Primary cutaneous lymphomas (CLs) constitute a spectrum of diseases characterized by a clonal accumulation of lymphocytes in the skin. Most CLs display a Th2 cytokine profile, including expression of interleukin-10 (IL-10). Because the up-regulation of HLA-G, a nonclassical class Ib molecule inducible by IL-10, might account for the immunescape of the malignant clone, HLA-G and IL-10 expression has been investigated in 45 cases of primary CL (10 of B-cell and 35 of T-cell origin) with quantitative polymerase chain reaction (PCR) and immunohistochemistry. HLA-G message was present in all cutaneous B-cell (CBCL) and T-cell (CTCL) lymphomas evaluated. Immunohistochemistry revealed HLA-G protein expression in 23 (51%) of 45 cases (7 of 10 CBCL, 16 of 35 CTCL). While in CBCL mostly indolent types displayed HLA-G positivity, in CTCL HLA-G expression was associated with high-grade histology and advanced stage of the disease. Except for neoplastic and infiltrating lymphocytes, other cells such as macrophages and dendritic cells showed HLA-G immunoreactivity. Furthermore, IL-10 protein expression was demonstrated in 16 (73%) of 22 HLA-G+ cases, which correlated with HLA-G protein presence (P < .001). HLA-G up-regulation together with IL-10 expression in CL might additionally contribute to the evasion of immunosurveillance and facilitate the transition from low- to high-grade lymphomas.

Primary cutaneous lymphomas (CLs) comprise a spectrum of heterogeneous entities characterized by a clonal accumulation of lymphocytes initially restricted to the skin.1 Despite their rather benign course, cutaneous T-cell (CTCLs) and B-cell lymphomas (CBCLs) may spread to extracutaneous sites, confirming that they are systemic disease of the lymphoid system despite their clinical confinement to the skin.2 Decreased cell-mediated cytotoxity (in both natural killer [NK] cell and lymphokine-activated killer cell activity), decreased T-cell response to antigens, and several other immune phenomena imply the existence of general immune dysregulation in CTCL patients.3,4 Most of the abnormalities might be attributed to the pleiotropic immunosuppressive effects of interleukin-10 (IL-10), as demonstrated in different CL types.5-8 

Despite the evidence of antitumor humoral and cell-mediated immune response in CL,9-11 the disease may evolve further, eventually killing the patient. Besides the production of immunosuppressive cytokines, alterations of HLA class I status responsible for the presentation of tumor peptides to T cells could be the reason for an ineffective antitumor response.12 So far only a few studies have been conducted investigating expression of HLA class I molecules, mainly in nodal lymphomas.13-15 The loss of major histocompatibility complex class I antigens was detected preferentially in large cell lymphomas and in total was less frequent (2.3%) in comparison to nonhematologic cancer (up to 50%).12 15 Therefore, this mechanism is less likely to take place in lymphomas.

The expression of one nonclassical HLA class I molecule, HLA-G, has been implicated as an additional way of tumor immunescape.16-19 HLA-G differs from other HLA class I molecules—hence the prefix “nonclassical”—by its quasimonomorphism and distribution restricted to immune privileged sites, such as fetomaternal interface and thymus.20,21 The HLA-G gene is transcribed in a variety of human adult tissues, peripheral blood B and T lymphocytes, and monocytes but not NK cells.21-25 Additionally, alternative splicing creates at least 7 different HLA-G isoforms, of which 4 are membrane-bound and 3 are coding for soluble proteins.21,26 The shift toward Th2 cytokine profile, dominated by IL-10, is thought to be responsible for HLA-G expression in placenta.27,28Initially, HLA-G was shown to protect from NK-mediated lysis of HLA-deficient targets through interactions with killer cell inhibitory receptors (KIR).20,29,30 The fact that other cells, such as lymphocytes, macrophages, and dendritic cells, harbor HLA-G–specific inhibitory ligands, such as p49, ILT-2, and ILT-4,31-33 explained HLA-G–mediated inhibition of a cytotoxic T-lymphocyte–specific response.34 HLA-G leader sequence-derived peptide stabilizes cell surface expression of another nonclassical class I molecule, HLA-E, which constitutes a preferential ligand for a widely expressed KIR, CD94/NKG2A.35Intratumoral-activated macrophages and dendritic cells have been described recently to express HLA-G protein,36 suggesting that HLA-G–mediated immune modulation might further contribute to the impairment and circumvention of an efficient immune response.

Because lymphocytes have the potential to transcribe and express HLA-G depending on the environmental conditions, we investigated HLA-G expression in primary CLs. We report here that HLA-G message and protein were expressed in both cutaneous CBCL and CTCL. In CBCL, mostly indolent types displayed HLA-G immunoreactivity, while in CTCL HLA-G expression was associated with large cell morphology and advanced stage of the disease. In addition, we have demonstrated IL-10 immunoreactivity in the proximity of HLA-G+ cells, suggesting that IL-10 was responsible for HLA-G up-regulation in CL.

