Abstract
Human leukocyte antigen-G is a non-classical major histocompatibility complex class I antigen with potent immune-inhibitory function. Human leukocyte antigen-G benefit patients in allotransplantation and autoimmune diseases by interacting with its receptors, immunoglobulin-like transcripts. Here we observed significantly less human leukocyte antigen-G in plasma from immune thrombocytopenia patients positive for anti-platelet autoantibodies compared with autoantibodies-negative patients or healthy controls. Besides, human leukocyte antigen-G is positively correlated with platelet counts in both patients and healthy controls. We also found less membrane-bound human leukocyte antigen-G and immunoglobulin-like transcripts on CD4+ and CD14+ cells in patients. Recombinant human leukocyte antigen-G upregulated immunoglobulin-like transcript 2 expression on CD4+ and immunoglobulin-like transcript 4 on CD14+ cells. Human leukocyte antigen-G upregulated IL-4 and IL-10, and downregulated tumor necrosis factor-α, IL-12 and IL-17 secreted by patient peripheral blood mononuclear cells, suggesting a stimulation of Th2 differentiation and downregulation of Th1 and Th17 immune response. Human leukocyte antigen-G-modulated dendritic cells from immune thrombocytopenia patients showed decreased expression of CD80 and CD86, and suppressed CD4+ T-cell proliferation compared to unmodulated cells. Moreover, human leukocyte antigen-G modulated cells from patients induced less platelet apoptosis. Human leukocyte antigen-G administration also significantly alleviated thrombocytopenia in a murine model of ITP. In conclusion, our data demonstrated that impaired expression of human leukocyte antigen-G and immunoglobulin-like transcripts is involved in the pathogenesis of immune thrombocytopenia; Recombinant human leukocyte antigen-G can correct this abnormality via upregulation of immunoglobulin-like transcripts, indicating that human leukocyte antigen-G can be a diagnostic marker and a therapeutic option for immune thrombocytopenia.Introduction
Immune thrombocytopenia (ITP) is a common autoimmune bleeding disorder with decreased platelet counts.1-4 The therapeutic regimens for ITP patients, including glucocorticosteroids, intravenous immunoglobulin, thrombopoietin receptor agonists, rituximab and other immunoregulatory drugs, are not always effective, and only about one-third of ITP patients can achieve longterm remission.5-7 Autoantibodies targeting platelets enhanced T-helper cell activity, initiated abnormal activation of cytotoxic T lymphocytes and disturbed cytokine profiles to accelerate platelet clearance and suppress platelet production in ITP patients.1-4,8 Moreover, the deficiency and dysfunction of immunoregulatory cells (i.e., regulatory T cells [Tregs]),9-11 tolerogenic dendritic cells12 and myeloid-derived suppressor cells13 also contribute to the loss of immune tolerance in ITP.
HLA-G is a non-classical major histocompatibility complex class I antigen found to be expressed in placental trophoblast cells in 1990s.14 There are four membrane-bound (mbHLA-G1 to -G4) and three soluble (sHLA-G5 to -G7) isoforms generated by alternative splicing of the primary transcript.15 It was initially believed that HLA-G in placental cells plays an important role in maternal-fetal immunotolerance by interacting with inhibitory receptors (immunoglobulin-like transcript 2 [ILT-2/LILRB1/CD85j], immunoglobulin-like transcript 4 [ILT-4/LILRB2/CD85d] and KIR2DL4 receptor) on leukocytes, which leads to CTL apoptosis, natural killer (NK) cells immobilization, mononuclear cells inhibition and suppressive cytokine environment.16,17 Subsequently HLA-G was found not be restricted to placenta cells, but it is also expressed in human peripheral blood mononuclear cell (PBMC) and plasma.18,19 Additionally to the immune inhibitory capability mentioned above, HLA-G-ILT2/ILT4 interaction suppressed lymphocyte proliferation20,21 and inhibited antibody secretion of activated B cells.21 Ideas that HLA-G protein facilitates allotransplantation by preventing immune rejection via these actions on immune cells was established. After liver and kidney transplantation, an increased HLA-G level was detected in patients with a reduced incidence of rejection.22-24
Recently, HLA-G-induced immune tolerance has been intensively investigated. The expression of HLA-G (and its cognate receptors, ILT) was correlated with immune escape, which suggests unfavorable outcomes in cancer patients.25,26 On the other hand, HLA-G-induced tolerance can benefit patients with autoimmune diseases. Low soluble HLA-G (sHLA-G) level was found in patients with rheumatoid arthritis (RA) and multiple sclerosis, and was associated with disease progression in both cases.27,28 Moreover, the decrease of the number and/or function of ILT2 and ILT4 was involved in the pathogenesis of systemic lupus erythematosus (SLE).29,30 Another study by Kim et al.31 showed that functional HLA-G 14-bp polymorphism was associated with susceptibility to RA and SLE. However, the role of HLA-G in ITP remains obscure.
LeMaoult et al.32 reported that both membrane-bound HLA-G (mHLA-G) and secreted HLA-G5 (sHLA-G5) upregulated ILT2/ILT4 expression on APC and CD4+ T cells without the stimulation from exogenous antigens.32 This encouraged the development of recombinant human HLA-G protein (rhHLA-G). Further investigation demonstrated that in vitro synthetic HLA-G protein induced the tolerance of skin grafts in vivo successfully.33 Nevertheless, little has been defined about the effect of rhHLA-G in autoimmune disease.
In this study, we investigated whether there is aberrant HLA/ILT expression and their contribution to the loss of immune tolerance in ITP. Our data showed that the expression of HLA-G and ILT were both decreased in ITP patients, which may result in impaired function of PBMC and dendritic cells (DC). rhHLA-G upregulated ILT on CD4+ T cells and monocytes, and corrected dysfunction of immune cells. These findings indicate that HLA-G has the potential as a diagnostic marker and therapeutic option for ITP.
