Abstract
The role of ICOS and its ligand (ICOSL) have both been shown to be essential for proper humoral responses as well as autoimmune Ab development in mouse models of lupus. In this paper, we report a specific role for the metalloprotease ADAM10 on B cells in regulating both ICOSL and ICOS in a mouse model of increased humoral immunity using B6mir146a−/− mice and a model of lymphoproliferative disease using the well-characterized lpr model. B6lpr mice lacking ADAM10 on B cells (A10Blpr) have decreased nodal proliferation and T cell accumulation compared with control B6lpr mice. Additionally, A10Blpr mice have a drastic reduction in autoimmune anti-dsDNA Ab production. In line with this, we found a significant reduction in follicular helper T cells and germinal center B cells in these mice. We also show that lymphoproliferation in this model is closely tied to elevated ICOS levels and decreased ICOSL levels. Overall, our data not only show a role of B cell ADAM10 in control autoimmunity but also increase our understanding of the regulation of ICOS and ICOSL in the context of autoimmunity.
Introduction
The role of the ICOS of T cells (ICOS) has been well characterized and has been shown to be essential for follicular helper T cell (TFH), T effector cell (Teff), and Th2 functions (1–4). Loss of ICOS has been shown to result in blunted thymus-dependent (T-dependent) Ab responses as well as absent germinal center (GC) formation. The only known ligand for ICOS (ICOSL) is expressed on APCs, such as dendritic cells, B cells, and other hematopoietic and nonhematopoietic cells (5). ICOSL-deficient mice have a similar phenotype to ICOS-deficient mice, suggesting the importance of their cognate interaction (6).
Studies regarding the protein and transcriptional regulation of ICOS and ICOSL in autoimmune disease have been limited. In steady-state mice, Icos transcriptional levels have been shown to rapidly increase upon TCR stimulation (1, 4). Additionally, several studies have shown the importance of ICOS posttranscriptional regulation through Roquin-1 and mir146a (7–9). A loss-of-function mutation in Roquin-1 (sanroque mice) or loss of mir146a in mice results in elevated ICOS levels, ultimately resulting in exaggerated GC responses and Ab production (8, 9). Additionally, sanroque mice develop an autoimmune phenotype resembling lupus with autoantibody production (7, 8). These studies clearly show the importance of proper ICOS regulation in maintaining homeostasis between productive GC responses and autoimmunity.
Disease progression in lupus-prone B6.MRL-Faslpr/J (Faslpr) mice and other lupus-prone models is also associated with altered levels of ICOS and ICOSL (10–13). Several groups have shown that ICOS is required for class-switched autoantibody production in MRL.Faslpr and Sle1 mice (10, 13). The origin of the T cells responsible for this B cell help and autoantibody production are thought to be extrafollicular in nature but resemble TFH in gene expression and cytokine production, mainly IL-21 and CD40L (14–18). Several studies have contradictory results regarding the role of ICOS in Teff function in the Faslpr model. More recently it has been suggested that ICOSL on CD11c+ cells promotes T cell survival and effector function in the kidneys (12). However, these mice were not protected from the development of autoimmune Abs, whereas B cell conditional ICOSL knockout mice developed reduced autoantibody, suggesting differential roles for B cell and dendritic cell ICOSL (12) and further suggesting a multifaceted role for ICOSL.
We have recently shown that mice that conditionally lack a disintegrin and metalloproteinase 10 (ADAM10) on B cells (A10B) have elevated ICOSL on this cell due to the inability to shed ICOSL from the cell surface (19). These mice having decreased GC responses and Ab production. The mechanism for this defect in humoral immunity was the finding that the increase in ICOSL on the B cell surface led to a posttranslational downregulation of surface ICOS levels on T cells. This regulation was examined in naive, 4-hydroxy-3-nitrophenylacetyl hapten conjugated to keyhole limpet hemocyanin (NP31-KLH)–immunized, experimental autoimmune encephalomyelitis, and house dust mite–challenged mice and was able to effectively downregulate ICOS levels to block TFH responses and affinity-matured Ab production (19). These studies suggested that in addition to proper translational regulation of ICOS and ICOSL, proper posttranslational regulation of these proteins is just as important for regulating humoral immunity.
In this study, first we show that B cell ADAM10 is necessary for the enhanced ICOS and TFH expression that is associated with the B6mir146a−/− mice (9) and that loss of B cell ADAM10 ablated the increased TFH accumulation seen in these mice. Additionally, we show that loss of B cell ADAM10 in the lupus-prone Faslpr mouse model results in decreased TFH accumulation and more importantly a decrease in anti-dsDNA Abs. Our results indicate that B cell ADAM10 represents a novel mechanism of ICOSL and ICOS regulation in the context of humoral autoimmunity in models where enhanced immune responses are seen, and this novel mechanism even extends to the lupus model (one of the most severe of autoimmune diseases).
Materials and Methods
Mice
All mice were maintained at the Virginia Commonwealth University Animal Facility in accordance with guidelines by the U.S. National Institutes of Health and the American Association for the Accreditation of Laboratory Animals Care. C57BL/6J ADAM10-floxed mice crossed to the CD19-cre mouse were generated previously (20). Faslpr mice were purchased from The Jackson Laboratory (000482). These mice were crossed to Adam10fl/fl-floxed CD19cre+/− mice. For lpr studies, Faslpr/lpr Adam10fl/fl Cre−/− are referred to as B6lpr, Faslpr/lpr Adam10fl/fl Cre+/− are referred to as A10Blpr, Fas−/− Adam10fl/fl Cre−/− are referred to as B6, and Fas−/− Adam10fl/fl Cre+/− are referred to as A10B. B6mir146a−/− mice were purchased from The Jackson Laboratory (016239) and crossed to Adam10fl/fl-floxed CD19cre+/− mice. For Mir146a studies, mir146a−/− Adam10fl/fl Cre−/− mice are referred to as B6mir146a−/−, mir146a−/− Adam10fl/fl Cre+/− mice are referred to as A10Bmir146a−/−, mir146a+/+ Adam10fl/fl Cre−/− mice are referred to as B6, and mir146a−/− Adam10fl/fl Cre+/− mice are referred to as A10B.