Tissue and blood samples; cell lines

Skin biopsy samples from 45 patients with primary CLs were taken primarily for diagnostic purposes after obtaining a prior informed consent of the patient. The specimens used in further investigations were a surplus material available after all routine diagnostic procedures. The healthy skin specimens were obtained from the Department of Plastic Surgery, University Hospital Zurich following breast reduction. The histologic diagnoses are summarized in Table 1 and were assessed in agreement with the European Organization for Research and Treatment of Cancer (EORTC) classification of primary CLs.37 The assignment to a certain stage was done according to the recommendations of EORTC Cutaneous Lymphoma Project Group.37 Peripheral blood mononuclear cells (PBMCs) were isolated from patients' blood and buffy coat preparation from healthy volunteers (obtained from a local blood bank) using a Ficoll-Hypaque density gradient centrifugation (Pharmacia Biotech, Uppsala, Sweden). Skin biopsy samples and PBMCs from 2 patients (CO) with secondary CBCL were included as controls. The HLA-G+ human choriocarcinoma cell line JEG-3 (ATCC, Manassas, VA) was cultured in Dulbecco modified Eagle medium (Biochrom, Berlin, Germany) supplemented with 10% heat-inactivated fetal calf serum with antibiotics-antimycotic (Gibco BRL, Life Technologies AG, Basel, Switzerland), 1 mM sodium pyruvate (Gibco BRL), and 2 mM L-glutamine (Biochrom). First trimester trophoblast tissue was obtained from elective termination of pregnancy after informed consent of the patient and used as an additional positive control.

Table 1.

Data of the CL patients evaluated for HLA-G and IL-10 protein expression

Patient no.GenderAge, yHistologic classification (EORTC)StageHLA-G protein expressionIL-10
expression
BL1 75 Follicular B-cell lymphoma  ++ 
BL2 32 Follicular B-cell lymphoma  ++ − 
BL3 19 Follicular B-cell lymphoma  − 
BL4 59 Follicular B-cell lymphoma  ++ 
BL5 58 MZBCL  ++ ++ 
BL6 82 Diffuse large B-cell lymphoma  
BL7 81 Diffuse large B-cell lymphoma  − ND 
BL8 49 Diffuse large B-cell lymphoma  ND 
BL9 77 Large B-cell lymphoma of the leg  − − 
BL10 57 T-cell-rich B-cell lymphoma  − − 
TL1 36 Lymphomatoid papulosis  − − 
TL2 45 Lymphomatoid papulosis  − − 
TL3 65 Lymphomatoid papulosis  − − 
TL4 54 Lymphomatoid papulosis  − − 
TL5 52 Lymphomatoid papulosis  − − 
TL6 40 MF Ia − − 
TL7 56 MF Ib − − 
TL8 87 MF Ib − − 
TL9 64 MF Ib − − 
TL10 66 MF IIb − − 
TL11 74 MF IIb − − 
TL12 52 MF IIb − − 
TL13 55 MF III ++ ++ 
TL14 77 MF IVb 
TL15 87 Sézary syndrome IVa − − 
TL16 45 CD8+ initial CTCL Ia 
TL17 50 CD8+, TIA-1+, CD30 large cell CTCL IIb ++ ++ 
TL18 68 CD30+ large cell CTCL IIb −  
TL19 56 CD30+ large cell CTCL IIb − −  
TL20 67 CD30+large cell CTCL IIb − 
TL21 29 CD30+ large cell CTCL IIb − −  
TL22 45 CD30+ large cell CTCL IIb − −  
TL23 33 CD30large cell CTCL IIb − − 
TL24 18 CD30 large cell CTCL IIb ++ −  
TL25 65 CD30 large cell CTCL IIb ++ 
TL26 76 CD30 large cell CTCL IVb ++ ++ 
TL27 36 CD30 large cell CTCL IVb − − 
TL28 50 CD30 large cell CTCL IVb ++ 
TL29 69 CD30+ pleomorphic CTCL IIb − −  
TL30 62 CD30+pleomorphic CTCL IVa 
TL31 52 CD30 pleomorphic small/medium-sized CTCL IIb ++ 
TL32 80 CD30 pleomorphic small/medium-sized CTCL IIb 
TL33 62 CD30 pleomorphic small/medium-sized CTCL IIb 
TL34 50 CD30 pleomorphic small/medium-sized CTCL IVb − 
TL35 28 GSS IIb ++ ++ 
Patient no.GenderAge, yHistologic classification (EORTC)StageHLA-G protein expressionIL-10
expression
BL1 75 Follicular B-cell lymphoma  ++ 
BL2 32 Follicular B-cell lymphoma  ++ − 
BL3 19 Follicular B-cell lymphoma  − 
BL4 59 Follicular B-cell lymphoma  ++ 
BL5 58 MZBCL  ++ ++ 
BL6 82 Diffuse large B-cell lymphoma  
BL7 81 Diffuse large B-cell lymphoma  − ND 
BL8 49 Diffuse large B-cell lymphoma  ND 
BL9 77 Large B-cell lymphoma of the leg  − − 
BL10 57 T-cell-rich B-cell lymphoma  − − 
TL1 36 Lymphomatoid papulosis  − − 
TL2 45 Lymphomatoid papulosis  − − 
TL3 65 Lymphomatoid papulosis  − − 
TL4 54 Lymphomatoid papulosis  − − 
TL5 52 Lymphomatoid papulosis  − − 
TL6 40 MF Ia − − 
TL7 56 MF Ib − − 
TL8 87 MF Ib − − 
TL9 64 MF Ib − − 
TL10 66 MF IIb − − 
TL11 74 MF IIb − − 
TL12 52 MF IIb − − 
TL13 55 MF III ++ ++ 
TL14 77 MF IVb 
TL15 87 Sézary syndrome IVa − − 
TL16 45 CD8+ initial CTCL Ia 
TL17 50 CD8+, TIA-1+, CD30 large cell CTCL IIb ++ ++ 
TL18 68 CD30+ large cell CTCL IIb −  
TL19 56 CD30+ large cell CTCL IIb − −  
TL20 67 CD30+large cell CTCL IIb − 
TL21 29 CD30+ large cell CTCL IIb − −  
TL22 45 CD30+ large cell CTCL IIb − −  
TL23 33 CD30large cell CTCL IIb − − 
TL24 18 CD30 large cell CTCL IIb ++ −  
TL25 65 CD30 large cell CTCL IIb ++ 
TL26 76 CD30 large cell CTCL IVb ++ ++ 
TL27 36 CD30 large cell CTCL IVb − − 
TL28 50 CD30 large cell CTCL IVb ++ 
TL29 69 CD30+ pleomorphic CTCL IIb − −  
TL30 62 CD30+pleomorphic CTCL IVa 
TL31 52 CD30 pleomorphic small/medium-sized CTCL IIb ++ 
TL32 80 CD30 pleomorphic small/medium-sized CTCL IIb 
TL33 62 CD30 pleomorphic small/medium-sized CTCL IIb 
TL34 50 CD30 pleomorphic small/medium-sized CTCL IVb − 
TL35 28 GSS IIb ++ ++ 