Methods
Patients and controls
Plasma samples from 50 newly diagnosed ITP patients were collected for the detection of sHLA-G with ELISA. Among them, PBMC from 17 ITP patients were isolated for the in vitro investigation of mHLA-G and ILT (Table 1-2). Samples from 15 healthy volunteers were used as controls. The diagnosis of ITP was based on established practice guidelines.1 Fourteen patients with autoantibody received standard ITP treatment high-dose dexamethasone (HD-DXM; 40 mg/day for 4 consecutive days), as described in the Online Supplementary Appendix and shown in Table 2. Enrollment took place between March 2013 and December 2018. The study was approved by the Medical Ethical Committee of Qilu Hospital, Shandong University. Informed consent was obtained from all patients before collecting samples in accordance with the Declaration of Helsinki.
Reagents
RhHLA-G (Abcam, Cambridge, UK) was dissolved in PBS to prepare a stock solution of 60 g/mL, and stored at -80 °C, according to the manufacturer’s instructions.
In vitro platelet apoptosis assay
PBMC were cultured with 600 ng/mL rhHLA-G for 3 days. Freshly isolated human platelets were adjusted to 1x108/mL with 5M prostaglandin E1. rhHLA-G-modulated PBMC (1×106) were then co-cultured with autologous or allogeneic platelets (1×107) as effector cells and target cells, respectively, and incubated at 37°C for 4 hours (h). Platelet apoptosis was detected using a mitochondrial membrane potential assay kit (Beyotime Biotechnology, Shanghai, China).
DC suppression assays
Freshly isolated CD4+ T cells were labeled with carboxyfluorescein diacetate succinimidyl ester (5 mM; Sigma-Aldrich, St. Louis, MO, USA) for 15 minutes (min) at 37°C and quenched by adding 3 mL of cold fetal calf serum (FCS) for 5 min at 4°C. CD4+ cells were washed with RPMI 1640 culture medium three times and seeded at 105 cells/well in U-bottom 96-well plates containing 1 mg/mL anti-human CD3 (Biolegend) and 50 U/mL recombinant human IL-2 (rhIL-2; R&D Systems) in triplicates. DC with or without rhHLA-G pre-modulation were added at a 5:1 ratio (T cells : DC). All cells were cultured for 5 days in humidified air with 5% CO2 at 37°C, then collected for flow cytometry analysis.
Animal model and the treatment of HLA-G
Blood was drawn by retro-orbital bleeding. Platelets from C57BL/6 wild-type mice were prepared to immunize C57BL/6 CD61 knockout (KO) mice by transfusion 10 mL of 108 platelets weekly for 4 consecutive weeks.34,35 The immunized CD61 KO mice were sacrificed, and their spleens were homogenized to prepare splenocyte suspension. On the day of transplantation, the C57BL/6 SCID mice were subjected to 200 cGy total body irradiation, and then injected with the indicated splenocytes (5×104) with or without HLA-G protein administration. Blood from saphenous veins was collected and platelet counts were detected weekly, as described in the Online Supplementary Appendix.34,35
Statistical analysis
Data are presented as mean ± standard deviation (SD). Differences between rhHLA-G modulated and unmodulated cells were determined by paired Student t tests. Correlative analysis was determined by nonparametric Pearson correlation. All the other statistics were analyzed with the Mann-Whitney U test. P<0.05 was considered a significant difference.
Results
HLA-G and ILT expression reduced in ITP patients
Platelet antibodies were identified by monoclonal antibody immobilization of platelet antigens (MAIPA) assay (shown in Table 1). In order to investigate the level of sHLA-G in ITP patients, platelet poor plasma (PPP) was isolated and sHLA-G was measured by enzyme linked immunosorbent assay (ELISA). Plasma HLA-G was reduced in ITP patients with anti-platelet antibodies compared to patients without autoantibodies and healthy controls. Among these patients, 11 were positive for anti- GPIIb/IIIa antibodies, 12 for anti-GPIb/IX antibodies, and 11 for both. However, no significant differences were found in the sHLA-G levels based on antibody type. ITP patients without anti-platelet antibodies showed no significant difference compared to healthy controls (Figure 1A-B). We then stratified ITP patients into platelet high (≥10×109/L) group and platelet low (<10×109/L) group and found that patients with high platelets showed significantly higher sHLA-G level compared to those with low platelets (Figure 1C). In order to assess whether the ITP group develops an increase in sHLA-G after normalization of their platelet counts due to standard ITP treatments, we tested sHLA-G level in 14 patients who received HD-DXM therapy. As shown in Table 2, 12 ITP patients responded to HD-DXM treatment and after treatment, the sHLA-G level in their plasma was significantly increased (Figure 1D). Thus, HLA-G might also be involved in the recovering process by HD-DXM treatments. Moreover, the level of sHLA-G positively correlated with platelet counts in patients with or without antiplatelet antibodies (r=0.602, P<0.01; r=0.584, P<0.05; Figure 1E-F). Interestingly, the sHLA-G level was also positively correlated with the platelet counts in healthy control (r=0.580, P<0.05, Figure 1G).
Cell surface expression of mHLA-G was evaluated as shown by flow cytometry. Expression of mHLA-G on CD4+ T cells and CD14+ monocytes was significantly lower in ITP patients than in healthy controls. However, CD8+ T cells and CD19+ B cells did not show a significant difference in the expression of mHLA-G between ITP patients and healthy controls (Figure 1H-K).