Histology
Kidneys and lungs were excised and trimmed followed by fixation in 10% formalin for 48 h. Organs were paraffin embedded, sectioned, deparaffinized, and rehydrated as described (21). Sections were stained with H&E and imaged using an Olympus BX41 microscope.
Fluorescence-linked immunosorbent assay
Serum was collected from mice by cardiac puncture and allowed to clot for at least 30 min at room temperature. Serum was then separated from clotted blood components by centrifugation and stored at −20°C until use. For total Ab assays, 384-well FLUOTRAC 600 (Greiner Bio-One) were coated with 20 μl of 5 μg/ml goat anti-mouse IgG1 (1071-01; Southern Biotech), IgG2a (1081-01; Southern Biotech), IgG3 (1101-01; Southern Biotech), or IgM (1021-01; Southern Biotech) in borate-buffered saline overnight at 4°C. Plates were washed (PBS with 0.02% Tween-20) and blocked with 20 μl of 5% FBS in PBS and for 2 h at room temperature. Twenty microliters of serum dilutions and Ab standards were added to wells and incubated overnight at 4°C. Plates were washed and Abs were detected using 20 μl of biotinylated goat anti-mouse IgG (1036-08; Southern Biotech) for all IgG isotypes and 20 μl of biotinylated goat anti-mouse IgM (1021-08; Southern Biotech) for IgM assays at 2 μg/ml overnight at 4°C. Plates were washed and incubated with 20 μl of 2 μg/ml PE-streptavidin (405204; BioLegend) in block for 2 h at room temperature. Plates were then washed, and 30 μl of PBS was added to all wells and analyzed with a SpectraMax M5 with an excitation wavelength of 496 and emission wavelength of 574. For anti-dsDNA assays, the 384-well FLUOTRAC 600 plates were coated with 20 μg/ml protamine sulfate in BBS overnight at 4°C. Plates were washed and incubated with 5 μg/ml highly polymerized calf thymus DNA (Worthington) in PBS overnight at 4°C. Plates were washed and serum dilutions were added and incubated overnight at 4°C. After washing, detection was performed as stated for total Ab assays.
Immunofluorescence microscopy
Kidneys were frozen on dry ice in OCT compound (Tissue-Tek). Ten-micrometer sections were cut from frozen blocks using a cryostat (Frigocut 2800E; Jung), fixed in absolute ice-cold acetone for 10 min and air dried. Sections were rehydrated in PBS and blocked in 4% horse serum and 3% BSA in PBS for 60 min at room temperature. Sections were washed and labeled with 5 μg/ml FITC goat anti-mouse IgM (1021-02; Southern Biotech) or 5 μg/ml Alexa Fluor 647 goat anti-mouse IgG-Fc (1033-31; Southern Biotech) for 60 min at room temperature. Images were captured on a LSM700 Axio Imager 2.
Abs and flow cytometry
Lymph nodes were excised and teased apart followed by mechanical digestion through a 100-μm cell strainer. Spleens were excised and mechanically forced through a 100-μm cell strainer followed by ACK lysis of RBCs. Kidneys were excised and cut into <1 mm pieces using a razor and then digested at 37°C for 45 min with 0.05 mg/ml Liberase TM (Roche) and 0.05 mg/ml DNAse1 (Worthington). Digested tissue was then mechanically passed through a 100-μm filter followed by ACK (Quality Biologic) lysis of RBCs. Single-cell suspensions of all organs were washed with PBS, and live/dead staining was done using Zombie Aqua (423102; BioLegend) according to the manufacturer’s protocol. Cells were washed with FACS buffer (5% FBS in PBS with 2 mM EDTA). Fc receptors were blocked with 5 μg 2.4g2 (22) for 10 min at 4°C. Abs were added at indicated concentrations (Table I) for 45 min at 4°C. Cells were washed two times with FACS buffer and fixed in Fixation Buffer (420801; BioLegend), or secondaries were added and incubated for 30 min at 4°C followed by fixation. For intracellular staining, following fixation, cells were permeabilized using Intracellular Stain Permeabilization Buffer (421002; BioLegend) according to the manufacturer’s protocol. Cells were stained for intracellular markers for 60 min at room temperature and washed and fixed. Flow cytometry data were collected on an LSR Fortessa II and analyzed in FCS Express 5. Gating strategies and cell surface phenotypes can be found in Supplemental Fig. 1.
Immunizations
Ten micrograms of NP31-KLH (BioSearch Technologies) was added to PBS containing 0.25 mg alum/ml (Imject, Sigma-Aldrich) in a volume of 25 μl and injected into each hind footpad. Popliteal lymph nodes (popLN) were isolated from mice 14 d postimmunization.
Statistical analyses
All statistical analyses used GraphPad Prism 7.3. Individual figure legends indicate statistical tests used for different experiments. Overall, Bartlett test for equal variance was used, and if variances were not equal, equivalent nonparametric tests as indicated in figure legends were used.
Results
B cell ADAM10 blocks the increased humoral immune responses seen in the absence of mir146a
We first sought to examine the ability of B cell ADAM10 to regulate elevated humoral immune responses seen in the B6mir146a−/− mice. B6mir146a−/− mice have elevated ICOS levels as well as elevated TFH responses following immunization (9). ADAM10 B cell conditional knockout mice (A10B) (20) were crossed to B6mir146a−/− mice (9) to make A10Bmir146a−/− mice to examine whether B cell ADAM10 could effectively regulate humoral immune responses in this model. We first examined ICOSL levels and saw that A10Bmir146a−/− mice had elevated levels of ICOSL on B cells similar to A10B mice. In both cases, ICOSL was significantly higher than wild-type C57BL/6J (B6) or B6mir146a−/− mice (Fig. 1A). B6mir146a−/− CD4+ T cells from the nondraining popLN repeated the previously reported increase in ICOS compared with B6 mice (9); however, loss of B cell ADAM10 resulted in decreased ICOS levels in both situations (Fig. 1B). Additionally, when adoptively transferred CD4+ T cells from B6mir146a−/− mice into either B6 or A10B mice, we saw that ICOS levels on donor T cells from B6mir146a−/− mice dropped significantly in the A10B recipients but not the B6 recipients (Fig. 1C).