− indicates no expression detectable with 4H84 mAb, accordingly for IL-10; +, single-cell immunoreactivity with 4H84 mAb, accordingly for IL-10; ++, strong immunoreactivity with 4H84 mAb, accordingly for IL-10; and ND, not done.

*

Skin material was available for quantitative PCR.

PBMCs were available for quantitative PCR.

Transformed from MF.

RNA extraction; cDNA synthesis

Total RNA was extracted from 17 frozen tissue samples using a High Pure RNA Tissue Kit and from PBMCs of 10 patients using a High Pure RNA Isolation Kit (both from Roche Molecular Biochemicals [RMB], Mannheim, Germany) according to the manufacturer's recommendations. Approximately 1 μg of RNA was reverse transcribed using oligo-p(dT)15 priming and avian myeloblastosis virus reverse transcriptase (1st Strand cDNA Synthesis Kit for RT-PCR [avian myeloblastosis virus], RMB) at 42°C for 1 hour. Complementary DNAs (cDNAs) were then stored at −20°C until further use.

Real-time quantitative PCR

HLA-G–specific polymerase chain reaction (PCR) amplifications were carried out using following primer sets: U522 (exon 3) and L922 (exon 5) detecting mainly HLA-G1 isoform; G.257(exon 2) and G.1225 (3′-UT) screening for all alternatively spliced HLA-G messenger RNA (mRNA) forms, as previously described38 (Table2). Design of the primers and optimization for the use in the LightCycler quantification system was done with the OLIGO 5.0 Primer Analysis Software (Molecular Biology Insights, Cascade, CO). Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) primers and plasmids pCRII.GAPDH were kindly provided by Andreas Trojan (Division of Oncology, Department of Internal Medicine, University Hospital Zurich, Switzerland). A Hot-Start PCR was performed using 2 μL ready-to-use mastermix (LightCycler-Faststart DNA Master SYBR Green I, RMB, containing termostable recombinant Taq polymerase, reaction buffer, dATP, dCTP, dGTP, dUTP), 0.5 μM of each oligonucleotide primer (desalted PCR grade, Microsynth, Balgach, Switzerland), variable free MgCl2 concentrations (Table 2), 2 μL cDNA, and a water to a final volume of 20 μL. After initial denaturation of 7 minutes, amplification occurred as a 3-step cycling procedure: denaturation, 95°C, 15 seconds, ramp rate 20°C per second; annealing (Table 2), 10 seconds, ramp rate 20°C per second; and elongation at 72°C (Table 1), ramp rate 2°C per second, for 40 cycles. The acquisition of fluorescence was done at 87°C to avoid contribution of nonspecific products to the overall signal. Finally, the temperature was raised gradually (0.2°C per second) starting from 70°C to 99°C for the melting curve analysis

Table 2.

Used oligonucleotides and PCR conditions

PrimersSequenceAmplicon size, base pairsAnnealing temperature, elongation timeFree MgCl2
U522 (exon 3) 5′-CAA TGT GGC TGA ACA AAG GA-3′ 420 60°C, 25 s 4 mM 
L922 (exon 5) 5′-CCA GCA ACG ATA CCC ATG-3′    
G.257 (exon 2) 5′-GGA AGA GGA GAC ACG GAA CA-3′ G1-988; G2,4-712; G3-436; G5-1110; G6-834 60°C, 90 s 2 mM  
G.1225 (3′-UT) 5′-TGA GAC AGA GAC GGA GAC AT-3′    
GAP1U19 5′-GAA GGT GAA GGT CGG AGT C-3′ 224 55°C, 10 s 4 mM  
GAP206L19 5′-GAA GAT GGT GAT GGG ATT T-3′    
PrimersSequenceAmplicon size, base pairsAnnealing temperature, elongation timeFree MgCl2
U522 (exon 3) 5′-CAA TGT GGC TGA ACA AAG GA-3′ 420 60°C, 25 s 4 mM 
L922 (exon 5) 5′-CCA GCA ACG ATA CCC ATG-3′    
G.257 (exon 2) 5′-GGA AGA GGA GAC ACG GAA CA-3′ G1-988; G2,4-712; G3-436; G5-1110; G6-834 60°C, 90 s 2 mM  
G.1225 (3′-UT) 5′-TGA GAC AGA GAC GGA GAC AT-3′    
GAP1U19 5′-GAA GGT GAA GGT CGG AGT C-3′ 224 55°C, 10 s 4 mM  
GAP206L19 5′-GAA GAT GGT GAT GGG ATT T-3′    