We then analyzed ILT2 expression on CD4+ T cells, CD8+ T cells, CD14+ monocytes, and CD19+ B cells, as well as ILT4 on CD14+ cells. Significantly lower ILT2 expression was found on CD4+ T cells from ITP patients compared with those from healthy controls. By contrast, no significant difference was found on CD8+, CD14+, or CD19+ cells. Decreased ILT4 expression was also observed on CD14+ cells from ITP patients (Figure 2). These data indicate aberrant expression of mHLA-G and ILT in ITP patients. However, no correlation was found between mHLA-G/ILT and platelet count in our study (Online Supplementary Figure S1). The expression of ILT2 on CD4+, CD8+, CD14+ or CD19+ cells and the expression of ILT4 on CD14+ cells showed no obvious changes after HDDXM treatment (Online Supplementary Figure S2).
rhHLA-G upregulated ILT expression on CD4+ and CD14+ cells
In order to test whether exposure to exogenous HLA-G would lead to an upregulation of inhibitory receptors on PBMC, we exposed these cells to rhHLA-G and monitored ILT2/ILT4 expression. rhHLA-G upregulated ILT2 expression on CD4+ T cells, as well as ILT4 on CD14+ monocytes in both ITP patients and controls. However, rhHLA-G had no significant effect on ILT2 expression on CD8+, CD14+, and CD19+ cells (Figure 2).
rhHLA-G reprogrammed PBMC cytokine secretion and regulated T-cell differentiation
Cytokines were measured in the supernatant of PBMC cultured with rhHLA-G. TNF-a, IL-12, and IL-17 levels were significantly reduced and IL-1β, IL-2, IL-4, IL-10, GCSF, and GM-CSF levels were significantly elevated in rhHLA-G-treated systems compared with untreated systems. No significant difference in IFN-g levels was found in the supernatant after rhHLA-G treatment (Figure 3A-J). IL-5, IL-6, IL-7, IL-8, IL-13, MCP-1, and MIP-1 levels secreted by ITP patient PBMC were below the detection limit.
In order to investigate whether rhHLA-G could enhance the expression of the Treg population, we also detected the percentage of CD4+CD25+Foxp3+ Tregs. Tregs percentages in ITP patients were significantly lower than in healthy controls; However, rhHLA-G exposure did not expand Tregs in ITP patients (Figure 3K).
rhHLA-G exposure attenuated ITP patient PBMC-induced platelet apoptosis
Given that rhHLA-G upregulated ILT and reprogrammed cytokine secretion, we hypothesized that the inhibitory functions of PBMC were restored. In order to testify the hypothesis, we measured the ability of PBMC to induce platelet apoptosis. PBMC from ITP patients induced significantly higher apoptosis of both autologous and healthy control platelets compared with healthy controls and the addition of rhHLA-G attenuated platelet destruction by ITP PBMC. The decrease of platelet apoptosis demonstrates a protective effect of rhHLA-G in ITP. In order to determine whether the platelet apoptosis was mediated by PBMC cytotoxicity and by the platelet themselves, healthy control PBMC were cultured with autologous platelets and ITP platelets respectively. The results showed that rhHLA-G modulation did not change the ability of healthy control PBMC to induce both autologous and ITP platelet apoptosis (Figure 4).
Since Fcg-receptor-mediated platelet phagocytosis is one of the most important mechanisms for antibody-mediated platelet destruction in ITP, we detected the Fcg-receptor (CD16) expression and antibody-mediated platelet phagocytic capability by monocytes/macrophages. Our results showed that treatment of rhHLA-G had no significant effect on monocytic Fcg-receptor (CD16) expression. Therefore, no difference was observed in the antibodymediated platelet phagocytic capability between rhHLAG modulated and unmodulated groups (Online Supplementary Figure S3), indicating that rhHLA-G protects ITP platelets through mechanisms other than by inhibiting platelet phagocytosis.
rhHLA-G suppressed DC maturation and T-cell proliferation in vitro
In order to assess whether rhHLA-G inhibits maturation of monocyte-derived DC, surface expression of CD80 and CD86 on DC was analyzed by flow cytometry. DC from ITP patients expressed higher levels of CD80 and CD86 than those from healthy controls. In the presence of rhHLA-G, CD80 and CD86 expression was markedly downregulated in both ITP patients and healthy controls (Figure 5A-D).
DC maturation, which increases the expression of costimulatory molecules, such as CD80, CD86 and related cytokines, could contribute to T-cell proliferation and differentiation. In order to further evaluate DC-mediated T-cell proliferation, allogeneic CD4+ T cells were cocultured with DC with or without rhHLA-G premodulation. DC from ITP patients stimulated significantly more CD4+ T-cell proliferation compared with those from healthy controls in vitro. However, rhHLA-G-modulated DC from ITP patients lost their suppressive effect on CD4+ T-cell proliferation compared to unmodulated cells (Figure 5E-F).
HLA-G treatment alleviated thrombocytopenia in a murine model of ITP
HLA-G capture by gold nanoparticle was observed by transmission electron microscopy (Figure 6A). The SCID mice engrafted with splenocytes from immunized CD61 KO mice exhibited profound thrombocytopenia within 28 days after transplantation. The SCID mice were separated into four groups. Group 1 received an intraperitoneal injection of 100 mL AuNP coated with 7.5ug HLA-G on the day of the splenocyte transplantation; group 2 received an intraperitoneal injection of same amount of control AuNP on the same day; group 3 received 7.5 ug HLA-G protein; group 4 received the same volume of phosphate-buffered saline as a blank control. According to routine monitored platelet counts, mice in group 1 showed significantly higher platelet counts than mice in group 2 at week 3 and 4 after irradiation. Mice in group 3 showed significantly higher platelet counts than mice in group 4, but lower platelet counts than mice in group 1 (Figure 6C). No significant difference in the survival rate among the four groups was observed in our study (Figure 6B).
