Skip to main content
Clinical and Experimental Immunology logoLink to Clinical and Experimental Immunology
. 2018 May 23;193(2):152–159. doi: 10.1111/cei.13134

Evaluation of membrane‐bound and soluble forms of human leucocyte antigen‐G in systemic sclerosis

P Contini 1, S Negrini 1, G Murdaca 1, M Borro 1, F Puppo 1,
PMCID: PMC6046504  PMID: 29660112

Summary

Systemic sclerosis (SSc) is a complex disease characterized by immune dysregulation, extensive vascular damage and widespread fibrosis. Human leucocyte antigen‐G (HLA‐G) is a non‐classic class I major histocompatibility complex (MHC) molecule characterized by complex immunomodulating properties. HLA‐G is expressed on the membrane of different cell lineages in both physiological and pathological conditions. HLA‐G is also detectable in soluble form (sHLA‐G) deriving from the shedding of surface isoforms (sHLA‐G1) or the secretion of soluble isoforms (HLA‐G5). Several immunosuppressive functions have been attributed to both membrane‐bound and soluble HLA‐G molecules. The plasma levels of sHLA‐G were higher in SSc patients (444·27 ± 304·84 U/ml) compared to controls (16·74 ± 20·58 U/ml) (P < 0·0001). The plasma levels of transforming growth factor (TGF)‐β were higher in SSc patients (18 937 ± 15 217 pg/ml) compared to controls (11 099 ± 6081 pg/ml; P = 0·003), and a significant correlation was found between TGF‐β and the plasma levels of total sHLA‐G (r = 0·65; P < 0·01), sHLA‐G1 (r = 0·60; P = 0·003) and HLA‐G5 (r = 0·47; P = 0·02). The percentage of HLA‐G‐positive monocytes (0·98 ± 1·72), CD4+ (0·37 ± 0·68), CD8+ (2·05 ± 3·74) and CD4+CD8+ double‐positive cells (14·53 ± 16·88) was higher in SSc patients than in controls (0·11 ± 0·08, 0·01 ± 0·01, 0·01 ± 0·01 and 0·39 ± 0·40, respectively) (P < 0·0001). These data indicate that in SSc the secretion and/or shedding of soluble HLA‐G molecules and the membrane expression of HLA‐G by peripheral blood mononuclear cells (PBMC) is clearly elevated, suggesting an involvement of HLA‐G molecules in the immune dysregulation of SSc.

Keywords: HLA‐G, scleroderma, systemic sclerosis, TGF‐β

Introduction

Systemic sclerosis (SSc) is a connective tissue disease characterized by diffuse fibrosis and obliterative vascular lesions occurring in skin and internal organs 1. SSc is classified into limited (lSSc) and diffuse (dSSc) forms presenting different cutaneous and visceral involvement and specific autoantibody patterns 2. Although the causative factors remain to be characterized, three pathogenetic events underlie SSc development, namely vascular damage, immune dysregulation and fibroblast activation 3, 4, 5. In particular, endothelin‐1 (ET‐1) levels are increased, leading to activation of vascular endothelial cells, arteriolar hyperreactivity, myointimal proliferation and vascular occlusion 6, whereas vascular epidermal growth factor (VEGF) levels are decreased, contributing to the impaired vascularization of digital arteries 7. As far as the immune system is concerned, autoantibodies 8, high levels of B cell activating factor (BAFF) 9 and perturbations of dendritic cells 10 have been reported. Furthermore, alterations of the normal functional balance between proinflammatory subpopulations, in particular T helper type 17 (Th17) and regulatory T cell (Treg) subpopulations, including both CD4+ and CD8+ Treg subsets, have been demonstrated in patients affected by SSc 11, 12, 13. Of note, double‐positive CD4/CD8 T lymphocytes have been described in dermal lesions of patients with SSc; these cells secrete elevated amounts of interleukin (IL)‐4, which has profibrotic properties 14. Finally, connective tissue growth factor (CTGF), platelet‐derived growth factor (PDGF) and transforming growth factor (TGF)‐β levels are increased in SSc, thus favouring fibroblast activation and extracellular matrix synthesis 1, 15, 16.

Human leucocyte antigen (HLA)‐G is a human non‐classical major histocompatibility complex (MHC) molecule expressed mainly in membrane‐bound form at the fetal–maternal interface on the extravillous cytotrophoblast 17 and in placental tissue 18, where it contributes to the development of maternal tolerance to the semi‐allogeneic fetus 19, 20. It is also expressed physiologically in a few adult tissues, such as thymus 21, cornea 22, pancreas 23 and bronchial epithelial cells 24, as well as in different cell types such as activated monocytes and erythroid and endothelial precursors 25. The expression of HLA‐G antigens has been reported recently in some solid tumours, transplanted organs and cutaneous inflammatory diseases as well as on virally infected cells 26, 27. HLA‐G is also detectable in soluble form (sHLA‐G) in several body fluids, deriving from the secretion of soluble isoforms (HLA‐G5) and/or the shedding of proteolytically cleaved surface isoforms (sHLA‐G1) 28, 29, 30. Elevated levels of sHLA‐G molecules have been detected in plasma of patients affected by various pathological conditions 31, 32, 33, 34, 35, as well as in cerebrospinal fluid of patients with multiple sclerosis 36.