External standards for the GAPDH quantification consisted of 6 serial 1:10 dilutions (5 × 102 to 5 × 107molecules per reaction) of pCRII.GAPDH plasmids. Plasmids containing full-length HLA-G insert pLNCX.G139 were used as 6 external standards for HLA-G1 quantitative PCR, containing 102 to 107 copies per reaction (a kind gift from D.E. Geraghty, Fred Hutchinson Cancer Research Center, Seattle, WA). External standards were run concomitantly with patient samples under identical conditions. The PCR reactions were run as triplicates, and run data were analyzed with the quantification program V3.39 (RMB). The fluorescence signal was plotted against the cycle number for all samples and external standards. The Fit Points method option was used in the course of analyzing quantification data, allowing the definition of a noise band and subsequent background fluorescence subtraction and resulting in the display of only log-linear and plateau amplification phases. The point when the signal rose above the background level, the so-called crossing point, was then determined for each standard dilution. The standard curve was then generated for each run, plotting the crossing point against the log concentration of the standards. The mean crossing point value, SD, and coefficient of variation were than calculated for each standard and patient sample. The single HLA-G1 and GAPDH standard curves as a result of triplicate runs were then used as references to calculate the number of target molecules in each sample. Results were expressed initially as the absolute copy number per microliter. Normalization of the estimated HLA-G1 amount was achieved by calculating the ratios between HLA-G1 and GAPDH copy number in 1 μL cDNA to compensate the variations in quantity and quality of starting mRNA.40 The normalized values were then multiplied by the constant 104.

Ten microliters of each amplification product was analyzed for appropriate length by electrophoresis on 1.6% agarose gel stained with ethidium bromide. The estimated size of the amplified fragments matched the calculated size. In addition, product identity was confirmed by melting curve analysis, an application in the LightCycler analysis program. The specificity of the obtained PCR products was finally confirmed by sequencing.

Immunohistochemistry

The paraffin-embedded material was available for all 27 cases. The following antibodies were used: 4H84 (1:500 dilution), IgG1 mouse monoclonal antibody (mAb) raised against denatured HLA-G α1 domain (kindly provided by M. McMaster, University of California, San Francisco, CA); anti–major histocompatibility complex class I mAb TP25.99, reacting with HLA-A, -B, and -C but not HLA-G (kind gift from S. Ferrone, Roswell Park Cancer Institute, Buffalo, NY); anti–human IL-10 (E-10) mouse IgG2b mAb (Santa Cruz Biotechnology, Santa Cruz, CA); anti–human CD79a mouse IgG1 mAb and anti–human CD3 mouse polyclonal antibody (both from DAKO, Denmark); anti–human CD1a mouse IgG1 mAb (Immunotech, Marseille, France); and isotype-matched controls IgG1, IgG2a, and IgG2b (DAKO). Imunohistochemical staining was performed using the alkaline phosphatase–antialkaline phosphatase (APAAP) technique after microwave antigen recovery. To determine whether HLA-G expression coincides with IL-10 presence and to further determine the phenotype of HLA-G+ cells, paraffin-embedded tissue sections were double-stained with anti–IL-10 or anti-CD79a, -CD3, and anti-CD1a mAb and 4H84 (1:300 dilution) antibody employing the APAAP method. Briefly, after deparaffinization and antigen retrieval, nonspecific binding sites were blocked by incubating slides with 20% AB serum/phosphate–buffered saline for at least 15 minutes at room temperature. Tissue sections were incubated with different primary antibodies or isotype-matched control and, secondarily, always with 4H84 mAb for 60 minutes. Each specific antibody application was followed by 2 cycles of sequential incubations with rabbit anti–mouse IgG and APAAP complexes (DAKO). The immunoreaction was visualized with developing solutions containing blue-purple 5-bromo-4-chloro-3-indoxyl phosphate with nitroblue tetrazolium chloride (DAKO), which labeled primary antibody and red neufuchsin (DAKO) marking HLA-G. Finally, sections were counterstained with hematoxylin. All the incubations were performed at room temperature in a moist chamber.

Data analysis

Statistical analysis was performed using an SPSS statistical software (version 10.0; SPSS).

Differential HLA-G gene expression in primary CLs

Amplification with the G.257/G.1225 primer pair screening for all HLA-G mRNA-spliced isoforms demonstrated variable HLA-G gene expression in all lymphomatous skin lesions available for reverse transcriptase (RT)–PCR as well as in nonlesional skin of 7 patients and healthy skin samples (Figure 1A). Furthermore, the full-length transcript HLA-G1 was present in all cases investigated (Figure 2A). Quantitative RT-PCR revealed up to 2 orders of magnitude difference in HLA-G1 transcriptional levels between lymphoma cases, being most pronounced in patients with mycosis fungoides (MF) (Figure 2A). The HLA-G gene expression in lymphoma samples did not statistically differ from the uninvolved skin group and from the healthy skin. Although the CTCL group transcribed HLA-G more abundantly in comparison to the CBCL group, the difference was not statistically significant and the correlation with the particular lymphoma subtype could not be established. When comparing HLA-G transcription grouped according to the pattern of protein expression evaluated by immunohistochemistry (Figure 2B), HLA-G1 transcription in cases negative for HLA-G protein expression was significantly higher from the ones expressing HLA-G (Kruskal- Wallis test, H = 9.167, P = .010). In addition, the absence of HLA-G immunoreactivity correlated with high HLA-G1 mRNA levels (one-tailed Spearman rank correlation test, rho = −.490,P = .023).