Discussion
ITP is characterized by autoantibody-mediated platelet phagocytosis,1-3 in which the number deficiency and/or dysfunction of immune regulatory cells, such as Tregs, tolerogenic DC, and MDSC, leads to the loss of immune tolerance in ITP patients. As an important immunoregulatory molecule, HLA-G executes its inhibitory immune function through suppression of T cells, B cells, and APC.16,19
In this study, the levels of sHLA-G in ITP plasma is correlated with the presence of anti-platelet autoantibodies, but no significant difference was observed between Abnegative ITP patients and healthy controls, indicating the involvement of HLA-G in the pathogenesis of autoantibody- mediated platelet clearance. A previous study showed that HLA-G inhibited B-cell proliferation and antibody secretion,21 which may explain why patients with low levels of sHLA-G tended to have high levels of anti-platelet antibodies. Furthermore, sHLA-G positively correlated with platelet counts in ITP patients and in healthy controls, suggesting that HLA-G may have other protective effects.
sHLA-G exerts its inhibitory immune function by inter- acting with receptors ILT2 and ILT4, which were differentially expressed on CD4+, CD8+, CD14+, and CD19+ cells. In ITP patients, we observed decreased expression of ILT2 on CD4+ cells, as well as ILT4 on CD14+ cells in comparision with healthy controls. Consistent with the previous study,32 we found that rhHLA-G upregulated ILT4 expression in CD14+ monocytes, and ILT2 expression in CD4+ cells, but not on CD8+, CD14+, and CD19+ cells in ITP patients, probably due to the relatively high baseline expression of ILT2 on CD14+ and CD19+ cells. Shiroishi et al.36 reported that CD8 is also the receptor for HLA-G, and it can compete with ILT2 and ILT4 for HLA-G binding. sHLA-G triggers Fas/Fas-Ligand (FasL)-mediated apoptosis in CD8+ T cells by interacting with CD8 molecules,37 which could alleviate the suppression on megakaryocyte apoptosis and increase platelet production.38 ILT upregulation by rhHLA-G indicates that sHLA-G could also regulate immune cells.
Cytokines are crucial factors in maintaining self-tolerance by controlling cell proliferation, differentiation, and migration. We and other groups demonstrated that type 1 (IL-2, IFN-a, IL-12, TNF-a), type 17 (IL-17) cytokines were significantly increased and type 2 (IL-4, IL-5, IL-10, IL-13) were decreased in ITP patients.39-41 Patients whose platelet count responded to therapeutic regimens often experienced correction of a disturbed cytokine profile. Agaugué S et al.42 found that HLA-G contributed to tumor escape by expanding MDSC and supporting Th2 versus Th1/Th17. Meanwhile, HLA-G could also help restore the balance of cytokines in ITP patients. We found that rhHLA-G upregulated IL-4 and IL-10 secretion, and downregulated TNF-a, IL-12, and IL-17 secretion by ITP patient PBMC, indicating a promotion of Th2 and inhibition of Th1 and Th17 by rhHLA-G. Our results demostrated rhHLA-G did not expand the CD4+ CD25+ Foxp3+ Tregs. We suspect that rhHLA-G might exert its suppressive function through other mechanisms. A novel subset of regulatory T cells expressing HLA-G was described, and it was proven that CD4+ HLA-G+ Tregs suppressed T-cell proliferation by secreting IL-10 and sHLA-G.43,44 A more recent study by Pankratz et al.45 showed that CD4+ HLA-G+ Tregs are the key player in inflammation and can ameliorate graft-versus- host diseases in vivo. Here, the proportion of CD4+ HLA-G+ cells was significantly decreased in ITP patients, further establishing the role of HLA-G in reconstructing immune tolerance in ITP patients.
In addition, rhHLA-G exerted their immunosuppressive effects by promoting IL-1β, IL-2, G-CSF, and GMCSF production. A previous study demonstrated that GM-CSF suppresses the immune response by expanding MDSC population, which may inhibit CD4+ T-cell proliferation and CTL-mediated platelet lysis.46 Pickup et al.47 found that G-CSF promoted differentiation of CD11b+ Ly6G+cells, increased IL-10 secretion from myeloid cells and inhibited T-cell proliferation by upregulating inducible nitric oxide synthase, arginase, IL-6, IL-10, IL- 1β, and vascular endothelial growth factor, which are beneficial and protective in autoimmune diseases. On the other hand, IL-10 and IL-1β combined with GM-CSF and/or IL-2 are able to induce HLA-G protein expression.48 This may initiate a possible positive feedback loop connecting cytokines and immune cells, and supporting immune tolerance.
Binding of HLA-G to its cognate receptor ILT may repair the disturbed cytokine profile and restore the immunosuppressive functions of various cells, such as T cells and DC. In our study, rhHLA-G could not affect platelet apoptosis directly, while rhHLA-G modulation attenuated platelets apoptosis when cocultured with PBMC from ITP patients, indicating the protective effect of HLA-G is through modulation of PBMC. sHLA-G interacts with CD8 molecules and induces apoptosis of cytotoxic T cells.37 In addition, HLA-G can suppress the cytolytic function of CD8+ T cells through downregulation of granzyme B expression, and impair perforin granules polarization toward target cells,49,50 which might explain its protective effect to platelets. Therefore, rhHLA-G may be a potential strategy to protect their own platelets from destruction in clinical practice.
