In vitro activity and function of B7-H4-Ig fusion protein

Abstract

B7-H4 has been shown to inhibit T cell proliferation, cytokine production and cell cycle in vitro. B7-H4 deficient mice develop exacerbated disease in the mouse models of Rheumatoid Arthritis (RA), Type 1 Diabetes (T1D) and Experimental Autoimmune Encephalomyelitis (EAE). On the other hand, B7-H4-Ig fusion protein has been documented to assuage the symptoms in mouse models of RA, T1D, and multiple sclerosis in vivo. In the present study, B7-H4-Ig bound to the majority of human peripheral blood monocytes and NK cells, but not to either normal or activated T cells. B7-H4-Ig fusion protein was assayed for its effects in allogeneic mixed lymphocyte culture (MLC) systems. Soluble B7- H4-Ig had no significant effect in the MLC, but with a tendency to promote allogeneic response. Immobilized, but not soluble B7-H4-Ig inhibited plastic bound anti-CD3 mediated activation of T cells. This inhibition however was largely due to B7-H4-Ig mediated displacement of anti-CD3 antibody from the plastic plate. Finally, B7-H4-Ig had no effect on the cytotoxicity mediated by NK and LAK cells in PBMC. Our findings thus caution against the interpretation of suppressive effect observed solely in plate-bound anti-CD3 mediated T cell co-stimulation in vitro.

Share and Cite:

Rasmussen, S. , Kosicki, M. , Svendsen, S. , Claesson, M. and Kristensen, N. (2013) In vitro activity and function of B7-H4-Ig fusion protein. Open Journal of Immunology, 3, 24-32. doi: 10.4236/oji.2013.31004.

1. INTRODUCTION

The B7 family member B7-H4, discovered ten years ago, has been shown to inhibit TCR-mediated proliferation, cell-cycle progression and IL-2 production by CD4+ and CD8+ T cells in murine [1-3] and human [4]

in vitro systems. Regulatory T cells (Treg) have been shown to induce IL-10 secretion and upregulation of B7-H4 in APCs [5]. B7-H4 mRNA is broadly expressed in many tissue and cell types, including tumors [6]. B7-H4 protein , however, is limited to activated T cells, B cells, monocytes, dendritic cells [2] activated hepatic stellar cells, synovia cells in rheumatoid arthritis, islet beta cell, renal tubular and lung epithelial cells [7,8]. A number of cancers have been shown to over-express B7-H4 protein, including human ovarian, breast, prostate and lung cancer, renal-cell carcinoma and uterine adenocarcinoma [9]. In ovarian carcinoma, B7-H4 expressing tumor-associated macrophages inhibit Tumor Associated Antigen (TAA)-specific T-cell effector function [10]. B7-H4 blockade with a monoclonal antibody in the EAE mouse model led to exacerbated disease and to an accumulation of CD8 + T cells and macrophages in the brain [1]. In vivo administration of B7-H4Ig in a murine graft-versus-host disease model reduced both T cell proliferation, CTL activity and improved animal survival [2]. A soluble form of B7-H4 has been implicated in pathogenesis of rheumatoid arthritis and genetic deletion of B7-H4 accelerated the progression of collagen-induced arthritis in mice [11]. B7-H4 knockout mice suffer from a more severe experimental diabetes due to increased islet infiltration of Th1 and Th17 cells, while mice overexpressing B7-H4 were protected from diabetes by reducing IFN-production in CD4 T cells, without skewing towards Th2 phenotype or activation of Tregs [12]. Treating NOD mice with B7-H4-Ig resulted in reduced incidence of diabetes through a transient increase in Tregs number and suppressive activity [13]. The receptor for B7-H4 has yet to be identified, but is believed to be expressed on subsets of immune cells, including activated T cells and neutrophils. The B7-H4 receptor most probably belongs to an unknown member of the co-inhibitory CD28 superfamily that includes BTLA, CTLA-4 and PD-1 [14].

In the present paper, we studied a recently developed recombinant human B7-H4-Ig fusion protein, B7-H4-Ig (provided by AmplimmunInc.U.S.A.) in human in vitro systems. Binding of B7-H4-Ig to normal and activated human peripheral blood mononuclear cells (PBMC) was examined. Whereas the majority of monocytes and NK cells were shown to bind B7-H4-Ig, less than 10% of normal B cells and neither naive, nor activated T cells were found to bind B7-H4-Ig. In a series of allogeneic mixed lymphocyte culture (MLC) experiments, B7-H4-I g showed a slight, but insignificant enhancing effect on proliferation. An inhibitory effect of B7-H4-Ig on immobilized anti-CD3 induced T cell proliferation was observed, but proved to be caused by B7-H4-Ig mediated displacement of anti-CD3 antibody from the culture well. Finally, neither the activity of NK nor LAK cells was influenced by exposure to B7-H4-Ig.