Fig. 1.

Results after RT-PCR amplification with G.257/G.1225 primer pair screening all HLA-G isoforms.

Results are shown in (A) lesional and nonlesional skin and in (B) PBMCs of CL patients. Due to the low sensitivity of ethidium bromide staining, for some samples the bands are not visible. Using a melting curve analysis (data not shown) and the primer pair amplifying HLA-G1, it was possible to confirm that all the cases were transcribing HLA-G. The cDNAs from JEG-3 cell line and first trimester placenta (Pla) were used as positive controls. Instead of cDNA template, water (–) was run as a negative control; hs indicates healthy skin; l, lesional skin; n, nonlesional skin; BC, buffy coat preparation; and MWM, molecular weight marker.

Fig. 1.

Results after RT-PCR amplification with G.257/G.1225 primer pair screening all HLA-G isoforms.

Results are shown in (A) lesional and nonlesional skin and in (B) PBMCs of CL patients. Due to the low sensitivity of ethidium bromide staining, for some samples the bands are not visible. Using a melting curve analysis (data not shown) and the primer pair amplifying HLA-G1, it was possible to confirm that all the cases were transcribing HLA-G. The cDNAs from JEG-3 cell line and first trimester placenta (Pla) were used as positive controls. Instead of cDNA template, water (–) was run as a negative control; hs indicates healthy skin; l, lesional skin; n, nonlesional skin; BC, buffy coat preparation; and MWM, molecular weight marker.

Close modal
Fig. 2.

Relative HLA-G1 transcriptional levels in primary CLs.

Values are expressed as mean ± SD GAPDH-balanced ratios (as a result of triplicate runs) multiplied by 104. (A) Pattern of HLA-G1 expression in lesional (n = 17) versus nonlesional (n = 7) and a control healthy skin. The Student t test was performed; t = 2.54, P = .022. (B) Relative HLA-G1 transcriptional levels grouped according to the immunoreactivity with 4H84 mAb. Levels of HLA-G1 mRNA expression in cases showing no 4H84 immunoreactivity (n = 7) versus single-cell positivity (n = 5) versus HLA-G protein–positive lymphoma cases (n = 4). In some cases HLA-G immunoreactivity coincided with IL-10 expression (▾). NL indicates nonlesional skin.

Fig. 2.

Relative HLA-G1 transcriptional levels in primary CLs.

Values are expressed as mean ± SD GAPDH-balanced ratios (as a result of triplicate runs) multiplied by 104. (A) Pattern of HLA-G1 expression in lesional (n = 17) versus nonlesional (n = 7) and a control healthy skin. The Student t test was performed; t = 2.54, P = .022. (B) Relative HLA-G1 transcriptional levels grouped according to the immunoreactivity with 4H84 mAb. Levels of HLA-G1 mRNA expression in cases showing no 4H84 immunoreactivity (n = 7) versus single-cell positivity (n = 5) versus HLA-G protein–positive lymphoma cases (n = 4). In some cases HLA-G immunoreactivity coincided with IL-10 expression (▾). NL indicates nonlesional skin.

Close modal

Further on, we sought to compare HLA-G transcriptional levels from the lesional skin with the one in patients' PBMCs, presuming that the malignant clone would be preferentially homing to the skin and exhibit difference in HLA-G gene expression due to the influence of microenvironmental factors. Again, HLA-G isoform screening revealed the positivity of all cases (Figure 1B). The full-length transcript was present in all cases within the patient group, varying significantly (Student t test, t = 2.693,P = .021), and in PBMCs obtained from a buffy coat preparation (Figure 3A). The levels of HLA-G1 mRNA did not statistically differ between the patient group and buffy coat sample. Of note, patients' own PBMCs demonstrated an average of 5 times higher HLA-G1 transcription than the lesional skin (Figure 3B). Interestingly, 4 MF patients (TL6, TL9, TL11, TL12) exhibited an average of 4 times higher levels of HLA-G1 in the skin (Figure3B). None of the 10 patients evaluated had a detectable B-cell or T-cell clone in the blood.

Fig. 3.

Relative HLA-G1 mRNA levels in PBMCs obtained from primary CL patients.

(A) Values are expressed as mean ± SD GAPDH-balanced ratios (as a result of triplicate runs) multiplied by 104. BC indicates buffy coat. (B) Relative HLA-G1 transcriptional levels in lesional skin and PBMCs of primary CL patients. Note that 4 patients with CTCL (TL6, TL9, TL11, TL12) exhibited an average 4 times higher HLA-G transcription in lesional skin than in PBMCs.

Fig. 3.

Relative HLA-G1 mRNA levels in PBMCs obtained from primary CL patients.

(A) Values are expressed as mean ± SD GAPDH-balanced ratios (as a result of triplicate runs) multiplied by 104. BC indicates buffy coat. (B) Relative HLA-G1 transcriptional levels in lesional skin and PBMCs of primary CL patients. Note that 4 patients with CTCL (TL6, TL9, TL11, TL12) exhibited an average 4 times higher HLA-G transcription in lesional skin than in PBMCs.