By reprogramming the cytokine profile in ITP patients, HLA-G established an optimal environment for the impaired cell populations, such as DC, to regain their tolerogenic function. We found that lower expression of mHLA-G and ILT4 on CD14+ cells from ITP patients, confirming the loss of immune tolerance in ITP. However, the effect of HLA-G on DC is still controversial. Liang et al.51 demonstrated that HLA-G-treated DC maintained a tolerogenic phenotype CD80low CD86low HLA-DRlow, and HLA-G inhibited DC maturation via IL-6 signaling pathway. However, Le Friec et al.52 found that sHLA-G inhibits human DC mediated T-cell proliferation without altering DC maturation processes. In order to determine the function of DC in ITP patients as well as the protective role of HLA-G, we sorted CD14+ cells and isolated DC in vitro, and then modulated them with rhHLA-G. As expected, binding of HLA-G to ILT2/4 on DC suppressed the expression of costimulatory molecules and showed decreased ability to stimulate T-cell proliferation. These results indicated that HLA-G could induce DC into tolerogenic DC.
As shown before, HLA-G exerts an immunosuppressive effect through interaction with ILT in humans. Interestingly, HLA-G could also interact with the murine receptor paired immunoglobulin-like inhibitory receptor (PIR-B), which is expressed on myeloid cell lineage and B lymphocytes, and triggers inhibitory signaling pathways. By binding with PIR-B, HLA-G aggregation could mimic the active polymeric structure found in vivo and has shown effectiveness in the context of allogeneic transplantation in mice.42 Moreover, the inhibitory ability of HLA-G polymers was about 100-fold higher than HLA-G monomer in arthritis mouse model.53 Here we demonstrated a single administration of HLA-G aggregated onto nanoparticles could alleviate thrombocytopenia in a murine model of ITP, further support the expectation of HLA-G as an immunosuppressive drug for autoimmune disease. PIR-B is absent on murine T and NK cells. Even though our in vitro study showed HLA-G could regulate T-cell differentiation, the HLA-G mediated effect on murine T and NK cells may be indirect.
Our data also demonstrated the safety of HLA-G protein (about 7.5 mg/per mouse) in the murine model of ITP. Liang et al.51 showed the effectiveness of HLA-G in inducing transplantation tolerance at a dosage of 20 ng per mouse. Kuroki et al.53 found HLA-G displayed significant anti-rheumatoid arthritis with a safety profile, even at a higher dose of 150 mg per mouse. Even though the optimal dosage of rhHLA-G used as a treatment option for ITP still needs to be further investigated, the recombinant HLA-G protein is expected to be safe in vivo.
Above all, the regulation of HLA-G and ILT expression is involved in the pathogenesis of ITP, and rhHLA-G could upregulate ILT expression and correct the dysfunction of immune cells. Several studies demonstrated that therapeutic reagents including dexamethasone and decitabine can induce HLA-G expression through different mechanisms. 54,55 Our results also showed increased sHLA-G expression in patients responding to HD-DXM (Figure 1D). Moreover, Pedersen and colleagues56 found that dexamethasone combined with 1a, 25-Dihydroxyvitamin D3 induced ILT expression. Hence, whether HLA-G is involved in HD-DXM response in ITP is worthwhile investigating in the future.
Conclusions
Decreased expression of HLA-G and ILT are involved in the immunopathogenesis of ITP. rhHLA-G upregulated HLA-G and ILT expression and corrected the dysfunction of immune cells in patients with ITP. Our study sheds new light on prognosis and treatment targeting HLA-G in the management of ITP.
Footnotes
- Received August 9, 2018
- Accepted February 12, 2020
Correspondence
Disclosures
No conflicts of interest to disclose.
Contributions
QF and JM designed the experiments, analyzed the data, and wrote the manuscript; XL and ZS performed the experiments, analyzed the data, and prepared the manuscript; JP, MH, HN evaluated the data and prepared the manuscript; MX corrected the manuscript; YS, YW, ZZ, HL, LS, YZ performed the experiment and analyzed data; JY, CM and CG analyzed data, and contributed to the manuscript preparation. All authors read and approved the final manuscript.
Funding
This work was supported by grants from the Natural Science Foundation of China (81800112, 91942306, 81770133, 81500096, 81900124, 81770114 and 81500094), State Key Clinical Specialty of China for Blood Disorders, and Tai Shan Scholar Foundation. HN was funded by the Canadian Institutes of Health Research Foundation grant (389035). HL acknowledges the Chinese "post-doctoral international exchange program" for a post-doctoral scholarship. Limei Wang (Advanced Medical Research Institute/Translational Medicine Core Facility of Advanced Medical Research Institute, Shandong University) provided help in flow cytometry.
Acknowledgments
ZZ acknowledges the Qilu Young Scholar program for funding.