Several immune functions have been attributed to both membrane‐bound and soluble HLA‐G molecules 37, 38. It has been proposed recently that HLA‐G should be qualified as an ‘immune checkpoint’ molecule 39. Most functions of HLA‐G molecules are immunosuppressive, as they inhibit the cytolytic function of natural killer (NK) cells and CD8+ T lymphocytes 40, 41, the alloproliferative response of CD4+ T cells 42, maturation of dendritic cells 43 and activation of B cells 44. In addition, HLA‐G molecules are able to trigger apoptosis in antigen‐specific CD8+ T lymphocytes 41, 45, 46. HLA‐G also seems to be involved in the tuning of immune responses, as in‐vitro studies indicate that incubation of peripheral blood mononuclear cells (PBMC) with HLA‐G‐expressing cells favour a shift towards a Th‐2 cytokine profile, whereas incubation with sHLA‐G protein may have a counterbalancing effect by creating an anti‐inflammatory environment due to the release of IL‐10 47, 48. Finally, HLA‐G‐positive Tregs have been detected in peripheral blood and inflamed tissues 49, 50, 51, 52.

HLA‐G molecules have been detected in approximately 50% of skin biopsies from SSc patients, and their expression has been associated with a better clinical outcome 53. Furthermore, in a recently published paper it has been reported that the serum levels of sHLA‐G are lower in SSc patients than in normal subjects, although the difference was not statistically significant, and that, within SSc patients, the lowest HLA‐G levels were associated with a more severe or active disease 54.

The aims of the present study were: (i) to determine the plasma levels of sHLA‐G molecules in a cohort of SSc patients with the limited or diffuse form of the disease; (ii) to correlate sHLA‐G levels with TGF‐β; and (iii) to evaluate the expression of HLA‐G in PBMC.

Materials and methods

Patients

Thirty‐five patients affected by SSc (28 females and seven males, aged 40–89 years) followed at the Clinical Immunology Unit (Department of Internal Medicine, University of Genoa, Genoa, Italy) were included in the present study. The patients satisfied the European League Against Rheumatism/American College of Rheumatology (EULAR/ACR) 2013 classification criteria for SSc 55. Patients were subclassified as having lSSc (23 subjects) or dSSc (12 subjects), according to the LeRoy criteria 2. Disease duration was determined from the onset of the first non‐Raynaud's manifestation. Forty healthy subjects, matched for sex and age, were recruited as controls. This study was approved by the local Ethical Committee and all patients signed a written informed consent. The study was conducted in accordance with the principles of the Declaration of Helsinki

Plasma samples

Plasma from both SSc patients and healthy donors was obtained by centrifugation (1125 g for 15 min at room temperature) from heparin tubes and stored at −80°C until use. Plasma was preferred to serum, as the amount of soluble HLA‐G molecules is higher in plasma than in serum 56.

sHLA‐G determination

The determination of sHLA‐G molecules was performed by sandwich immunoenzymatic assays. A commercially available assay (Exbio, Vestec, Czech Republic), which employs the MEM‐G9 monoclonal antibody (mAb) as capture antibody, was utilized to determine the total sHLA‐G amount. In order to determine the specific amount of sHLA‐G1 or HLA‐G5, immunoenzymatic assays were performed using the 01G mAb (Exbio) or the 5A6G7 mAb (Exbio) as capture antibodies, respectively. The HRP‐conjugated B2M mAb (Exbio), which recognizes β2‐microglobulin, was employed as detection antibody in all assays. Plates were read with an ELX800 ELISA reader (BioTek Instruments, Inc., Winooski, VT, USA) and results were expressed as U/ml.

TGF‐β determination

The determination of TGF‐β was performed by immunoenzymatic assay utilizing a commercially available kit (Bender MedSystems, Vienna, Austria) and results were expressed as pg/ml.