Close modal

HLA-G protein is expressed preferentially in indolent CBCL or in high-grade CTCL

Immunohistochemical staining with 4H84 mAb showed HLA-G protein up-regulation in 23 (51%) of 45 cases (7 of 10 CBCL, 16 of 35 CTCL) (Table 1). According to the number of HLA-G–expressing cells, observed HLA-G positivity was described as a strong (numerous cells dispersed within the malignant infiltrate) or as a single-cell positivity (scant, scattered cells throughout the infiltrate). Correspondingly, strong HLA-G expression could be detected in half of the positive cases in both lymphoma groups (6 of 10 CBCL, 8 of 16 CTCL). None of the samples displayed alterations in total HLA class I expression, evaluated with TP25.99 mAb.

No particular pattern of HLA-G expression could be elucidated. However, in CL types prone to create the structures similar to their physiologic counterparts, such as follicular center cell lymphoma (FCCL) or marginal zone B-cell lymphoma (MZBCL), the presence of HLA-G was restricted to the mantle zone of pseudofollicular structures (Figures 4B-C). The tendency of HLA-G+ cells toward a particular dermal compartment was not observed, although there was occasional close contact with epidermis (Figure 4A).

Fig. 4.

HLA-G immunoreactivity in primary CLs.

(A) FCCL (patient BL2, original magnification [OM] × 100). (B) FCCL (patient BL1, OM × 160). (C) MZBCL (patient BL5, OM × 100). (D) Large cell CD30, CD8+, TIA-1+ CTCL (patient TL17, OM × 200). (E) GSS (patient TL135., OM × 100). (F) A giant multinucleated HLA-G+ cell in GSS (OM × 200). (G) Double immunostaining for HLA-G (red) and CD79a (blue-purple) in FCCL (patient BL4, OM × 630). (H) Double immunolabeling of HLA-G (red) and CD3 (blue-purple) in FCCL (patient BL4, OM × 630). (I) HLA-G (red) and CD3 (blue-purple) immunoreactivity in CTCL (patient TL135., OM × 630). (J) Expression of HLA-G (red) and IL-10 (blue-purple) in CTCL, MF (patient TL13, OM × 200). Colocalization of HLA-G/IL-10 creates a mixed violet-purple pigment, as shown also in panel K (patient TL17, OM × 400). (L) Double immunostaining of HLA-G (red) and CD1a (blue-purple) in CTCL (patient TL4, OM × 400).

Fig. 4.

HLA-G immunoreactivity in primary CLs.

(A) FCCL (patient BL2, original magnification [OM] × 100). (B) FCCL (patient BL1, OM × 160). (C) MZBCL (patient BL5, OM × 100). (D) Large cell CD30, CD8+, TIA-1+ CTCL (patient TL17, OM × 200). (E) GSS (patient TL135., OM × 100). (F) A giant multinucleated HLA-G+ cell in GSS (OM × 200). (G) Double immunostaining for HLA-G (red) and CD79a (blue-purple) in FCCL (patient BL4, OM × 630). (H) Double immunolabeling of HLA-G (red) and CD3 (blue-purple) in FCCL (patient BL4, OM × 630). (I) HLA-G (red) and CD3 (blue-purple) immunoreactivity in CTCL (patient TL135., OM × 630). (J) Expression of HLA-G (red) and IL-10 (blue-purple) in CTCL, MF (patient TL13, OM × 200). Colocalization of HLA-G/IL-10 creates a mixed violet-purple pigment, as shown also in panel K (patient TL17, OM × 400). (L) Double immunostaining of HLA-G (red) and CD1a (blue-purple) in CTCL (patient TL4, OM × 400).

Close modal

Concerning the type of CL predominantly expressing HLA-G, in CBCL indolent types (FCCL and MZBCL) demonstrated prominent HLA-G protein expression (Figure 4C). Within the CTCL group, MF cases of a stage ranging from Ia to IIb were clearly negative for HLA-G. Conversely, in MF patients with advanced stage (III-IVb) as well as in patients with CD30- large or pleomorphic CTCL (Figure 4D,J), HLA-G immunoreactivity was a consistent phenomenon (Table 1). On the contrary, in CD30+ lesions HLA-G expression was either not detectable (lymphomatoid papulosis) or present as a rare, single-cell positivity (CD30+ large cell and pleomorphic lymphoma).

HLA-G protein expression was observed in case of granulomatous slack skin (GSS), a rare type of CTCL characterized by a dense granulomatous CD4+ T-cell infiltrate and by the presence of multinucleated giant cells involved in the destruction of elastic tissue (Figure 4E). Additionally, 2 cases of CTCL showing cytotoxic, CD8+ phenotype displayed a discrete but noticeable HLA-G protein presence.

We performed a series of double staining, labeling HLA-G together with common B-cell (CD79a) and T-cell (CD3) markers to characterize further HLA-G–expressing cells. In FCCL and MZBCL, small to medium-sized B lymphocytes expressed HLA-G, which was confirmed also by double staining with CD79a (Figure 4G). Moreover, infiltrating T cells (CD3+) were also shown to express HLA-G (Figure 4H). In diffuse large B-cell lymphoma, an additional HLA-G+ cell type was large cells with vesicular nuclei and prominent nucleoli. Macrophages and endothelial cells occasionally displayed HLA-G immunoreactivity independent of the CL histology type. Moreover, plasma cells were shown quite repeatedly to be HLA-G+ irrespective of the CL type. Whether it was an artifact or specific staining remains elusive.

