.
62
:
2467
2475
.
19
Nie
Y.
,
Lau
C.
,
Lie
A.
,
Chan
G.
,
Mok
M.
.
2010
.
Defective phenotype of mesenchymal stem cells in patients with systemic lupus erythematosus.
Lupus
19
:
850
859
.
20
Li
X.
,
Liu
L.
,
Meng
D.
,
Wang
D.
,
Zhang
J.
,
Shi
D.
,
Liu
H.
,
Xu
H.
,
Lu
L.
,
Sun
L.
.
2012
.
Enhanced apoptosis and senescence of bone-marrow-derived mesenchymal stem cells in patients with systemic lupus erythematosus.
Stem Cells Dev.
21
:
2387
2394
.
21
Bocelli-Tyndall
C.
,
Bracci
L.
,
Spagnoli
G.
,
Braccini
A.
,
Bouchenaki
M.
,
Ceredig
R.
,
Pistoia
V.
,
Martin
I.
,
Tyndall
A.
.
2007
.
Bone marrow mesenchymal stromal cells (BM-MSCs) from healthy donors and auto-immune disease patients reduce the proliferation of autologous- and allogeneic-stimulated lymphocytes in vitro.
Rheumatology (Oxford)
46
:
403
408
.
22
Sun
L.
,
Akiyama
K.
,
Zhang
H.
,
Yamaza
T.
,
Hou
Y.
,
Zhao
S.
,
Xu
T.
,
Le
A.
,
Shi
S.
.
2009
.
Mesenchymal stem cell transplantation reverses multiorgan dysfunction in systemic lupus erythematosus mice and humans.
Stem Cells
27
:
1421
1432
.
23
Zhou
K.
,
Zhang
H.
,
Jin
O.
,
Fen
X.
,
Yao
G.
,
Hou
Y.
,
Sun
L.
.
2008
.
Transplantation of human bone marrow mesenchymal stem cell ameliorates the autoimmune pathogenesis in MRL/lpr mice.
Cell. Mol. Immunol.,
5
:
417
424
.
24
Gu
F.
,
Molano
I.
,
Ruiz
P.
,
Sun
L.
,
Gilkeson
G. S.
.
2012
.
Differential effect of allogeneic versus syngeneic mesenchymal stem cell transplantation in MRL/lpr and (NZB/NZW)F1 mice.
Clin. Immunol.
145
:
142
152
.
25
Akiyama
K.
,
You
Y.-O.
,
Yamaza
T.
,
Chen
C.
,
Tang
L.
,
Jin
Y.
,
Chen
X.-D.
,
Gronthos
S.
,
Shi
S.
.
2012
.
Characterization of bone marrow derived mesenchymal stem cells in suspension.
Stem Cell Res. Ther.
3
:
40
.
26
Youd
M.
,
Blickarz
C.
,
Woodworth
L.
,
Touzjian
T.
,
Edling
A.
,
Tedstone
J.
,
Ruzek
M.
,
Tubo
R.
,
Kaplan
J.
,
Lodie
T.
.
2010
.
Allogeneic mesenchymal stem cells do not protect NZB×NZW F1 mice from developing lupus disease.
Clin. Exp. Immunol.
161
:
176
186
.
27
Schena
F.
,
Gambini
C.
,
Gregorio
A.
,
Mosconi
M.
,
Reverberi
D.
,
Gattorno
M.
,
Casazza
S.
,
Uccelli
A.
,
Moretta
L.
,
Martini
A.
,
Traggiai
E.
.
2010
.
Interferon-γ-dependent inhibition of B cell activation by bone marrow-derived mesenchymal stem cells in a murine model of systemic lupus erythematosus.
Arthritis Rheum.
62
:
2776
2786
.
28
Choi
E. W.
,
Shin
I. S.
,
Park
S. Y.
,
Park
J. H.
,
Kim
J. S.
,
Yoon
E. J.
,
Kang
S. K.
,
Ra
J. C.
,
Hong
S. H.
.
2012
.
Reversal of serologic, immunologic, and histologic dysfunction in mice with systemic lupus erythematosus by long-term serial adipose tissue-derived mesenchymal stem cell transplantation.
Arthritis Rheum.
64
:
243
253
.
29
Abumaree
M.
,
Al Jumah
M.
,
Pace
R. A.
,
Kalionis
B.
.
2012
.
Immunosuppressive properties of mesenchymal stem cells.
Stem Cell Rev.
8
:
375
392
.
30
Yao
J. Q. Y.
,
Yu
F.
.
2011
.
DEB: a web interface for RNA-seq digital gene expression analysis.
Bioinformation
7
:
44
45
.
31
Xu
Z.
,
Cuda
C. M.
,
Croker
B. P.
,
Morel
L.
.
2011
.
The NZM2410-derived lupus susceptibility locus Sle2c1 increases Th17 polarization and induces nephritis in fas-deficient mice.
Arthritis Rheum.
63
:
764
774
.
32
Nakajima
H.
,
Ito
M.
,
Morikawa
Y.
,
Komori
T.
,
Fukuchi
Y.
,
Shibata
F.
,
Okamoto
S.
,
Kitamura
T.
.
2009
.
Wnt modulators, SFRP-1, and SFRP-2 are expressed in osteoblasts and differentially regulate hematopoietic stem cells.
Biochem. Biophys. Res. Commun.
390
:
65
70
.
33
Tian
G.
,
Li
J.-L.
,
Wang
D.-G.
,
Zhou
D.
.
2014
.
Targeting IL-10 in auto-immune diseases.