Flow cytometric analysis

The expression of cell membrane antigens by peripheral blood mononuclear cells (PBMC) was analysed by direct immunofluorescence incubating 100 µl of peripheral blood from each individual with the fluorochrome‐conjugated anti‐HLA‐G mAb MEM‐G9 (Exbio), which reacts with the native form of HLA‐G1, and with the fluorochrome‐conjugated anti‐CD3, ‐CD4, ‐CD8, ‐CD14 and ‐CD45 mAbs (Beckman Coulter Europe, Cassina de'Pecchi, Italy) at 4°C for 30 min in the dark. Fluorochrome‐conjugated isotype matched antibodies were used as controls. After red blood cell lysis, analysis was performed by flow cytometry using a Navios flow cytometer equipped with kaluza software (Beckman Coulter Europe).

Statistical analysis

Values are expressed as mean ± standard deviation. Comparisons between sHLA‐G plasma levels in SSc patients and controls were performed by unpaired Student's t‐test. Correlations between sHLA‐G plasma levels and TFG‐β levels were performed by Pearson's correlation coefficient. Comparisons between the percentages of different PBMC subpopulations were performed by Mann–Whitney U‐test. A P‐value < 0·05 was considered statistically significant. Statistical analyses were performed using GraphPad Prism version 6.03 for Windows (GraphPad Software, San Diego, CA, USA).

Results

sHLA‐G and TGF‐β plasma levels

The plasma levels of total sHLA‐G molecules were significantly higher in SSc patients (444·27 ± 304·84 U/ml) compared to controls (16·74 ± 20·58 U/ml; P < 0·0001) (Fig. 1a). In SSc patients, plasma levels of sHLA‐G1 and HLA‐G5 isoforms were comparable (264·66 ± 226·95 U/ml and 181·44 ± 130·12 U/ml, respectively) (Fig. 1b). In addition, no significant differences were detected in total sHLA‐G, sHLA‐G1 and HLA‐G5 plasma levels between dSSc and lSSc patients (data not shown). As expected, the levels of sHLA‐G1 and HLA‐G5 correlated with those of total sHLA‐G (r = 0·92; P < 0·0001 and r = 0·72; P < 0·0001, respectively) (Fig. 1c,d). No correlation was found between the level of sHLA‐G molecules and the disease features of each single patient (e.g. lung function tests, pulmonary artery pressure, autoantibodies pattern, nailfold videocapillaroscopy pattern, disease's form and duration). The plasma levels of TGF‐β were significantly higher in SSc patients (18 937 ± 15 217 pg/ml) compared to controls (11 099 ± 6081 pg/ml; P = 0·003) (Fig. 2a). A significant correlation was found between the plasma levels of TGF‐β and the plasma levels of total sHLA‐G (r = 0·65; P < 0·01), sHLA‐G1 (= 0·60; P = 0·003) and HLA‐G5 (r = 0·47; P = 0·02) (Fig. 2b–d).

Figure 1.

Figure 1

Plasma levels of soluble human leucocyte antigen‐G (sHLA‐G) molecules. (a) Total sHLA‐G concentrations in systemic sclerosis (SSc) patients and healthy donors. (b) HLA‐G1 and sHLA‐G5 concentrations in SSc patients. (c,d) Correlations between total sHLA‐G concentration and sHLA‐G1 and sHLA‐G5 plasma levels in SSc patients.

Figure 2.

Figure 2

Transforming growth factor (TGF)‐β plasma concentration and correlation with soluble human leucocyte antigen‐G (sHLA‐G) levels. (a) TGF‐β levels in systemic sclerosis (SSc) patients and healthy controls. (b,c,d) Correlations between TGF‐β concentrations and total sHLA‐G, sHLA‐G1 and sHLA‐G5 plasma levels in SSc patients.

Determination of HLA‐G expression in PBMC

The percentage of HLA‐G‐positive monocytes was significantly higher in SSc patients (0·98 ± 1·72) than in controls (0·11 ± 0·08; P < 0·0001). The percentage of CD4+ and CD8+ cells expressing HLA‐G molecules was significantly higher in PBMC from SSc patients (0·37 ± 0·68 and 2·05 ± 3·74, respectively) compared to controls (0·01 ± 0·01; P < 0·0001 and 0·01 ± 0·01; P < 0·0001, respectively). The frequencies of T lymphocytes subpopulations were comparable between controls and SSc patients (the mean of CD3+CD4+ lymphocytes was 70·5 and 71·3% and the mean of CD3+CD8+ lymphocytes was 26·5 and 27·4% in controls and SSc patients, respectively). The mean of monocytes (CD45+CD14+) was 8·2 and 5·4% in controls and SSc patients, respectively. Moreover, SSc patients displayed a higher, although not significantly different, percentage of CD4+CD8+ double‐positive (DP) cells (4·95 ± 8·31) in comparison with controls (2·45 ± 1·79). In the context of DP cells, SSc patients showed a significantly higher percentage of HLA‐G+ cells compared to controls (14·53 ± 16·88 and 0·39 ± 0·40, respectively; P < 0·0002). Furthermore, among DP cells a subpopulation of CD4dullCD8high cells was detectable in both SSc patients and controls (1·43 ± 3·58 and 1·27 ± 0·65, respectively). Of interest, CD4dullCD8high cells from SSc patients presented a high percentage of HLA‐G+ cells (30·47 ± 26·75) whereas HLA‐G expression was virtually absent in the same subpopulation from healthy controls. If the limited and diffuse forms of disease were compared, no significant differences were observable in the percentage of HLA‐G‐positive cells among DP and CD4dullCD8high populations. Representative examples of HLA‐G expression by monocytes, CD4+, CD8+, DP and CD4dullCD8high cells from a SSc patient and a healthy control are shown in Figs 3 and 4. Of interest, a significant correlation was found between sHLA‐G plasma levels and the percentage of HLA‐G expressing monocytes (r = 0·4, P < 0·01).