References
- Rodeghiero F, Stasi R, Gernsheimer T. Standardization of terminology, definitions and outcome criteria in immune thrombocytopenic purpura of adults and children: report from an international working group. Blood. 2009; 113(11):2386-2393. https://doi.org/10.1182/blood-2008-07-162503PubMedGoogle Scholar
- Cines DB, Blanchette VS. Immune thrombocytopenic purpura. N Engl J Med. 2002; 346(13):995-1008. https://doi.org/10.1056/NEJMra010501PubMedGoogle Scholar
- Li J, Sullivan JA, Ni H. Pathophysiology of immune thrombocytopenia. Curr Opin Hematol. 2018; 25(5):373-381. https://doi.org/10.1097/MOH.0000000000000447PubMedGoogle Scholar
- Semple JW, Freedman J. Increased antiplatelet T helper lymphocyte reactivity in patients with autoimmune thrombocytopenia. Blood. 1991; 78(10):2619-2625. https://doi.org/10.1182/blood.V78.10.2619.2619PubMedGoogle Scholar
- Zeng Q, Zhu L, Tao L. Relative efficacy of steroid therapy in immune thrombocytopenia mediated by anti-platelet GPIIbIIIa versus GPIbalpha antibodies. Am J Hematol. 2012; 87(2):206-208. https://doi.org/10.1002/ajh.22211PubMedGoogle Scholar
- Peng J, Ma SH, Liu J. Association of autoantibody specificity and response to intravenous immunoglobulin G therapy in immune thrombocytopenia: a multicenter cohort study. J Thromb Haemost. 2014; 12(4):497-504. https://doi.org/10.1111/jth.12524PubMedGoogle Scholar
- Tao L, Zeng Q, Li J. Platelet desialylation correlates with efficacy of first-line therapies for immune thrombocytopenia. J Hematol Oncol. 2017; 10(1):46. https://doi.org/10.1186/s13045-017-0413-3PubMedPubMed CentralGoogle Scholar
- Audia S, Samson M, Mahevas M. Preferential splenic CD8(+) T-cell activation in rituximab-nonresponder patients with immune thrombocytopenia. Blood. 2013; 122(14):2477-2486. https://doi.org/10.1182/blood-2013-03-491415PubMedGoogle Scholar
- Yu J, Heck S, Patel V. Defective circulating CD25 regulatory T cells in patients with chronic immune thrombocytopenic purpura. Blood. 2008; 112(4):1325-1328. https://doi.org/10.1182/blood-2008-01-135335PubMedPubMed CentralGoogle Scholar
- Giri S, Pathak R, Aryal MR, Karmacharya P, Bhatt VR, Martin MG. Seasonal variation of immune thrombocytopenic purpura related hospitalizations among adults in the USA: analysis of the nationwide inpatient sample database. Ther Adv Hematol. 2015; 6(4):217-218. https://doi.org/10.1177/2040620715582146PubMedPubMed CentralGoogle Scholar
- Ma L, Simpson E, Li J. CD8+ T cells are predominantly protective and required for effective steroid therapy in murine models of immune thrombocytopenia. Blood. 2015; 126(2):247-256. https://doi.org/10.1182/blood-2015-03-635417PubMedPubMed CentralGoogle Scholar
- Ma J, Ning YN, Xu M. Thalidomide corrects impaired mesenchymal stem cell function in inducing tolerogenic DCs in patients with immune thrombocytopenia. Blood. 2013; 122(12):2074-2082. https://doi.org/10.1182/blood-2013-03-491555PubMedGoogle Scholar
- Hou Y, Feng Q, Xu M. High-dose dexamethasone corrects impaired myeloidderived suppressor cell function via Ets1 in immune thrombocytopenia. Blood. 2016; 127(12):1587-1597. https://doi.org/10.1182/blood-2015-10-674531PubMedGoogle Scholar
- Ellis SA, Palmer MS, McMichael AJ. Human trophoblast and the choriocarcinoma cell line BeWo express a truncated HLA Class I molecule. J Immunol. 1990; 144(2):731-735. Google Scholar
- Nardi Fda S, Konig L, Wagner B, Giebel B, Santos Manvailer LF, Rebmann V. Soluble monomers, dimers and HLA-G-expressing extracellular vesicles: the three dimensions of structural complexity to use HLA-G as a clinical biomarker. HLA. 2016; 88(3):77-86. https://doi.org/10.1111/tan.12844PubMedGoogle Scholar
- Hunt JS, Petroff MG, McIntire RH, Ober C. HLA-G and immune tolerance in pregnancy. FASEB J. 2005; 19(7):681-693. https://doi.org/10.1096/fj.04-2078revPubMedGoogle Scholar
- Wang Q, Song H, Cheng H. Structures of the four Ig-like domain LILRB2 and the four-domain LILRB1 and HLA-G1 complex. Cell Mol Immunol. 2020; 17(9):966-975. https://doi.org/10.1038/s41423-019-0258-5PubMedPubMed CentralGoogle Scholar
- Kirszenbaum M, Moreau P, Gluckman E, Dausset J, Carosella E. An alternatively spliced form of HLA-G mRNA in human trophoblasts and evidence for the presence of HLA-G transcript in adult lymphocytes. Proc Natl Acad Sci U S A. 1994; 91(10):4209-4213. https://doi.org/10.1073/pnas.91.10.4209PubMedPubMed CentralGoogle Scholar
- Le Friec G, Gros F, Sebti Y. Capacity of myeloid and plasmacytoid dendritic cells especially at mature stage to express and secrete HLA-G molecules. J Leukoc Biol. 2004; 76(6):1125-1133. https://doi.org/10.1189/jlb.0104015PubMedGoogle Scholar