Loss of B cell ADAM10 reverses increased GC responses seen in mir146a−/− mice. (A) ICOSL MFI as determined by flow cytometry from B220+ B cells from the contralateral (nondraining) popLN of NP-KLH–immunized mice. (B) ICOS MFI of CD4+ T cells as determined by flow cytometry from the contralateral popLN of NP-KLH–immunized mice. (C) ICOS MFI levels on B6mir146a−/− CD4+ T cells from naive mice adoptively transferred into either B6 or A10B mice and analyzed before and 24 h after adoptive transfer. (D) Representative pseudocolor dot plots of draining popLN CD4+ T cells from NP31-KLH–immunized mice indicating TFH in the gated area at day 14 postimmunization (p.i.). Relative (E) and absolute number of (F) TFH of CD4+ T cells in the immunized popLN at day 14 p.i. (G) Representative pseudocolor dot plots of draining popLN B220+ B cells from NP-KLH–immunized mice with GC B cells in gated area at day 14 p.i. Relative (H) and absolute (I) number of GC B cells of B220+ B cells in the draining popLN at day 14 p.i. (J) Representative histogram of CD4+ TFH ICOS draining popLN of NP-KLH–immunized mice at day 14 p.i. (K) Statistical analysis of ICOS MFI of TFH from (J). One-way ANOVA with Tukey posttest (A, B, E, G, and I). Data are pooled from two (A, B, E, G, and I; mean) independent experiments. n.s., not significant (p ≥ 0.05), *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. MFI, mean fluorescence intensity.
Loss of B cell ADAM10 reverses increased GC responses seen in mir146a−/− mice. (A) ICOSL MFI as determined by flow cytometry from B220+ B cells from the contralateral (nondraining) popLN of NP-KLH–immunized mice. (B) ICOS MFI of CD4+ T cells as determined by flow cytometry from the contralateral popLN of NP-KLH–immunized mice. (C) ICOS MFI levels on B6mir146a−/− CD4+ T cells from naive mice adoptively transferred into either B6 or A10B mice and analyzed before and 24 h after adoptive transfer. (D) Representative pseudocolor dot plots of draining popLN CD4+ T cells from NP31-KLH–immunized mice indicating TFH in the gated area at day 14 postimmunization (p.i.). Relative (E) and absolute number of (F) TFH of CD4+ T cells in the immunized popLN at day 14 p.i. (G) Representative pseudocolor dot plots of draining popLN B220+ B cells from NP-KLH–immunized mice with GC B cells in gated area at day 14 p.i. Relative (H) and absolute (I) number of GC B cells of B220+ B cells in the draining popLN at day 14 p.i. (J) Representative histogram of CD4+ TFH ICOS draining popLN of NP-KLH–immunized mice at day 14 p.i. (K) Statistical analysis of ICOS MFI of TFH from (J). One-way ANOVA with Tukey posttest (A, B, E, G, and I). Data are pooled from two (A, B, E, G, and I; mean) independent experiments. n.s., not significant (p ≥ 0.05), *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. MFI, mean fluorescence intensity.
Target . | Conjugate . | Company . | Clone . | Product No. . | Lot No. . | Concentration Used . |
---|---|---|---|---|---|---|
B220 | BUV395 | BD Biosciences | RA3-6B2 | 563793 | 6320735 | 1:400 |
CD127 | BUV737 | BD Biosciences | SB/199 | 564399 | 7339546 | 1:400 |
CD3e | BUV737 | BD Biosciences | 145-2C11 | 564618 | 7158789 | 1:400 |
CD62L | BV421 | BD Biosciences | MEL-14 | 562910 | 7054905 | 1:400 |
PD1 | BV421 | BioLegend | 29F.1A12 | 135218 | B213657 | 1:400 |
CD23 | BV421 | BioLegend | B3B4 | 101621 | B246546 | 1:400 |
CD90.2 | BV605 | BioLegend | 30-H12 | 105343 | B242640 | 1:400 |
CD19 | BV650 | BioLegend | 6D5 | 115541 | B252162 | 1:400 |
CD138 | BV711 | BioLegend | 281-2 | 142519 | B211818 | 1:400 |
ICOS | BV785 | BioLegend | C398.4A | 313534 | B244412 | 1:400 |
CD45.2 | PE | BioLegend | 104 | 109807 | B195069 | 1:400 |
ICOSL | PE | BioLegend | HK5.3 | 107405 | B225590 | 1:400 |
CD8a | PE/Dazzle594 | BioLegend | 53-6.7 | 100762 | B232092 | 1:400 |
CD45 | Allophycocyanin-Fire750 | BioLegend | 30-F11 | 103154 | B226658 | 1:400 |
CXCR5 | Biotin | BioLegend | L138D7 | 145510 | B214657 | 1:400 |
CD11b | PE/Cy7 | BioLegend | M1/70 | 101216 | B203625 | 1:400 |
ICOS | PE | BioLegend | C398.4A | 313508 | B186195 | 1:400 |
GL7 | AF647 | BD Biosciences | GL7 | 561529 | 249042 | 1:400 |
CD44 | AF700 | BioLegend | IM7 | 103026 | B244378 | 1:400 |
CD4 | Allophycocyanin-Fire750 | BioLegend | GK1.5 | 100460 | B244105 | 1:400 |
IFN-γ | FITC | BioLegend | XMG1.2 | 505806 | B237657 | 1:400 |
Streptavidin | PE/Cy7 | BioLegend | 405206 | B207311 | 1:200 |
Target . | Conjugate . | Company . | Clone . | Product No. . | Lot No. . | Concentration Used . |
---|---|---|---|---|---|---|
B220 | BUV395 | BD Biosciences | RA3-6B2 | 563793 | 6320735 | 1:400 |
CD127 | BUV737 | BD Biosciences | SB/199 | 564399 | 7339546 | 1:400 |
CD3e | BUV737 | BD Biosciences | 145-2C11 | 564618 | 7158789 | 1:400 |
CD62L | BV421 | BD Biosciences | MEL-14 | 562910 | 7054905 | 1:400 |
PD1 | BV421 | BioLegend | 29F.1A12 | 135218 | B213657 | 1:400 |
CD23 | BV421 | BioLegend | B3B4 | 101621 | B246546 | 1:400 |
CD90.2 | BV605 | BioLegend | 30-H12 | 105343 | B242640 | 1:400 |
CD19 | BV650 | BioLegend | 6D5 | 115541 | B252162 | 1:400 |