Figure 3.

Figure 3

Surface expression of human leucocyte antigen‐G (HLA‐G) molecule on peripheral blood mononuclear cells from systemic sclerosis (SSc) patients and healthy controls.

Figure 4.

Figure 4

Surface expression of human leucocyte antigen‐G (HLA‐G) molecule on CD4highCD8dull and CD4dullCD8high subpopulations from systemic sclerosis (SSc) patients and healthy controls.

Discussion

In the present investigation we have shown that the plasma levels of sHLA‐G molecules were significantly higher in SSc patients than in healthy subjects. Both sHLA‐G1 and HLA‐G5 isoforms accounted for this increase, and their levels correlated with those of total sHLA‐G. The plasma levels of total sHLA‐G, sHLA‐G1 and HLA‐G5 molecules did not differ significantly between patients affected by the diffuse or the localized form of disease. Moreover, no correlation was found between clinical characteristics (e.g. lung function tests, pulmonary artery pressure, autoantibody pattern, nailfold videocapillaroscopy pattern, disease form and duration) and the level of sHLA‐G molecules. Accordingly, we argue that the increase of sHLA‐G levels could be considered an intrinsic characteristic of the disease itself that is not related to specific clinical features of the single patient. Nevertheless, we cannot exclude that the lack of difference observed could be explained by the low number of subjects assessed. These findings are in disagreement with those published recently by other authors who did not find significant differences in sHLA‐G molecule levels between SSc patients and healthy donors 54. A possible explanation of this discrepancy might be the difference in the immunoenzymatic methods utilized by each research group for the quantitative determination of sHLA‐G molecules.

In the present study we have also shown that the plasma levels of TGF‐β were significantly higher in SSc patients than in controls and correlated with those of soluble HLA‐G molecules. This finding is in keeping with the results of in‐vitro experiments in which the production of TGF‐β1 by myelomonocytic cells was increased strongly after incubation with recombinant soluble HLA‐G molecules 57. Moreover, this observation supports the hypothesis that sHLA‐G may contribute to the immunopathogenesis of systemic sclerosis because TGF‐β has been identified as a major activator of fibroblasts in both mouse models and human disease 58, 59, 60, 61.

Further experiments have been performed in order to analyse the membrane expression of HLA‐G in PBMC from SSc patients. First, we have demonstrated that the percentage of HLA‐G positive monocytes is increased in SSc patients. This finding is in agreement with published data reporting that mononuclear phagocytes express HLA‐G mRNA and protein and that interferon (IFN)‐γ enhances HLA‐G expression selectively, which could influence inflammatory responses 62. Of note, the percentage of HLA‐G‐positive monocytes correlated significantly with the plasma levels of sHLA‐G molecules. Secondly, we found that the percentage of CD4+ cells and CD8+ cells expressing HLA‐G molecules was significantly higher in SSc patients than in controls. These CD4+HLA‐G+ and CD8+HLA‐G+ cells, that are different from the naturally occurring CD25+forkhead box protein 3 (FoxP3+) Tregs, may be involved in the modulation of immune responses in systemic sclerosis, as it has been reported that these HLA‐G‐expressing lymphocytes exert potent suppressive function and are present in inflamed tissues of patients affected by immune mediated disorders 50, 63, 64. Interestingly, HLA‐G molecules were detected in epidermal and dermal cells of skin biopsies from approximately 50% of patients with SSc, and HLA‐G expression was associated with a lower frequency of cutaneous ulcers, telangiectasias and polyarthralgias 53. Thirdly, we detected a significant increase in CD4+CD8+ DP cells in patients affected by SSc compared to healthy subjects. The function of circulating CD4+CD8+ DP T cells remains controversial, with conflicting reports describing cytotoxic activity in viral infections or immune suppressive roles in inflammatory disorders 65. Of note, CD4+CD8+ DP cells are present in the skin of patients with early active SSc and may contribute to the enhanced extracellular matrix deposition by fibroblasts through the production of high levels of IL‐4 14. Furthermore, CD4+CD8+ DP cells are found in other autoimmune diseases, such as systemic lupus erythematosus, where they exert a suppressive role in the production of autoantibodies 66. Finally, a subpopulation of CD4dullCD8high cells expressing HLA‐G‐positive cells was detected among CD4+CD8+ DP cells in SSc patients but not in healthy controls.