Cell Biochem. Biophys.
70
:
37
49
.
34
Wang
S.
,
Ding
L.
,
Liu
S. S.
,
Wang
C.
,
Leng
R. X.
,
Chen
G. M.
,
Fan
Y. G.
,
Pan
H. F.
,
Ye
D. Q.
.
2012
.
IL-33: a potential therapeutic target in autoimmune diseases.
J. Investig. Med.
60
:
1151
1156
.
35
Koga
T.
,
Hedrich
C. M.
,
Mizui
M.
,
Yoshida
N.
,
Otomo
K.
,
Lieberman
L. A.
,
Rauen
T.
,
Crispín
J. C.
,
Tsokos
G. C.
.
2014
.
CaMK4-dependent activation of AKT/mTOR and CREM-α underlies autoimmunity-associated Th17 imbalance.
J. Clin. Invest.
124
:
2234
2245
.
36
Villarreal
D. O.
,
Weiner
D. B.
.
2014
.
Interleukin 33: a switch-hitting cytokine.
Curr. Opin. Immunol.
28
:
102
106
.
37
Wakeland
E. K.
,
Liu
K.
,
Graham
R. R.
,
Behrens
T. W.
.
2001
.
Delineating the genetic basis of systemic lupus erythematosus.
Immunity
15
:
397
408
.
38
Wang
H.
,
Wu
M.
,
Liu
Y.
.
2012
.
Are mesenchymal stem cells major sources of safe signals in immune system?
Cell. Immunol.
272
:
112
116
.
39
Marigo
I.
,
Dazzi
F.
.
2011
.
The immunomodulatory properties of mesenchymal stem cells.
Semin. Immunopathol.
33
:
593
602
.
40
Hayashi
T.
2010
.
Therapeutic strategies for SLE involving cytokines: mechanism-oriented therapies especially IFN-γ targeting gene therapy.
J. Biomed. Biotechnol.
2010
:
461641
.
41
Krampera
M.
2011
.
Mesenchymal stromal cell “licensing”: a multistep process.
Leukemia
25
:
1408
1414
.
42
Sang
A.
,
Yin
Y.
,
Zheng
Y.-Y.
,
Morel
L.
.
2012
.
Animal models of molecular pathology: systemic lupus erythematosus.
Prog. Mol. Biol. Transl. Sci.
105
:
321
370
.
43
Carrion
F.
,
Nova
E.
,
Ruiz
C.
,
Diaz
F.
,
Inostroza
C.
,
Rojo
D.
,
Mönckeberg
G.
,
Figueroa
F. E.
.
2010
.
Autologous mesenchymal stem cell treatment increased T regulatory cells with no effect on disease activity in two systemic lupus erythematosus patients.
Lupus
19
:
317
322
.
44
Brinster
C.
,
Shevach
E. M.
.
2008
.
Costimulatory effects of IL-1 on the expansion/differentiation of CD4+CD25+Foxp3+ and CD4+CD25+Foxp3 T cells.
J. Leukoc. Biol.
84
:
480
487
.
45
Triplett
T. A.
,
Curti
B. D.
,
Bonafede
P. R.
,
Miller
W. L.
,
Walker
E. B.
,
Weinberg
A. D.
.
2012
.
Defining a functionally distinct subset of human memory CD4+ T cells that are CD25POS and FOXP3NEG.
Eur. J. Immunol.
42
:
1893
1905
.
46
Ma
X.
,
Che
N.
,
Gu
Z.
,
Huang
J.
,
Wang
D.
,
Liang
J.
,
Hou
Y.
,
Gilkeson
G.
,
Lu
L.
,
Sun
L.
.
2013
.
Allogenic mesenchymal stem cell transplantation ameliorates nephritis in lupus mice via inhibition of B-cell activation.
Cell Transplant.
22
:
2279
2290
.
47
Cigni
A.
,
Pileri
P. V.
,
Faedda
R.
,
Gallo
P.
,
Sini
A.
,
Satta
A. E.
,
Marras
R.
,
Carta
E.
,
Argiolas
D.
,
Rum
I.
,
Masala
A.
.
2014
.
Interleukin 1, interleukin 6, interleukin 10, and tumor necrosis factor α in active and quiescent systemic lupus erythematosus.
J. Investig. Med.
62
:
825
829
.
48
Song
G. G.
,
Kim
J.-H.
,
Seo
Y. H.
,
Choi
S. J.
,
Ji
J. D.
,
Lee
Y. H.
.
2014
.
Associations between interleukin 1 polymorphisms and susceptibility to systemic lupus erythematosus: a meta-analysis.
Hum. Immunol.
75
:
105
112
.
49
Li
P.
,
Li
S.-H.
,
Wu
J.
,
Zang
W.-F.
,
Dhingra
S.
,
Sun
L.
,
Weisel
R. D.
,
Li
R.-K.
.
2013
.
Interleukin-6 downregulation with mesenchymal stem cell differentiation results in loss of immunoprivilege.
J. Cell. Mol. Med.
17
:
1136
1145
.
50
Deng
Y.
,
Yi
S.
,
Wang
G.
,
Cheng
J.
,
Zhang
Y.
,
Chen
W.
,
Tai
Y.
,
Chen
S.
,
Chen
G.
,
Liu
W.
, et al
.
2014
.
Umbilical cord-derived mesenchymal stem cells instruct dendritic cells to acquire tolerogenic phenotypes through the IL-6-mediated upregulation of SOCS1.
Stem Cells Dev.
23
:
2080
2092
.

The authors have no financial conflicts of interest.

Supplementary data