CD138 | BV711 | BioLegend | 281-2 | 142519 | B211818 | 1:400 |
ICOS | BV785 | BioLegend | C398.4A | 313534 | B244412 | 1:400 |
CD45.2 | PE | BioLegend | 104 | 109807 | B195069 | 1:400 |
ICOSL | PE | BioLegend | HK5.3 | 107405 | B225590 | 1:400 |
CD8a | PE/Dazzle594 | BioLegend | 53-6.7 | 100762 | B232092 | 1:400 |
CD45 | Allophycocyanin-Fire750 | BioLegend | 30-F11 | 103154 | B226658 | 1:400 |
CXCR5 | Biotin | BioLegend | L138D7 | 145510 | B214657 | 1:400 |
CD11b | PE/Cy7 | BioLegend | M1/70 | 101216 | B203625 | 1:400 |
ICOS | PE | BioLegend | C398.4A | 313508 | B186195 | 1:400 |
GL7 | AF647 | BD Biosciences | GL7 | 561529 | 249042 | 1:400 |
CD44 | AF700 | BioLegend | IM7 | 103026 | B244378 | 1:400 |
CD4 | Allophycocyanin-Fire750 | BioLegend | GK1.5 | 100460 | B244105 | 1:400 |
IFN-γ | FITC | BioLegend | XMG1.2 | 505806 | B237657 | 1:400 |
Streptavidin | PE/Cy7 | BioLegend | 405206 | B207311 | 1:200 |
All mAbs were used at indicated concentrations for flow cytometry experiments according to protocols in 2Materials and Methods.
We next immunized these mice with a T-dependent immunization model using NP31-KLH and examined GC responses. As previously reported, B6mir146a−/− mice had a significant increase in TFH compared with B6 mice (Fig. 1D–F). However, A10Bmir146a−/− mice had similar TFH levels compared with A10B mice and significantly lower relative and absolute TFH numbers than both B6 and B6mir146a−/− mice (Fig. 1D–F). Supporting our previous findings (19), we saw significantly decreased absolute and relative GC B cells in A10B mice compared with B6 mice in the draining popLN (Fig. 1G–I). Additionally, we saw significantly reduced absolute and relative GC B cells numbers in A10Bmir146a−/− mice compared with B6mir146a−/− mice (Fig. 1G–I). As also reported (9), B6mir146a−/− mice had significantly higher TFH ICOS levels than B6 mice (Fig. 1J, 1K). However, TFH ICOS levels were significantly lower in A10Bmir146a−/− mice compared with both B6 and B6mir146a−/− mice and were not significantly different from A10B mice (Fig. 1J, 1K). Thus, loss of mir146 does not overcome the suppression of ICOS or the decreased GC response that was seen in A10 animals (19, 23).
Loss of B cell ADAM10 affects lymph node cellularity and T cell activation
We next turned to a more severe model where overstimulation of the immune response is seen, the Faslpr model (24). By appropriate backcrossing, B6lpr mice that lacked ADAM10 on their B cells (A10Blpr) were generated. The B6 strain has a delayed onset compared with the MRL, with 6 mo of age being the common onset of disease (24). Thus, our initial studies were performed at this age. Splenomegaly was evident in both B6lpr and A10Blpr strains, and no significant difference in splenic leukocyte (Fig. 2A) or infiltrating kidney leukocytes (Fig. 2B) and kidney T cells (Fig. 2C) were seen. A10Blpr mice had similar T cell subset distributions in the spleen (Supplemental Fig. 2A–D) and kidneys (Supplemental Fig. 2E–H) compared with B6lpr mice. We also did not observe any pathologic differences in the lungs or kidneys between A10Blpr and B6lpr mice as observed by H&E staining (Supplemental Fig. 2I, 2J). For unknown reasons, CD8+ T cell numbers in B6 and A10B were lower than B6lpr and A10Blpr (Supplemental Fig. 2C). There was a significant reduction (∼10-fold) in total cervical lymph node (cervLN) CD45+ cell number in the A10Blpr compared with B6lpr mice (Fig. 2D).
B cell ADAM10 regulates cervLN T cell activation in lpr mice. CD45+ cells were enumerated from the spleens (A) and kidneys (B) of indicated mice. (C) CD3+ T cells were enumerated from the kidneys of indicated mice. (D) CD45+ cells were enumerated from cervLN of indicated mice. (E) Representative pseudocolor dot plots of cervLN B220− CD3+ T cells. Relative (F) and absolute (G) numbers of CD4+ T cells that are activated (CD44+ CD62L−). Relative (H) and absolute (I) numbers Teff of CD4+ T cells. Relative (J) and absolute (K) numbers Tem of CD4+ T cells. One-way ANOVA with Tukey posttest (A, E, F, G, and H). Unpaired Student t test (B and C). Data are pooled from three (A, E, F, G, and H; mean) and two (B and C; mean) independent experiments. n.s., not significant (p ≥ 0.05), *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.
B cell ADAM10 regulates cervLN T cell activation in lpr mice. CD45+ cells were enumerated from the spleens (A) and kidneys (B) of indicated mice. (C) CD3+ T cells were enumerated from the kidneys of indicated mice. (D) CD45+ cells were enumerated from cervLN of indicated mice. (E) Representative pseudocolor dot plots of cervLN B220− CD3+ T cells. Relative (F) and absolute (G) numbers of CD4+ T cells that are activated (CD44+ CD62L−). Relative (H) and absolute (I) numbers Teff of CD4+ T cells. Relative (J) and absolute (K) numbers Tem of CD4+ T cells. One-way ANOVA with Tukey posttest (A, E, F, G, and H). Unpaired Student t test (B and C). Data are pooled from three (A, E, F, G, and H; mean) and two (B and C; mean) independent experiments. n.s., not significant (p ≥ 0.05), *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.