- Lila N, Rouas-Freiss N, Dausset J, Carpentier A, Carosella ED. Soluble HLA-G protein secreted by allo-specific CD4+ T cells suppresses the allo-proliferative response: a CD4+ T cell regulatory mechanism. Proc Natl Acad Sci U S A. 2001; 98(21):12150-12155. https://doi.org/10.1073/pnas.201407398PubMedPubMed CentralGoogle Scholar
- Naji A, Menier C, Morandi F. Binding of HLA-G to ITIM-bearing Ig-like transcript 2 receptor suppresses B cell responses. J Immunol. 2014; 192(4):1536-1546. https://doi.org/10.4049/jimmunol.1300438PubMedGoogle Scholar
- Zarkhin V, Talisetti A, Li L. Expression of soluble HLA-G identifies favorable outcomes in liver transplant recipients. Transplantation. 2010; 90(9):1000-1005. https://doi.org/10.1097/TP.0b013e3181f546afPubMedGoogle Scholar
- Le Rond S, Azema C, Krawice-Radanne I. Evidence to support the role of HLA-G5 in allograft acceptance through induction of immunosuppressive/ regulatory T cells. J Immunol. 2006; 176(5):3266-3276. https://doi.org/10.4049/jimmunol.176.5.3266PubMedGoogle Scholar
- Qiu J, Terasaki PI, Miller J, Mizutani K, Cai J, Carosella ED. Soluble HLA-G expression and renal graft acceptance. Am J Transplant. 2006; 6(9):2152-2156. https://doi.org/10.1111/j.1600-6143.2006.01417.xPubMedGoogle Scholar
- Rouas-Freiss N, Moreau P, Ferrone S, Carosella ED. HLA-G proteins in cancer: do they provide tumor cells with an escape mechanism?. Cancer Res. 2005; 65(22):10139-10144. https://doi.org/10.1158/0008-5472.CAN-05-0097PubMedGoogle Scholar
- Nuckel H, Rebmann V, Durig J, Duhrsen U, Grosse-Wilde H. HLA-G expression is associated with an unfavorable outcome and immunodeficiency in chronic lymphocytic leukemia. Blood. 2005; 105(4):1694-1698. https://doi.org/10.1182/blood-2004-08-3335PubMedGoogle Scholar
- Verbruggen LA, Rebmann V, Demanet C, De Cock S, Grosse-Wilde H. Soluble HLA-G in rheumatoid arthritis. Hum Immunol. 2006; 67(8):561-567. https://doi.org/10.1016/j.humimm.2006.03.023PubMedGoogle Scholar
- Mitsdoerffer M, Schreiner B, Kieseier BC. Monocyte-derived HLA-G acts as a strong inhibitor of autologous CD4 T cell activation and is upregulated by interferonbeta in vitro and in vivo: rationale for the therapy of multiple sclerosis. J Neuroimmunol. 2005; 159(1-2):155-164. https://doi.org/10.1016/j.jneuroim.2004.09.016PubMedGoogle Scholar
- Chen J, Shen B, Jiang Y. Analysis of immunoglobulin-like transcripts (ILTs) in lymphocytes with sHLA-G and IL10 from SLE patients. Clin Exp Med. 2013; 13(2):135-142. https://doi.org/10.1007/s10238-012-0185-6PubMedGoogle Scholar
- Guerra-de Blas Pdel C, Villasenor-Talavera YS, Cruz-Gonzalez Dde J. Analysis of the expression and function of immunoglobulin- like transcript 4 (ILT4, LILRB2) in dendritic cells from patients with systemic lupus erythematosus. J Immunol Res. 2016; 2016:4163094. https://doi.org/10.1155/2016/4163094PubMedPubMed CentralGoogle Scholar
- Kim SK, Jeong KH, Kang IJ, Chung JH, Shin MK, Lee MH. Relationship between the HLA-G 14 bp insertion/deletion polymorphism and susceptibility to autoimmune disease: a meta-analysis. Genet Mol Res. 2015; 14(4):15839-15847. https://doi.org/10.4238/2015.December.1.35PubMedGoogle Scholar
- LeMaoult J, Zafaranloo K, Le Danff C, Carosella ED. HLA-G up-regulates ILT2, ILT3, ILT4, and KIR2DL4 in antigen presenting cells, NK cells, and T cells. FASEB J. 2005; 19(6):662-664. https://doi.org/10.1096/fj.04-1617fjePubMedGoogle Scholar
- LeMaoult J, Daouya M, Wu J, Loustau M, Horuzsko A, Carosella ED. Synthetic HLAG proteins for therapeutic use in transplantation. FASEB J. 2013; 27(9):3643-3651. https://doi.org/10.1096/fj.13-228247PubMedPubMed CentralGoogle Scholar
- Ni H, Chen P, Spring CM. A novel murine model of fetal and neonatal alloimmune thrombocytopenia: response to intravenous IgG therapy. Blood. 2006; 107(7):2976-2983. https://doi.org/10.1182/blood-2005-06-2562PubMedPubMed CentralGoogle Scholar
- Chow L, Aslam R, Speck ER. A murine model of severe immune thrombocytopenia is induced by antibody- and CD8+ T cellmediated responses that are differentially sensitive to therapy. Blood. 2010; 115(6):1247-1253. https://doi.org/10.1182/blood-2009-09-244772PubMedGoogle Scholar
- Shiroishi M, Tsumoto K, Amano K. Human inhibitory receptors Ig-like transcript 2 (ILT2) and ILT4 compete with CD8 for MHC class I binding and bind preferentially to HLA-G. Proc Natl Acad Sci U S A. 2003; 100(15):8856-8861. https://doi.org/10.1073/pnas.1431057100PubMedPubMed CentralGoogle Scholar
- Fournel S, Aguerre-Girr M, Huc X. Cutting edge: soluble HLA-G1 triggers CD95/CD95 ligand-mediated apoptosis in activated CD8+ cells by interacting with CD8. J Immunol. 2000; 164(12):6100-6104. https://doi.org/10.4049/jimmunol.164.12.6100PubMedGoogle Scholar
- Li S, Wang L, Zhao C, Li L, Peng J, Hou M. CD8+ T cells suppress autologous megakaryocyte apoptosis in idiopathic thrombocytopenic purpura. Br J Haematol. 2007; 139(4):605-611. https://doi.org/10.1111/j.1365-2141.2007.06737.xPubMedGoogle Scholar