2. MATERIALS AND METHODS

2.1. Antibodies and Control Reagents

Anti-CD3 antibody (F101.01) recognizes a conformational determinant on the TcR/CD3 complex [15]. The antibody was a kind gift from Professor C. Geisler, University of Copenhagen and was used for T cell activation. The following mAbs were purchased from Becton-Dicki nson,New Jersey, U.S.A. as mouse anti-human Abs: FITC-conjugated anti-CD3; APC-conjugated anti -CD8, anti-CD14, and anti-CD56. Mouse anti-human FITCconjugated anti-CD4 and APC-conjugated anti C-D19 an tibodies were purchased from Elektra and PE-con jugated mouse anti-human B7-H4 (clone H74) was purchased from eBioscience (California, U.S.A.). B7 -H4-Ig (human B7-H4 extracellular domain fused with human IgG1) fusion proteins were produced in suspension culture in an animal protein-free-adapted CHOK- 1SV (Lon za Biologics, Allendale, NJ) cell line utilizing the glutamine synthetase gene expression system, and purified using Amplimmune’s protein purification process. KS is an inactive mutant of B7-DC-Ig (provided by Amplimmune Inc.), a fusion protein consisting of the Fc portion of human IgG1 protein and extracellular domain of B7-DC, in which a missense mutation in the active site changed lysine residue at position 113 to serine. The KS mutant was used as a negative, isotype control for B7- H4-Ig. Synagis (also known as palivizumab) was used as another negative isotype control for B7-H4-Ig, as well as a Fc-blocker for flow cytometry experiments. Synagis is a humanized (IgG1 Fc domain) monoclonal antibody against respiratory syncytial virus.

2.2. Cell Cultures

Human PBMCs were obtained from buffy coats of healthy donors in Danish Blood Donor Corps by Ficoll gradient centrifugation and stored at −140˚C. Thawing of the cells followed a standard protocol. Cells were cultured on 96-well round (MLCs) or flat-bottomed plates (anti-CD3 stimulated T cells) in 220 μl of RPMI-1640 medium supplied with Glutamax, 10% human serum (Valley Biomedical), 0.5% 2-mercaptoethanol and 0.5% penicillin/streptomycin. All cultures were conducted with a total of 0.2 million cells per well in an incubator at 37˚C, 5% CO2. One-way MLCswere prepared by irradiating one set of cells (“stimulator”) with 2000 rad from Cs-137 for 10 minutes. CD8 T cells were depleted using Dynabeads-CD8 (Invitrogen) following manufacturer’s manual. T cells were enriched 2-3 fold by two-step nonadherence to plastic. All cells were cultured for 72 hours and their proliferation assessed by tritium thymidine incorporation assay.

Immobilization of B7-H4-Ig, KS, anti-CD3 antibody and Synagis or a mixture of them was carried out with 40 μl of the agent incubated in flat-bottomed plates overnight at 4˚C and washed twice. In case of experiments shown in Figures 5-8 the indicated agent (B7-H4-Ig, KS or Synagis) was either added as “immobilized” (incubated overnight together with anti-CD3) or “soluble” (added together with the cells after the plate had been washed). In experiments shown in Figures 9 and 10 the plate was washed and B7-H4-Ig was added at 40 μl for two hours at 37˚C followed by washing and plating of cells essentially as described by Sica et al. [2].

2.3. Flow Cytometry

Cells for FACS analysis were incubated with 300 μg/ml Synagis for 15 min on ice (to block Fc receptors) followed by incubation for 1h on ice with either 100 μg/ml of B7-H4-Ig or PBS. After wash 1 x 106 Cells were stained with fluorochrome labeled antibodies containing 1% bovine serum albumin (BSA) in PBS for 20 minutes on ice. After washing twice, cells were fixed with 1% paraformaldehyde. Isotype control antibodies were included to define appropriate cut-off levels. For labelling of non-adherent, anti-CD3 activated T cells, these were incubated for 15 minutes with 300 μg/ml Synagis, washed, and incubated for 1 hour with 200 μg/ml B7-H4-Ig or PBS, then washed and incubated for 30 minutes in FITC-conjugated mouse anti-CD3 (BD Pharmingen) and PE-conjugated mouse anti-B7-H4 antibodies (eBioscience,clone H74), in PBS supplied with 0,5% BSA. Cells were then washed, resuspended in either FACS buffer or 1% paraformaldehyde and analysed using a FACScalibur flow cytometer (Becton-Dickinson). Data were analysed using cellquest software (BectonDickinson).