Collectively, these data indicate clearly that in SSc the secretion and/or shedding of soluble HLA‐G molecules and the membrane expression of HLA‐G by PBMC is elevated. A possible involvement of HLA‐G molecules in SSc pathogenesis might be suggested by the correlation between sHLA‐G and TFG‐β plasma levels leading to fibroblast activation and fibrosis development. Moreover, as the immunosuppressive role of HLA‐G is demonstrated widely 40, 41, 42, 43, 44, 45, 46, we suggest that the increased release of soluble HLA‐G forms and the up‐regulation of HLA‐G membrane expression by PBMC may reflect an attempt to control the immune derangement occurring in systemic sclerosis. This hypothesis is supported by previously published data indicating that the expression of HLA‐G in the skin is associated with a better disease prognosis 53, whereas low sHLA‐G serum levels associate with a worse clinical course 54. However, available data are scanty and sometimes conflicting, therefore further perspective basic and clinical investigations are required in order to define more clearly the modulatory role, if any, of HLA‐G molecules in SSc pathogenesis and clinical course.

Disclosure

The authors have no conflicting financial interests.

Acknowledgements

This work was supported by Gruppo Italiano Lotta alla Sclerodermia (GILS). The sponsor had no role in study design, in the collection, analysis and interpretation of data, in the writing of the report or in the decision to submit the article for publication.