- Zhang J, Ma D, Zhu X, Qu X, Ji C, Hou M. Elevated profile of Th17, Th1 and Tc1 cells in patients with immune thrombocytopenic purpura. Haematologica. 2009; 94(9):1326-1329. https://doi.org/10.3324/haematol.2009.007823PubMedPubMed CentralGoogle Scholar
- Rocha AM, Souza C, Rocha GA. The serum levels of the cytokines involved in the Th17 and Th1 cell commitment are increased in individuals with borderline thrombocytopenia. J Hematol Oncol. 2013; 6:28. https://doi.org/10.1186/1756-8722-6-28PubMedPubMed CentralGoogle Scholar
- Liu F, Wu C, Yang X. Polarization and apoptosis of T cell subsets in idiopathic thrombocytopenic purpura. Cell Mol Immunol. 2005; 2(5):387-392. Google Scholar
- Agaugue S, Carosella ED, Rouas-Freiss N. Role of HLA-G in tumor escape through expansion of myeloid-derived suppressor cells and cytokinic balance in favor of Th2 versus Th1/Th17. Blood. 2011; 117(26):7021-7031. https://doi.org/10.1182/blood-2010-07-294389PubMedGoogle Scholar
- Feger U, Tolosa E, Huang YH. HLA-G expression defines a novel regulatory T-cell subset present in human peripheral blood and sites of inflammation. Blood. 2007; 110(2):568-577. https://doi.org/10.1182/blood-2006-11-057125PubMedGoogle Scholar
- Huang YH, Zozulya AL, Weidenfeller C, Schwab N, Wiendl H. T cell suppression by naturally occurring HLA-G-expressing regulatory CD4+ T cells is IL-10-dependent and reversible. J Leukoc Biol. 2009; 86(2):273-281. https://doi.org/10.1189/jlb.1008649PubMedGoogle Scholar
- Pankratz S, Bittner S, Herrmann AM. Human CD4+ HLA-G+ regulatory T cells are potent suppressors of graft-versus-host disease in vivo. FASEB J. 2014; 28(8):3435-3445. https://doi.org/10.1096/fj.14-251074PubMedGoogle Scholar
- Bayne LJ, Beatty GL, Jhala N. Tumorderived granulocyte-macrophage colonystimulating factor regulates myeloid inflammation and T cell immunity in pancreatic cancer. Cancer Cell. 2012; 21(6):822-835. https://doi.org/10.1016/j.ccr.2012.04.025PubMedPubMed CentralGoogle Scholar
- Pickup MW, Owens P, Gorska AE. Development of aggressive pancreatic ductal adenocarcinomas depends on granulocyte colony stimulating factor secretion in carcinoma cells. Cancer Immunol Res. 2017; 5(9):718-729. https://doi.org/10.1158/2326-6066.CIR-16-0311PubMedPubMed CentralGoogle Scholar
- Moreau P, Flajollet S, Carosella ED. Nonclassical transcriptional regulation of HLAG: an update. J Cell Mol Med. 2009; 13(9B):2973-2989. https://doi.org/10.1111/j.1582-4934.2009.00800.xPubMedPubMed CentralGoogle Scholar
- Ajith A, Portik-Dobos V, Nguyen-Lefebvre AT. HLA-G dimer targets Granzyme B pathway to prolong human renal allograft survival. FASEB J. 2019; 33(4):5220-5236. https://doi.org/10.1096/fj.201802017RPubMedPubMed CentralGoogle Scholar
- Lesport E, Baudhuin J, LeMaoult J. Human melanoma cell secreting human leukocyte antigen-G5 inhibit natural killer cell cytotoxicity by impairing lytic granules polarization toward target cell. Hum Immunol. 2009; 70(12):1000-1005. https://doi.org/10.1016/j.humimm.2009.07.019PubMedGoogle Scholar
- Liang S, Ristich V, Arase H, Dausset J, Carosella ED, Horuzsko A. Modulation of dendritic cell differentiation by HLA-G and ILT4 requires the IL-6--STAT3 signaling pathway. Proc Natl Acad Sci U S A. 2008; 105(24):8357-8362. https://doi.org/10.1073/pnas.0803341105PubMedPubMed CentralGoogle Scholar
- Le Friec G, Laupeze B, Fardel O. Soluble HLA-G inhibits human dendritic cell-triggered allogeneic T-cell proliferation without altering dendritic differentiation and maturation processes. Hum Immunol. 2003; 64(8):752-761. https://doi.org/10.1016/S0198-8859(03)00091-0PubMedGoogle Scholar
- Kuroki K, Hirose K, Okabe Y. The longterm immunosuppressive effects of disulfide- linked HLA-G dimer in mice with collagen- induced arthritis. Hum Immunol. 2013; 74(4):433-438. https://doi.org/10.1016/j.humimm.2012.11.060PubMedGoogle Scholar
- Akhter A, Faridi RM, Das V, Pandey A, Naik S, Agrawal S. In vitro up-regulation of HLAG using dexamethasone and hydrocortisone in first-trimester trophoblast cells of women experiencing recurrent miscarriage. Tissue Antigens. 2012; 80(2):126-135. https://doi.org/10.1111/j.1399-0039.2012.01884.xPubMedGoogle Scholar
- Yan WH, Lin AF, Chang CC, Ferrone S. Induction of HLA-G expression in a melanoma cell line OCM-1A following the treatment with 5-aza-2'-deoxycytidine. Cell Res. 2005; 15(7):523-531. https://doi.org/10.1038/sj.cr.7290376PubMedGoogle Scholar
- Pedersen AE, Gad M, Walter MR, Claesson MH. Induction of regulatory dendritic cells by dexamethasone and 1alpha,25-dihydroxyvitamin D(3). Immunol Lett. 2004; 91(1):63-69. https://doi.org/10.1016/j.imlet.2003.11.004PubMedGoogle Scholar
Data Supplements
Figures & Tables
Article Information
This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.