In CTCL, overall scattered small to medium sized T-lymphocytes were the cells most frequently expressing HLA-G (Figure 4D,I,J). In large and pleomorphic CTCL, large and pleomorphic T cells, respectively, contributed to the HLA-G+ cell population (Figure 4D). A case of GSS demonstrated fine cytoplasmic HLA-G staining of multinucleated giant cells and accompanying spindle-shaped histiocytes in addition to scattered small lymphocytes (Figure 4E-F).

Simultaneous labeling of CD1a (Langerhans cells) and HLA-G demonstrated that HLA-G+ cells are often in close contact with CD1a-marked cells irrespective of the CL type. Apart from the morphology changes of Langerhans cells when they migrate to the dermis (more rounded, less dendritic in shape, decreased CD1a expression), we have noticed an occasional HLA-G coexpression (Figure 4L).

IL-10 expression coincides with HLA-G protein up-regulation

The intensity of IL-10 immunoreactivity was assessed according to the same criteria (see above) as for HLA-G. IL-10–producing cells were detected in 16 (73%) of 22 HLA-G+ CL cases (one-tailed Spearman rank correlation test, rho = . 761, P<.001) (Table 1). IL-10–secreting cells were either colabeled with HLA-G or located in the vicinity of HLA-G–expressing cells (Figure 4J-K). An inverse correlation between HLA-G1 transcription intensity and the expression of IL-10 could be demonstrated in the way that the highest HLA-G1 transcription was associated with absence of IL-10 in tumorous skin (one-tailed Spearman rank correlation test, rho = −.586,P = .007) (Figure 2B). Additionally, a statistical difference in HLA-G1 transcriptional levels could be shown between the groups exhibiting strong, single-cell, or no IL-10 expression (Kruskal-Wallis test, H = 6.954, P = .031).

Among the HLA class I antigens, 2 families are distinguishable, both sharing homology at the sequence level consistent with a broadly similar secondary structure.41 These 2 groups consist of ubiquitously expressed, highly polymorphic class Ia (classical) HLA-A, -B and -C molecules and selectively expressed, rather monomorphic class Ib (nonclassical) molecules, principally HLA-E, -F, -G, all of them having features making them quite distinct from the members of class Ia group.21,30,42 HLA-G has gained interest because of its involvement in tumor immunescape and progression.16,18,19 43 We have investigated HLA-G expression in different CL subtypes, presuming that its expression on one side could account for the lesions' immunologic “stealthiness” and, on the other side, be an additional feature involved in the transition from low- to high-grade lymphoma.

We have demonstrated that HLA-G message was present in lesional and uninvolved skin of CL patients as well as in normal skin. A case-to-case diversity of the isoforms detected is probably due to a malignant transformation itself and a different cellular environment.23,24 We have shown the full-length HLA-G transcript in all of the samples, confirming that this is the most commonly transcribed HLA-G form in PBMCs and keratinocytes.24,44 Only in situations in which the translation of the HLA-G1 protein is impaired (ie, individuals homozygous for the null allele HLA-G*0105N), the other shorter isoforms might serve as a “biological back-up” undertaking necessary HLA-G functions, although the HLA-G transcription and splicing are not affected.45 Therefore, we have focused our quantitative PCR primarily on this isoform.

Comparison of the HLA-G1 transcription intensity of lesional skin samples and nonlesional ones with healthy skin showed no statistical difference, possibly due to the abundant HLA-G message in keratinocytes in healthy skin.44 Because the lymphocytes are the predominant cell population within the lymphomatous lesions, the end PCR result is likely to reflect the transcription of the infiltrate rather than the one of solely active epidermal basal cell layer. On the other hand, observed significant case-to-case differences in HLA-G1 levels are probably reflecting the interindividual differences of patients, unique biology of the lesions, or various clinical stages of the disease. With respect to that, the CTCL group exhibited the highest HLA-G1 transcriptional levels, particularly low-stage MF, but not the HLA-G protein expression. The inverse correlation between the HLA-G transcription and HLA-G (or IL-10) protein expression might be in part explained by the effects that IL-10 exerts on HLA-G transcription and protein expression. Namely, after IL-10 stimulation monocytes up-regulated cell surface HLA-G expression, but the mRNA levels remained unchanged.46 On the other hand, IL-10–induced HLA-G expression in purified trophoblast cells was followed by a significant increase in HLA-G mRNA, suggesting a cell type–dependable use of different regulatory mechanisms, ie, transcriptional or posttranscriptional regulation of HLA-G expression.46-48Another explanation for high HLA-G1 mRNA levels in HLA-Gearly CTCL cases might be a different, interferon-γ–polarized cytokine profile in these lesions,49,50 knowing the capability of interferon-α to produce increased in HLA-G mRNA without protein up-regulation.25,51 52 It is conceivable that such a particular micromilieu could account for the difference between the skin and PBMC HLA-G1 transcriptional levels noted in 4 CTCL cases.