When we examined cervLN CD4+ T cells for T cell activation status, we saw that there was a significantly reduced percentage of activated CD4+ T cells (CD44+ CD62L−) in A10Blpr mice versus B6lpr mice (Fig. 2E–G). Additionally, both B6 and A10B mice had significantly fewer relative and absolute numbers of activated CD4+ T cells compared with their lpr counterpart (Fig. 2E–G). To further examine T cell activation states, we used CD127 as a marker for differentiation between T effector memory cells (Tem) and Teff (12) gated on CD4+ CD44+ CD62L− T cells. We saw a significant decrease in absolute but not relative levels of CD4+ Teff in the cervLN of A10Blpr mice versus B6lpr mice (Fig. 2E, 2H, 2I). Additionally, we saw significant decreases in both relative and absolute numbers of CD4+ Tem in the cervLN of A10Blpr mice versus B6lpr mice (Fig. 2E, 2J, 2K). We did not see a difference in relative or absolute CD4+ Tem or CD4+ Teff between A10B and B6 mice, but we did see a difference in absolute and relative CD4+ Tem or CD4+ Teff between B6 and B6lpr mice as well as A10B and A10Blpr mice (Fig. 2E, 2H–K).
A10Blpr mice have decreased GC responses
With our previous report showing that A10B mice have elevated ICOSL levels and decreased ICOS levels (19), we next investigated whether these altered ICOS and ICOSL levels were also present in the Faslpr model. B cells from A10Blpr mice had a significant increase in ICOSL levels compared with B6lpr mice; however, these ICOSL levels were significantly decreased when compared with A10B mice (Fig. 3A). We saw significantly lower ICOS levels on CD4+ T cells in A10Blpr mice compared with B6lpr mice (Fig. 3B). Interestingly, B6lpr mice had significantly higher levels of ICOS on CD4+ T cells than age-matched B6 mice, and A10Blpr mice had significantly higher levels of ICOS than A10B mice (Fig. 3B).
B cell ADAM10 controls GC responses in lpr mice through ICOSL regulation. cervLN B cell ICOSL (A) and CD4+ T cell ICOS (B) levels were determined by flow cytometry. (C) Representative pseudocolor dot plots of cervLN CD19+ B220+ B cells; gate indicates GL7+ population. (D) Percentage of GL7+ cells gated from CD19+ B220+. (E) Absolute number of GL7+ B cells in the cervLN. (F) Representative pseudocolor dot plots of CD3+ CD4+ T cells from the cervLN of indicated mice. (G) Percentage of TFH gated from CD3+ CD4+ cells in the cervLN. (H) Absolute number of TFH in the cervLN of indicated groups. (I) Representative histogram of ICOS expression on TFH in the cervLN. (J) ICOS MFI of cervLN TFH as determined by flow cytometry. n.s., not significant (p ≥ 0.05), *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. One-way ANOVA with Tukey posttest (A, B, D, E, G, H, and J). Data are pooled from three (A, B, D, E, G, H, and J; mean) independent experiments. MFI, mean fluorescence intensity.
B cell ADAM10 controls GC responses in lpr mice through ICOSL regulation. cervLN B cell ICOSL (A) and CD4+ T cell ICOS (B) levels were determined by flow cytometry. (C) Representative pseudocolor dot plots of cervLN CD19+ B220+ B cells; gate indicates GL7+ population. (D) Percentage of GL7+ cells gated from CD19+ B220+. (E) Absolute number of GL7+ B cells in the cervLN. (F) Representative pseudocolor dot plots of CD3+ CD4+ T cells from the cervLN of indicated mice. (G) Percentage of TFH gated from CD3+ CD4+ cells in the cervLN. (H) Absolute number of TFH in the cervLN of indicated groups. (I) Representative histogram of ICOS expression on TFH in the cervLN. (J) ICOS MFI of cervLN TFH as determined by flow cytometry. n.s., not significant (p ≥ 0.05), *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. One-way ANOVA with Tukey posttest (A, B, D, E, G, H, and J). Data are pooled from three (A, B, D, E, G, H, and J; mean) independent experiments. MFI, mean fluorescence intensity.
Because of the role of ICOS and ICOSL in GC reactions, we next examined GC responses in the cervLN in these mice. We did not see a difference in relative number of GL7+ cells in the cervLN (Fig. 3C, 3D) but did observe decreased absolute numbers of GL7+ B cells (Fig. 3E) in A10Blpr mice, potentially due to decreased cervLN cellularity in A10Blpr mice. When we examined the spleen, there was a significant decrease in percentage and absolute number of GL7+ GC B cells in the spleen (Supplemental Fig. 3A–C) in A10Blpr mice.
TFH were decreased in both percentage as well as absolute number in the cervLN of A10Blpr mice compared with B6lpr mice at 6 mo of age (Fig. 3F–H). TFH were significantly decreased in absolute number but not relative number in the spleens of A10Blpr mice compared B6lpr mice (Supplemental Fig. 3D–F). In both the spleen and cervLN, TFH were decreased in B6lpr compared with B6 as well as A10Blpr compared with A10B mice (Fig. 3F–H, Supplemental Fig. 3D–F). CervLN TFH ICOS levels were significantly decreased in A10Blpr mice compared with B6lpr mice (Fig. 3I, 3J). This trend was similar in age-matched A10B and B6 controls, albeit with higher ICOS levels.
We saw no difference in relative or absolute numbers of CD4+ IFN-γ+ T cells between A10Blpr mice B6lpr mice in CD4+ T cells in both the spleen (Supplemental Fig. 3G–J) and cervLN (Supplemental Fig. 3K–M). However, the absolute number of CD4+ IFN-γ+ T cells in the cervLN trended down but was NS in the A10Blpr mice because of the decrease in cellularity seen in these mice (Supplemental Fig. 3L).