2.4. Simultaneous ELISA and Proliferation Assays

Two plates were incubated with a range of anti-CD3 concentrations overnight at 4˚C. They were washed twice with PBS, incubated with 40 μl of 50 μg/ml of B7-H4-Ig or PBS for 2 hours at 37˚C and washed twice with PBS. ELISA assay for anti-CD3 binding was performed on the first plate. To the second plate enriched human T cells were added and incubated at 37˚C/5% CO2 for 3 days. Tritium thymidine incorporation was performed to assess the proliferation.

2.5. 51Cr-Release Assay

Freshly harvested PBMCs from healthy donors were washed twice and resuspended in medium containing 10% FCS, at 5 × 106 cells/ml and directly tested for cytotoxicity or incubated for 24 h min at 37˚C in the atmosphere of 5% CO2 in air in the presence of a stimulatingcytokine cocktailof (IL-12 and IL-15: 50 ng/ml, IL-2: 2000 U/ml and IFN-α: 10.000 U/ml) (ImmunoTools GmbH; Germany) to generate LAK cells. B7-H4 -Ig was added to the effector cells at 50µg/ml and the formulation buffer and PBS was added as controls.K562 and Daudi cells were used as targets for NK and LAK respectively. Target cells were labeled with 30μCi/mil of 51Cr (Perkin Elmer, Billerica, MA) for 1 h at 37˚C. The labeled cells were washed and resuspended in medium. Cells were co-incubated at effector to target (E:T) ratios at 100:1 - 12.5:1. Co-cultures were set up in 96 well V-bottom plates (Nunc; Denmark) in four replicates and incubated for 4h at 37˚C in the atmosphere of 5% CO2 in air. Controls included target cells incubated in medium alone for spontaneous release or lysed with 10% triton X-100 for maximal release. Radioactivity was measured by Wallac Wizard 1470 Automatic Gamma Counter. Specific release was calculated using the following formula:

% specific lysis = (sample cpm − spontaneous cpm)/ (maximal cpm − spontaneous cpm) × 100%.

2.6. Data Treatment and Statistics

Data were analysed in R, using ggplot2 package for visualization. Statistical significance was assessed by onesample t-test on medium control normalized data. A posthoc Benjamini-Hochberg correction was employed to control for multiple testing. The results of simultaneous ELISA and proliferation assays were analysed by fitting a four-point logistic model over a range of anti-CD3 concentrations and response values (c.p.m. and OD values in proliferation and ELISA assays, respectively) in control samples. Using this model anti-CD3 concentrations corresponding to response values in B7-H4-Ig treated samples were reversely predicted. Ratio of predicted and originally applied anti-CD3 antibody concentrations was calculated (equalling 1 for control samples) and plotted.

Figure 1. The frequencies of T cells (CD3+), B cells (CD19+), NK cells (CD56+) and monocytes (CD14+) in the responder cell populations day 0 and day 3 in an allogeneic two-way MLC.

3. RESULTS

3.1. FACS Gating Strategy

Figure 1 shows the FACS gating strategy for PBMC. The frequencies of T cells (CD3+), B cells (CD19+), NK cells (CD56+) and monocytes (CD14+) are shown in the FACS plot of normal PBMC and day 3 responder cells in an allogeneic two-way MLC stained with cell type specific antibodies. As shown, the frequency of the four major cell types changes only slightly during the three days of MLC. Numbers are the percent of labeled cells in the respective gates.

Conflicts of Interest

The authors declare no conflicts of interest.