References

  • 1. Gabrielli A, Avvedimento EV, Krieg T. Scleroderma. N Engl J Med 2009; 360:1989–2003. [DOI] [PubMed] [Google Scholar]
  • 2. LeRoy EC, Black C, Fleischmajer R et al Scleroderma (systemic sclerosis): classification, subsets and pathogenesis. J Rheumatol 1988; 15:202–5. [PubMed] [Google Scholar]
  • 3. Dumoitier N, Lofek S, Mouthon L. Pathophysiology of systemic sclerosis: state of the art in 2014. Presse Med 2014; 43:e267–78. [DOI] [PubMed] [Google Scholar]
  • 4. Negrini S, Fenoglio D, Parodi A et al Phenotypic alterations involved in CD8+ Treg impairment in systemic sclerosis. Front Immunol 2017; 8:18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Pattanaik D, Brown M, Postlethwaite BC, Postlethwaite AE. Pathogenesis of systemic sclerosis. Front Immunol 2015; 6:272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Jing J, Dou TT, Yang JQ et al Role of endothelin‐1 in the skin fibrosis of systemic sclerosis. Eur Cytokine Netw 2015; 26:10–4. [DOI] [PubMed] [Google Scholar]
  • 7. Distler O, Distler JH, Scheid A et al Uncontrolled expression of vascular endothelial growth factor and its receptors leads to insufficient skin angiogenesis in patients with systemic sclerosis. Circ Res 2004; 95:109–16. [DOI] [PubMed] [Google Scholar]
  • 8. Sato S, Fujimoto M, Hasegawa M, Takehara K, Tedder TF. Altered B lymphocyte function induces systemic autoimmunity in systemic sclerosis. Mol Immunol 2004; 41:1123–33. [DOI] [PubMed] [Google Scholar]
  • 9. Bosello S, Pers JO, Rochas C et al BAFF and rheumatic autoimmune disorders: implications for disease management and therapy. Int J Immunopathol Pharmacol 2007; 20:1–8. [DOI] [PubMed] [Google Scholar]
  • 10. Lu TT. Dendritic cells: novel players in fibrosis and scleroderma. Curr Rheumatol Rep 2012; 14:30–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Fenoglio D, Battaglia F, Parodi A et al Alteration of Th17 and Treg cell subpopulations co‐exist in patients affected with systemic sclerosis. Clin Immunol 2011; 139:249–57. [DOI] [PubMed] [Google Scholar]
  • 12. Papp G, Horvath IF, Barath S et al Altered T‐cell and regulatory cell repertoire in patients with diffuse cutaneous systemic sclerosis. Scand J Rheumatol 2011; 40:205–10. [DOI] [PubMed] [Google Scholar]
  • 13. Fenoglio D, Bernuzzi F, Battaglia F et al Th17 and regulatory T lymphocytes in primary biliary cirrhosis and systemic sclerosis as models of autoimmune fibrotic diseases. Autoimmun Rev 2012; 12:300–4. [DOI] [PubMed] [Google Scholar]
  • 14. Parel Y, Aurrand‐Lions M, Scheja A, Dayer JM, Roosnek E, Chizzolini C. Presence of CD4+CD8+ double‐positive T cells with very high interleukin‐4 production potential in lesional skin of patients with systemic sclerosis. Arthritis Rheum 2007; 56:3459–67. [DOI] [PubMed] [Google Scholar]
  • 15. Ayers NB, Sun C, Chen SY. Transforming growth factor‐b signaling in systemic sclerosis. J Biomed Res 2017; doi: 10.7555/JBR.31.20170034. [Epub ahead of print] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Iwayama T, Olson LE. Involvement of PDGF in fibrosis and scleroderma: recent insights from animal models and potential therapeutic opportunities. Curr Rheumatol Rep 2013; 15:304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Ellis SA, Palmer MS, McMichael AJ. Human trophoblast and the choriocarcinoma cell line BeWo express a truncated HLA class I molecule. J Immunol 1990; 144:731–5. [PubMed] [Google Scholar]
  • 18. Hviid TV, Larsen LG, Hoegh AM, Bzorek M. HLA‐G expression in placenta in relation to HLA‐G genotype and polymorphisms. Am J Reprod Immunol 2004; 52:212–7. [DOI] [PubMed] [Google Scholar]
  • 19. Kovats S, Main EK, Librach C, Stubblebine M, Fisher SJ, DeMars R. A class I antigen, HLA‐G, expressed in human trophoblasts. Science 1990; 248:220–3. [DOI] [PubMed] [Google Scholar]
  • 20. Ferreira LM, Meissner TB, Tilburgs T, Strominger JL. HLA‐G: at the interface of maternal–fetal tolerance. Trends Immunol 2017; 38:272–86. [DOI] [PubMed] [Google Scholar]
  • 21. Crisa L, McMaster MT, Ishii JK, Fisher SJ, Salomon DR. Identification of a thymic epithelial cell subset sharing expression of the class Ib HLA‐G molecule with fetal trophoblasts. J Exp Med 1997; 186:289–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Le Discorde M, Moreau P, Sabatier P, Legeais JM, Carosella ED. Expression of HLA‐G in human cornea, an immune‐privileged tissue. Hum Immunol 2003; 64:1039–44. [DOI] [PubMed] [Google Scholar]
  • 23. Cirulli V, Zalatan J, McMaster M et al The class I HLA repertoire of pancreatic islets comprises the nonclassical class Ib antigen HLA‐G. Diabetes 2006; 55:1214–22. [DOI] [PubMed] [Google Scholar]