A10Blpr mice have decreased autoantibody levels
Because of the decreased GC and TFH numbers in the A10Blpr mice, we examined whether there were any differences in total serum Abs as well as autoantibody production in these mice. Six-month-old A10Blpr mice had a significant decrease in total IgG1 and IgG2a levels but had no significant difference in total IgM or IgG3 (Fig. 4A). Anti-dsDNA–specific Abs were used to evaluate auto-Ab. We saw a modest decrease in anti-dsDNA IgM at 6 mo of age but a larger and significant decrease at 8 mo in A10Blpr mice compared with B6lpr mice (Fig. 4B). Anti-dsDNA IgG was clearly suppressed at both 6 and 8 mo of age in A10Blpr mice compared with B6lpr mice (Fig. 4C). Although 6-mo-old A10Blpr mice did not have a noticeable difference in kidney IgM deposition, there was noticeable decrease in IgG Ab deposition in the kidneys of A10Blpr mice. (Fig. 4D, 4E). Additionally, CD138+ cells in A10Blpr mice were significantly decreased in both percentage and absolute number in the cervLN (Fig. 4F–H) as well as in the spleen (Fig. 4I–K).
Loss of B cell ADAM10 in lpr mice decreases autoantibody production. (A) Serum total Ab levels from 6-mo-old mice as determined by fluorescence-linked immunoabsorbant assay. Serum anti-dsDNA IgM (B) and IgG (C) levels as determined by fluorescence-linked immunoabsorbant assay from mice based on age. Immunofluorescent microscopy of IgM (D) and IgG (E) complex deposition in kidney glomeruli of 6-mo-old mice. (F) Representative pseudocolor dot plots of cervLN CD19+ B220+ B cells gating for CD138+ B cells. Number of plots indicates mean percentage of CD138+ B cells of total B cells. Percentage (G) and absolute number (H) of CD138+ B cells in the cervLN of indicated 6-mo-old mice. (I) Representative pseudocolor dot plots of spleen CD19+ B220+ B cells gating for CD138+ B cells. Number of plots indicates mean percentage of CD138+ of total B cells. Percentage (J) and absolute number (K) of CD138+ B cells in the spleen of indicated 6-mo-old mice. One-way ANOVA with Tukey posttest (A, B, C, G, H, J, and K). Data are pooled from three (A, G, H, J, and K; mean) and three (B and C; mean ± SEM) independent experiments. n.s., not significant (p ≥ 0.05), *p < 0.05, **p < 0.01, ***p < 0.001.
Loss of B cell ADAM10 in lpr mice decreases autoantibody production. (A) Serum total Ab levels from 6-mo-old mice as determined by fluorescence-linked immunoabsorbant assay. Serum anti-dsDNA IgM (B) and IgG (C) levels as determined by fluorescence-linked immunoabsorbant assay from mice based on age. Immunofluorescent microscopy of IgM (D) and IgG (E) complex deposition in kidney glomeruli of 6-mo-old mice. (F) Representative pseudocolor dot plots of cervLN CD19+ B220+ B cells gating for CD138+ B cells. Number of plots indicates mean percentage of CD138+ B cells of total B cells. Percentage (G) and absolute number (H) of CD138+ B cells in the cervLN of indicated 6-mo-old mice. (I) Representative pseudocolor dot plots of spleen CD19+ B220+ B cells gating for CD138+ B cells. Number of plots indicates mean percentage of CD138+ of total B cells. Percentage (J) and absolute number (K) of CD138+ B cells in the spleen of indicated 6-mo-old mice. One-way ANOVA with Tukey posttest (A, B, C, G, H, J, and K). Data are pooled from three (A, G, H, J, and K; mean) and three (B and C; mean ± SEM) independent experiments. n.s., not significant (p ≥ 0.05), *p < 0.05, **p < 0.01, ***p < 0.001.
Increased T cell numbers alter regulation of ICOSL by ADAM10
Overall, the previously mentioned data indicates a strong modulation to the lpr phenotype following loss of B cell ADAM10. As shown in Fig. 3A and 3B, cells from 6-mo-old A10Blpr mice had significantly higher ICOSL levels compared with B cells from B6lpr mice; however, the ICOSL on B cells from A10Blpr mice was decreased compared with age-matched A10B mice. Examination of ICOSL levels at different ages in A10Blpr mice revealed a steady decrease in ICOSL levels with increasing age (Fig. 5A). This decrease in ICOSL levels was also seen in B6lpr mice, albeit to a much lesser extent and at a younger age (Fig. 5A). There was a drastic drop in ICOSL levels seen between 4 and 5 mo in A10Blpr mice, which coincided with T cell ICOS levels greatly increasing during this same time period (Fig. 5A, 5B). This is further seen when examining the correlation between decreasing ICOSL levels and increasing ICOS levels (Supplemental Fig. 4A). The increasing ICOS levels correlate with increasing T cell numbers in the cervLN (Fig. 5C). There was a striking difference in ICOS:ICOSL ratios between B6lpr and A10Blpr mice that continued with age but did narrow between 4 and 5 mo (Fig. 5D). Interestingly, this increase in ICOS:ICOSL ratios between 4 and 5 mo of age in A10Blpr mice (Fig. 5D) overlaps with the age in which lymphoproliferation and increased cellularity began to occur in these mice (Fig. 5E) as well as TFH accumulation (Fig. 5F).
Elevated ICOS correlates with disease progression in lpr mice. (A) ICOSL MFI of B220+ CD19+ B cells as determined by flow cytometry versus age. (B) ICOS MFI of CD3+ CD4+ T cells as determined by flow cytometry versus age. (C) ICOS MFI of CD3+ CD4+ CD44+ CD62L− Teff plots against absolute CD3+ cell number in the cervLN versus age. (D) ICOS MFI to ICOSL MFI ratio plotted as a function of age. (E) CD45+ cell count in the cervLNs of indicated groups as a function of age. (F) CD4+ TFH number in the cervLN of indicated groups as a function of age. One-way ANOVA with Tukey posttest (A, B, D, E, and F). Data are pooled from three (A, B, D, E, and F; mean ± SEM) independent experiments. n.s., not significant (p ≥ 0.05), *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. MFI, mean fluorescence intensity.