References

[1] Prasad, D.V.R., Richards, S., Mai, X.M. and Dong, C. (2003) B7S1, a novel B7 family member that negatively regulates T cell activation. Immunity, 18, 863-873. doi:10.1016/S1074-7613(03)00147-X
[2] Sica, G.L., Choi, I.H., Zhu, G.F., Tamada, et al. (2003) B7-H4, a molecule of the B7 family, negatively regulates T cell immunity. Immunity, 18, 849-861. doi:10.1016/S1074-7613(03)00152-3
[3] Zang, X., Loke, P., Kim, J., Murphy, K., Waitz, R. and Allison, J.P. (2003) B7x: A widely expressed B7 family member that inhibits T cell activation. Proceedings of the National Academy of Sciences, 100, 10388-10392. doi:10.1073/pnas.1434299100
[4] Mao, Y.X., Chen, Y.J., Ge, Y., Ma, H.B., et al. (2006) Recombinant human B7-H4 expressed in Escherichia coli inhibits T lymphocyte proliferation and IL-2 secretion in vitro. Acta pharmacologica Sinica, 27, 741-746. doi:10.1111/j.1745-7254.2006.00338.x
[5] Kryczek, I., Wei, S., Zou, L., Zhu, G., et al. (2006) Cutting edge: Induction of B7-H4 on APCs through IL-10: Novel suppressive mode for regulatory T cells. Journal of Immunology, 177, 40-44.
[6] Choi, I.H., Zhu, G., Sica, G.L., Strome, S.E., Cheville, J.C., Lau, J.S., Zhu, Y., Flies, D. B., Tamada, K. and Chen, L. (2003) Genomic organization and expression analysis of B7-H4, an immune inhibitory molecule of the B7 family. Journal of Immunology, 171, 4650-4654.
[7] Chinnadurai, R. and Grakoui, A. (2010) B7-H4 mediates inhibition of T cell responses by activated murine hepatic stellate cells. Hepatology, 52, 2177-2185. doi:10.1002/hep.23953
[8] Chen, Y., Yang, C., Xie, Z., Zou, L., Ruan, Z., Zhang, X., Tang, Y., Fei, L., Jia, Z. and Wu, Y. (2006) Expression of the novel co-stimulatory molecule B7-H4 by renal tubular epithelial cells. Kidney International, 70, 2092-2099.
[9] Zou, W. and Chen, L. (2008) Inhibitory B7-family molecules in the tumour microenvironment. Nature Reviews, Immunology, 8, 467-477. doi:10.1038/nri2326
[10] Kryczek I., Zou L., Rodriguez P., Zhu, G. et al. (2006) B7-H4 expression identifies a novel suppressive macrophage population in human ovarian carcinoma. Journal Experimental Medicine, 203, 871-881. doi:10.1084/jem.20050930
[11] Azuma T., Zhu G.F., Xu H.Y., Rietz A.C., et al. (2009) Potential Role of Decoy B7-H4 in the Pathogenesis of Rheumatoid Arthritis: A Mouse Model Informed by Clinical Data. Plos Medicine, 6, Article ID: e1000166. doi:10.1371/journal.pmed.1000166
[12] Wei J., Loke P., Zang X. and Allison J.P. (2011) Tissue-specific expression of B7x protects from CD4 T cell-mediated autoimmunity. Journal Experimental Medicine, 208, 1683-1694. doi:10.1084/jem.20100639
[13] Wang X. Hao J., Metzger D.L., Mui A., et al. (2011) Early treatment of NOD mice with B7-H4 reduces the incidence of autoimmune diabetes. Diabetes, 60, 3246-3255. doi:10.2337/db11-0375
[14] Chen L. (2004) Co-inhibitory molecules of the B7-CD28 family in the control of T-cell immunity. Nature Reviews Immunology, 4, 336-347. doi:10.1038/nri1349
[15] Geisler C., Plesner T., Pallesen G., Skj?dt et al. (1988) Characterization and expression of the human T cell receptor-T3 complex by monoclonal antibody F101.01. Scandinavian Journal of Immunology, 27, 685-696. doi:10.1111/j.1365-3083.1988.tb02402.x
[16] Wang X., Hao J., Metzger D.L., Ao Z., et al. (2012) B7-H4 treatment of T cells inhibits ERK, JNK, p38, and AKT Activation. PloS One, 7, Article ID: e28232. doi:10.1371/journal.pone.0028232
[17] Suh W.K., Wang S., Duncan G.S., Miyazaki Y., Mak, T.W., et al. (2006) Generation and characterization of B7-H4/B7S1/ B7x-deficient mice. Molecular and Cellular Biology, 26, 6403-6411. doi:10.1128/MCB.00755-06

Copyright © 2024 by authors and Scientific Research Publishing Inc.

Creative Commons License

This work and the related PDF file are licensed under a Creative Commons Attribution 4.0 International License.