  • 24. Brugiere O, Thabut G, Pretolani M et al Immunohistochemical study of HLA‐G expression in lung transplant recipients. Am J Transplant 2009; 9:1427–38. [DOI] [PubMed] [Google Scholar]
  • 25. Menier C, Rabreau M, Challier JC, Le Discorde M, Carosella ED, Rouas‐Freiss N. Erythroblasts secrete the nonclassical HLA‐G molecule from primitive to definitive hematopoiesis. Blood 2004; 104:3153–60. [DOI] [PubMed] [Google Scholar]
  • 26. Carosella ED, Favier B, Rouas‐Freiss N, Moreau P, Lemaoult J. Beyond the increasing complexity of the immunomodulatory HLA‐G molecule. Blood 2008; 111:4862–70. [DOI] [PubMed] [Google Scholar]
  • 27. Fainardi E, Castellazzi M, Stignani M et al Emerging topics and new perspectives on HLA‐G. Cell Mol Life Sci 2011; 68:433–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Paul P, Cabestre FA, Ibrahim EC et al Identification of HLA‐G7 as a new splice variant of the HLA‐G mRNA and expression of soluble HLA‐G5, ‐G6, and ‐G7 transcripts in human transfected cells. Hum Immunol 2000; 61:1138–49. [DOI] [PubMed] [Google Scholar]
  • 29. Rebmann V, LeMaoult J, Rouas‐Freiss N, Carosella ED, Grosse‐Wilde H. Quantification and identification of soluble HLA‐G isoforms. Tissue Antigens 2007; 69:143–9. [DOI] [PubMed] [Google Scholar]
  • 30. Tabayoyong WB, Zavazava N. Soluble HLA revisited. Leuk Res 2007; 31:121–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Carosella ED, Moreau P, Aractingi S, Rouas‐Freiss N. HLA‐G: a shield against inflammatory aggression. Trends Immunol 2001; 22:553–5. [DOI] [PubMed] [Google Scholar]
  • 32. Pistoia V, Morandi F, Wang X, Ferrone S. Soluble HLA‐G: are they clinically relevant? Semin Cancer Biol 2007; 17:469–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Carosella ED, Moreau P, Lemaoult J, Rouas‐Freiss N. HLA‐G: from biology to clinical benefits. Trends Immunol 2008; 29:125–32. [DOI] [PubMed] [Google Scholar]
  • 34. Murdaca G, Contini P, Setti M et al Behavior of non‐classical soluble HLA class G antigens in human immunodeficiency virus 1‐infected patients before and after HAART: comparison with classical soluble HLA‐A, ‐B, ‐C antigens and potential role in immune‐reconstitution. Clin Immunol 2009; 133:238–44. [DOI] [PubMed] [Google Scholar]
  • 35. Menier C, Rouas‐Freiss N, Favier B, LeMaoult J, Moreau P, Carosella ED. Recent advances on the non‐classical major histocompatibility complex class I HLA‐G molecule. Tissue Antigens 2010; 75:201–6. [DOI] [PubMed] [Google Scholar]
  • 36. Fainardi E, Rizzo R, Melchiorri L et al CSF levels of soluble HLA‐G and Fas molecules are inversely associated to MRI evidence of disease activity in patients with relapsing–remitting multiple sclerosis. Mult Scler 2008; 14:446–54. [DOI] [PubMed] [Google Scholar]
  • 37. Carosella ED. The tolerogenic molecule HLA‐G. Immunol Lett 2011; 138:22–4. [DOI] [PubMed] [Google Scholar]
  • 38. Gonzalez A, Rebmann V, LeMaoult J, Horn PA, Carosella ED, Alegre E. The immunosuppressive molecule HLA‐G and its clinical implications. Crit Rev Clin Lab Sci 2012; 49:63–84. [DOI] [PubMed] [Google Scholar]
  • 39. Carosella ED, Rouas‐Freiss N, Tronik‐Le Roux D, Moreau P, LeMaoult J. HLA‐G: an immune checkpoint molecule. Adv Immunol 2015; 127:33–144. [DOI] [PubMed] [Google Scholar]
  • 40. Rouas‐Freiss N, Goncalves RM, Menier C, Dausset J, Carosella ED. Direct evidence to support the role of HLA‐G in protecting the fetus from maternal uterine natural killer cytolysis. Proc Natl Acad Sci USA 1997; 94:11520–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Contini P, Ghio M, Poggi A et al Soluble HLA‐A,‐B,‐C and ‐G molecules induce apoptosis in T and NK CD8+ cells and inhibit cytotoxic T cell activity through CD8 ligation. Eur J Immunol 2003; 33:125–34. [DOI] [PubMed] [Google Scholar]
  • 42. 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 USA 2001; 98:12150–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Le Friec G, Laupeze B, Fardel O et al Soluble HLA‐G inhibits human dendritic cell‐triggered allogeneic T‐cell proliferation without altering dendritic differentiation and maturation processes. Hum Immunol 2003; 64:752–61. [DOI] [PubMed] [Google Scholar]
  • 44. Naji A, Menier C, Morandi F et al Binding of HLA‐G to ITIM‐bearing Ig‐like transcript 2 receptor suppresses B cell responses. J Immunol 2014; 192:1536–46. [DOI] [PubMed] [Google Scholar]
  • 45. Fournel S, Aguerre‐Girr M, Huc X et al Cutting edge: soluble HLA‐G1 triggers CD95/CD95 ligand‐mediated apoptosis in activated CD8+ cells by interacting with CD8. J Immunol 2000; 164:6100–4. [DOI] [PubMed] [Google Scholar]