Elevated ICOS correlates with disease progression in lpr mice. (A) ICOSL MFI of B220+ CD19+ B cells as determined by flow cytometry versus age. (B) ICOS MFI of CD3+ CD4+ T cells as determined by flow cytometry versus age. (C) ICOS MFI of CD3+ CD4+ CD44+ CD62L− Teff plots against absolute CD3+ cell number in the cervLN versus age. (D) ICOS MFI to ICOSL MFI ratio plotted as a function of age. (E) CD45+ cell count in the cervLNs of indicated groups as a function of age. (F) CD4+ TFH number in the cervLN of indicated groups as a function of age. One-way ANOVA with Tukey posttest (A, B, D, E, and F). Data are pooled from three (A, B, D, E, and F; mean ± SEM) independent experiments. n.s., not significant (p ≥ 0.05), *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. MFI, mean fluorescence intensity.
Discussion
We have previously shown that ADAM10 is responsible for proteolytic ICOSL regulation on B cells (19). Loss of A10B results in elevated ICOSL levels and a compensatory posttranslational decrease in T cell ICOS. This dysregulation of the ICOS:ICOSL axis resulted in almost complete loss of TFH and affinity-matured Ab production in T-dependent immunization models (19, 23). This study built on those findings and examined the ability of ADAM10 to regulate the ICOS:ICOSL axis in two separate autoimmune models in which ICOS is important for pathogenesis.
In this study, we first assessed whether B cell ADAM10 was still able to regulate the ICOS:ICOSL axis in a model known to have elevated ICOS levels due to increased transcriptional levels. B6mir146a−/− mice have been shown to have elevated ICOS levels on resting T cells as well as increased TFH numbers and TFH ICOS levels following immunization (9). Mir146a was shown to regulate ICOS transcriptional levels in cooperation with Argonaute2 and Roquin, and its loss resulted in elevated ICOS transcriptional levels (9, 25). The loss of A10B in B6mir146a−/− mice resulted in decreased ICOS on T cells and decreased TFH and GC responses, completely reversing the phenotype seen in B6mir146a−/− mice. These results further support the importance of posttranslational regulation of ICOSL and ICOS by ADAM10-mediated catabolism of ICOSL.
A number of studies have shown an important role for both ICOS and ICOSL in murine lupus models (10–14). Several reports point to ICOS being essential for the development of autoantibody production while being dispensable for the accumulation of Th17 and Th1 effector cells (10, 11, 13). In A10Blpr mice, we did not see a difference in Th1 cells in either the spleen of cervLN (Supplemental Fig. 3). Other groups have shown that ICOS stimulation is essential for T cell–mediated interstitial nephritis in MRLlpr mice, which is not seen in the B6 version of this model (12, 24). However, we did see decreased IgG deposition in the glomeruli of A10Blpr mice (Fig. 4E). Studies using conditional ICOSL knockout mice have shown that B cell ICOSL is not fully necessary for autoantibody production in MRLlpr mice and is also dispensable for the accumulation of IL-21–producing effector helper T cells (Tefh) in the kidneys and spleens (12). In addition, loss of ICOSL on dendritic cells also did not affect autoantibody production but did cause a reduction in Tefh accumulation in the kidneys as well as decreased kidney pathology (12). Other groups have shown that ICOS−/− mice have a loss of Teff in the kidney when crossed to MRLlpr mice (10, 11, 14). In our study, we did not see a difference in kidney T cell infiltration (Fig. 2C).
Our ADAM10 B cell conditional knockout model examines the role of ICOS and ICOSL in the Faslpr model in a different context than genetic knockouts of either protein. Although our previous results have indicated an important role for B cell ADAM10 in the regulation of ICOSL levels (19), aberrant regulation of ICOS and ICOSL in A10Blpr mice was still seen. With increasing age, we observed an increase in T cell surface ICOS and a corresponding decrease in B cell surface ICOSL. In B6lpr mice, this decrease in ICOSL and increase in ICOS occurs between 2 and 3 mo, whereas in A10Blpr mice, it occurs between 4 and 5 mo (Fig. 5A, 5B) and can be seen more drastically when examining the ICOS:ICOSL ratio (Fig. 5D). Intriguingly, this increase in the ICOS:ICOSL ratio coincides with the first signs of lymphoproliferation in these mice (Fig. 5E). With these findings, we propose a model in which disease progression in lpr mice begins with the accumulation of T cells due to loss of FAS-mediated cell death. With this increase in T cell numbers, we see a decrease in ICOSL levels. This ultimately allows increased T cell accumulation of ICOS, which eventually leads to increased T cell activation, proliferation, and the ability to provide B cell help. This B cell help then allows for increased Ab and autoantibody production.
ICOS and ICOSL interaction have been shown to be necessary for class-switched Ab responses during T-dependent immunization models (2). Mice that lack either ICOS or ICOSL have drastically decreased class-switched Ab. In our model, A10Blpr mice have decreased total IgG1 and IgG2a at 6 mo compared with B6lpr mice. We did not see a difference in IgM in A10Blpr mice at this timepoint, which is not surprising as IgM is not a class-switched Ab. However, we did see a difference in anti-dsDNA IgM in A10Blpr mice compared with B6lpr mice, suggesting that A10Blpr mice have decreased loss of tolerance compared with B6lpr mice. We saw a significant difference in anti-dsDNA IgG at both 6 and 8 mo of age, further supporting the role for ADAM10 in regulating the ICOS:ICOSL axis in the lpr model. We were not able to examine the mice past 8 mo of age due to increased mortality in B6lpr mice.
We hypothesize that early T cell lymphoproliferation in B6lpr mice results in more interaction with B cell ICOSL, resulting in ICOSL shedding and ultimately decreasing B cell ICOSL levels. These decreased ICOSL levels are then responsible for the increased T cell ICOS levels seen, as we have previously shown that interaction of ICOS with ICOSL results in decreased ICOS levels through internalization of surface ICOS (19), further supporting this is the finding that ICOSL−/− mice have elevated T cell ICOS levels (26). We propose that T cells are able to be activated more extensively and differentiate to TFH and Tefh once their ICOS levels increase. ICOS stimulation for these cells could likely be coming from other APCs, such as dendritic cells.