  • 46. Contini P, Ghio M, Merlo A, Poggi A, Indiveri F, Puppo F. Apoptosis of antigen‐specific T lymphocytes upon the engagement of CD8 by soluble HLA class I molecules is Fas ligand/Fas mediated: evidence for the involvement of p56lck, calcium calmodulin kinase II, and Calcium‐independent protein kinase C signaling pathways and for NF‐kappaB and NF‐AT nuclear translocation. J Immunol 2005; 175:7244–54. [DOI] [PubMed] [Google Scholar]
  • 47. Kanai T, Fujii T, Kozuma S et al Soluble HLA‐G influences the release of cytokines from allogeneic peripheral blood mononuclear cells in culture. Mol Hum Reprod 2001; 7:195–200. [DOI] [PubMed] [Google Scholar]
  • 48. Kanai T, Fujii T, Unno N et al Human leukocyte antigen‐G‐expressing cells differently modulate the release of cytokines from mononuclear cells present in the decidua versus peripheral blood. Am J Reprod Immunol 2001; 45:94–9. [DOI] [PubMed] [Google Scholar]
  • 49. LeMaoult J, Caumartin J, Daouya M et al Immune regulation by pretenders: cell‐to‐cell transfers of HLA‐G make effector T cells act as regulatory cells. Blood 2007; 109:2040–8. [DOI] [PubMed] [Google Scholar]
  • 50. Feger U, Tolosa E, Huang YH et al HLA‐G expression defines a novel regulatory T‐cell subset present in human peripheral blood and sites of inflammation. Blood 2007; 110:568–77. [DOI] [PubMed] [Google Scholar]
  • 51. Carosella ED, Gregori S, LeMaoult J. The tolerogenic interplay(s) among HLA‐G, myeloid APCs, and regulatory cells. Blood 2011; 118:6499–505. [DOI] [PubMed] [Google Scholar]
  • 52. Huang YH, Zozulya AL, Weidenfeller C et al Specific central nervous system recruitment of HLA‐G(+) regulatory T cells in multiple sclerosis. Ann Neurol 2009; 66:171–83. [DOI] [PubMed] [Google Scholar]
  • 53. Wastowski IJ, Sampaio‐Barros PD, Amstalden EM et al HLA‐G expression in the skin of patients with systemic sclerosis. J Rheumatol 2009; 36:1230–4. [DOI] [PubMed] [Google Scholar]
  • 54. Favoino E, Favia IE, Vettori S et al Clinical correlates of human leucocyte antigen (HLA)‐G in systemic sclerosis. Clin Exp Immunol 2015; 181:100–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. van den Hoogen F, Khanna D, Fransen J et al 2013 classification criteria for systemic sclerosis: an American College of Rheumatology/European League against Rheumatism collaborative initiative. Arthritis Rheum 2013; 65:2737–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Rudstein‐Svetlicky N, Loewenthal R, Horejsi V, Gazit E. HLA‐G levels in serum and plasma. Tissue Antigens 2006; 67:111–6. [DOI] [PubMed] [Google Scholar]
  • 57. McIntire RH, Morales PJ, Petroff MG, Colonna M, Hunt JS. Recombinant HLA‐G5 and ‐G6 drive U937 myelomonocytic cell production of TGF‐beta1. J Leukoc Biol 2004; 76:1220–8. [DOI] [PubMed] [Google Scholar]
  • 58. Smith EA, LeRoy EC. A possible role for transforming growth factor‐beta in systemic sclerosis. J Invest Dermatol 1990; 95:125S–7S. [DOI] [PubMed] [Google Scholar]
  • 59. Kawakami T, Ihn H, Xu W, Smith E, LeRoy C, Trojanowska M. Increased expression of TGF‐beta receptors by scleroderma fibroblasts: evidence for contribution of autocrine TGF‐beta signaling to scleroderma phenotype. J Invest Dermatol 1998; 110:47–51. [DOI] [PubMed] [Google Scholar]
  • 60. Dantas AT, Goncalves SM, de Almeida AR et al Reassessing the role of the active TGF‐beta1 as a biomarker in systemic sclerosis: association of serum levels with clinical manifestations. Dis Markers 2016; 2016:6064830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Abraham DJ, Eckes B, Rajkumar V, Krieg T. New developments in fibroblast and myofibroblast biology: implications for fibrosis and scleroderma. Curr Rheumatol Rep 2007; 9:136–43. [DOI] [PubMed] [Google Scholar]
  • 62. Yang Y, Chu W, Geraghty DE, Hunt JS. Expression of HLA‐G in human mononuclear phagocytes and selective induction by IFN‐gamma. J Immunol 1996; 31:231. [PubMed] [Google Scholar]
  • 63. Pankratz S, Bittner S, Herrmann AM et al Human CD4+ HLA‐G+ regulatory T cells are potent suppressors of graft‐versus‐host disease in vivo . FASEB J 2014; 28:3435–45. [DOI] [PubMed] [Google Scholar]
  • 64. Pankratz S, Ruck T, Meuth SG, Wiendl H. CD4(+)HLA‐G(+) regulatory T cells: molecular signature and pathophysiological relevance. Hum Immunol 2016; 77:727–33. [DOI] [PubMed] [Google Scholar]
  • 65. Overgaard NH, Jung JW, Steptoe RJ, Wells JW. CD4+/CD8+ double‐positive T cells: more than just a developmental stage? J Leukoc Biol 2015; 97:31–8. [DOI] [PubMed] [Google Scholar]
  • 66. Wu Y, Cai B, Feng W et al Double positive CD4+CD8+ T cells: key suppressive role in the production of autoantibodies in systemic lupus erythematosus. Indian J Med Res 2014; 140:513–9. [PMC free article] [PubMed] [Google Scholar]

Articles from Clinical and Experimental Immunology are provided here courtesy of British Society for Immunology

RESOURCES