An interesting finding in this study was that that 6-mo-old A10Blpr mice had decreased B cell ICOSL levels compared with age-matched A10B controls (Fig. 2A). There are several possibilities to explain this finding. Although A10B mice have elevated ICOSL protein levels compared with wild-type mice, the transcriptional level of Icosl is greatly reduced (19), suggesting that excessive interaction of ICOS and ICOSL signals B cells to further downregulate Icosl transcription, ultimately resulting in decreased ICOSL surface expression. It is also possible that with the rapid lymphoproliferation seen in this model, we are losing penetrance of the Cre transgene in A10Blpr mice, which could explain why the mice begin to develop lymphoproliferation and autoantibody production at later timepoints. However, if this were the case, we would expect to see a bimodal pattern of ICOSL expression on the surface of B cells in A10Blpr mice, but that is not the case.
Another possibility for the decreased ICOSL levels in A10Blpr mice is that a different protease, most likely ADAM17, is able to cleave ICOSL in the context of autoimmunity. A10B mice have been shown to have elevated ADAM17 levels, which may allow for more shedding of ICOSL (27). We, and others, have shown that ADAM17 has the ability to shed ICOSL in response to several stimuli, most notably PMA stimulation (19, 28). Mice that lack both ADAM10 and ADAM17 on B cells have even higher levels of ICOSL compared with just the loss of ADAM10 (19). However, this seems to only be the case in conjunction with the loss of ADAM10, as mice deficient in ADAM17 on B cells do not have elevated ICOSL levels compared with wild-type mice (19). The ability of ADAM17 to control ICOSL in the absence of ADAM10 in the lpr model warrants further investigation, and we hypothesize that loss of ADAM17 in addition to A10B in lpr mice would further protect from autoantibody development.
Because of limitations of this study, we cannot exclude the possibility that other substrates of ADAM10 may account for some of the phenotype seen in A10Blpr mice. ADAM10 is known to shed a wide range of substrates, including chemokine receptors, as well as participate in Notch signaling (20, 29). We have previously reported that in A10B mice, reversing the dysregulation of ICOSL and ICOS by blocking excess ICOSL interaction with ICOS was able to restore humoral responses following a T-dependent immunization (19). However, the correlation we see with decreased ICOSL and increased ICOS certainly implicates this change as being relevant to the increased autoimmmunity that is seen.
Lower ICOS levels were observed on TFH present in A10Blpr mice, suggesting they may have decreased ability to fully offer B cell help for Ab production, particularly when isotype switching is involved (13, 30, 31). Several reports have suggested the source of autoantibody production is extrafollicular (14, 18) in the lpr model, so we cannot conclusively determine that the decrease in TFH is directly responsible for decreased autoantibody production, at least with respect to this occurring in GCs. However, all CD4+ T cells in A10Blpr mice had decreased ICOS levels compared with B6lpr mice. These extrafollicular helper T cells may also have decreased ability to aid in extrafollicular autoantibody production, particularly in the context of IL-21 and CD40L, which are reliant on ICOS stimulation (15, 32, 33). This is further supported with the significant decrease seen in anti-dsDNA IgM and IgG levels at 8 mo.
Overall, these results show that indirectly regulating ICOS by blocking ICOSL catabolism is a potent way to control humoral immunity, even in a severe autoimmune model. Additionally, the A10Blpr mice offer a model to study the role of ICOSL and ICOS in the development of lymphoproliferation and autoantibody production in lpr mice, without genetic deletion of either ICOS or ICOSL. Additionally, our results further exemplify the importance of tight regulation of this system to properly function and particularly the importance of ADAM10 in this regulation. However, because this model examines the genetic loss of ADAM10 on B cells before disease onset, we cannot determine whether blocking ADAM10 following onset of disease would be beneficial to reverse disease pathologies. We would exercise caution in the use of ADAM10 inhibitors for the treatment of established autoimmunity, as we cannot preclude the possibility that inhibiting ICOSL shedding through ADAM10 inhibition may actually further exacerbate disease severity when ICOS levels are already elevated.
The results further our understanding of ICOS:ICOSL regulation in the context of autoimmunity as well as other models of increased humoral immune responses. Our results suggest that proteolytic regulation of ICOSL on B cells by ADAM10 is able to extensively impact the ICOS- and ICOSL-dependent responses. These findings support the role for ICOSL catabolism in controlling ICOS levels and T cell activation, particularly in the context of autoimmunity reliant on ICOS stimulation. Additionally, this work suggests that measuring ICOS:ICOSL ratios may be a method to monitor disease progression and severity in autoimmune models reliant on these costimulatory molecules. Overall, altering the proteolytic regulation of ICOSL by ADAM10 may be a way to modulate the immune response, particularly in the context of humoral autoimmunity.
Acknowledgements
We thank Matthew Zellner for help in mouse colony management.
Footnotes
This work was supported by National Institutes of Health (NIH)/National Institute of Allergy and Infectious Diseases R01AI18697A1 (to D.H.C.). Microscopy was performed at the Virginia Commonwealth University (VCU) Microscopy Facility, and flow cytometry used the VCU Flow Cytometry Core. Both cores are supported in part by funding from NIH–National Cancer Institute Cancer Center Support Grant P30 CA016059.
The online version of this article contains supplemental material.
Abbreviations used in this article:
- ADAM10
a disintegrin and metalloproteinase 10
- B6
C57BL/6J
- cervLN
cervical lymph node
- GC
germinal center
- ICOSL
ligand for ICOS
- NP31-KLH
4-hydroxy-3-nitrophenylacetyl hapten conjugated to keyhole limpet hemocyanin
- popLN
popliteal lymph node
- T-dependent
thymus-dependent
- Teff
T effector cell
- Tefh
effector helper T cell
- Tem
T effector memory cell
- TFH
follicular helper T cell
- WT
wild-type.
References
Disclosures
The authors have no financial conflicts of interest.