Irrespective of the CL type, the strongest HLA-G immunoreactivity accounted for the 20% of the cellular infiltrate. Two populations of cells expressing HLA-G were identified: lymphoid cells of B or T phenotype and cells of myeloid origin, such as macrophages and dendritic cells. Surprisingly, no change in total HLA class I pattern could be detected. This might be due to the central role of lymphocytes in immune response in a way that any change of HLA class I status would be a disadvantage, rendering them immediately susceptible to destruction. Moreover, coexpression of HLA-G on targets with intact HLA class I molecules would favor evasion of immunosurveillance, inhibiting NK- and cytotoxic T-lymphocyte–mediated antitumor response.53 54 

Shift from a Th1 to Th2 cytokine profile and subsequent expression of IL-10 has been associated with the disease progression and impairment of tumor immunosurveillance in different malignancies as well as in advanced MF.1,7,55Additionally, IL-10 has been shown to be an autocrine growth factor in B-cell lymphomas.8 56 In our study HLA-G immunoreactivity was often associated with IL-10 expression in CTCL and CBCL, implying that the IL-10 might be one of the factors participating in the up-regulation of HLA-G. Moreover, the induction of HLA-G might serve as an additional way that IL-10 modulates and abrogates the CL-associated immune response.

In CBCL, HLA-G–expressing cells seemed to avoid “germinal centers” and to reside in the mantle zone. The mantle zone is a region of intensive immune interactions, such as antigen presentation and immunomodulation, where HLA-G+ cells might be also involved, because HLA-G is capable of presenting peptides57,58 and modulating the release of cytokines in PBMCs upon direct contact.59,60 Indeed, in our CL cases dendritic cells were often in close contact with HLA-G–bearing cells; however, the nature of this interaction remains unclear. Occasionally in CBCL, and according to the distribution of infiltrate in CTCL, HLA-G–expressing cells were located often subepidermally. Epidermal keratinocytes are potent producers of various cytokines,61 62 which could have a profound influence on the malignant infiltrate as well as on the HLA-G up-modulation.

HLA-G expression in CTCL was clearly evident in MF patients with advanced stage or in ones that underwent large cell transformation and also in CD30 large cell CTCL. All of these entities are associated with aggressive clinical behavior and poor survival.37,63,64 CD30 CTCL with the predominance of small or medium-sized pleomorphic T cells has been described as a separate entity, due to its better prognosis.37 Accordingly, HLA-G expression in these cases was not so pronounced, suggesting its true up-regulation only in highly malignant conditions such as CD30 large cell lymphomas or after a large-cell transformation in MF. Another group of primary CL, primary cutaneous CD8+ cytotoxic CTCL, was recently shown to express an HLA-G ligand, KIR molecule ILT-2.65Concurrent expression of HLA-G (as shown in 2 CD8+CTCL cases in our study) and interaction with clonal and host KIRs66 could account for growth/survival advantage of the dominant clone.65 Together with a shift toward a Th2 cytokine profile, HLA-G induction might be responsible for the circumvention of host immune response and subsequent disease aggressiveness and progression. HLA-G could be contributing as well to the tumor-associated angiogenesis, as hypothesized in placenta,67,68 due to its consistent expression by endothelial cells in our study. Within the group of CD30+primary cutaneous lymphoproliferative disorders, HLA-G expression could not be detected in self-healing lymphomatoid papulosis, while in CD30+ large cell CTCL it was limited to a sparse, single-cell positivity. This might be understandable, knowing that primary large CTCL expressing CD30 antigen has a significantly better prognosis than CD30 large cell CTCL.37 69 

Apart from lymphoid cells, intralesional macrophages and dendritic cells displayed HLA-G immunoreactivity in many CL cases, initially shown in lung cancer.36 As a curiosity, large multinucleate giant cells in GSS, which are of monocyte-macrophage lineage,70 demonstrated strong HLA-G expression, a feature apparently inherent to monocyte lineage.71 72 

Given the fact that B and T lymphocytes as well as macrophages and dendritic cells bear KIRs (ILT-2, ILT-4, p49),66,73some of which have been shown to bind soluble HLA-G1 tetrameric complexes,74 an open question remains as to whether HLA-G immunoreactivity in these cells is a phenomenon per se or an artifact due to the binding of soluble HLA-G molecules (ie, HLA-G5, -G6, -G7) to these receptors, particularly because the antibodies specific to the soluble protein are not yet available for use in immunohistochemistry. This might account for a strong HLA-G positivity in indolent, low-grade CBCL and of plasma cells in general, knowing that ILT-2 is present virtually on all B cells and its expression changes during the course of B-cell differentiation, ie, going up as the cells mature.31,35 75 The dedifferentiation is probably the reason why the large cell CBCL displayed a weak HLA-G positivity.

In conclusion, we have demonstrated for the first time expression of HLA-G and its association with IL-10 production in different types of primary CLs. The distinct pattern of HLA-G expression in CL suggests that this nonclassical molecule might be either interfering with the mounting of an efficient tumor-associated immune response in CL or facilitating the transition from low- to high-grade lymphoma. Whereas the role of class Ia antigens in immune recognition is well understood both in structural and functional terms, in the field of nonclassical HLA molecules and their putative ligands many questions remain to be answered.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 U.S.C. section 1734.

1
Dummer
R
Willers
J
Kamarashev
J
Urosevic
M
Döbbeling
U
Burg
G
Pathogenesis of cutaneous lymphomas.
Semin Cutan Med Surg.
19
2000
78
86
2
Burg
G
Kempf
W
Haeffner
AC
et al
Cutaneous lymphomas.
Curr Probl Dermatol.
9
1997
137
204
3
Dummer
R
Kohl
O
Gillessen
J
Kagi
M
Burg
G
Peripheral blood mononuclear cells in patients with nonleukemic cutaneous T-cell lymphoma. Reduced proliferation and preferential secretion of a T helper-2-like cytokine pattern on stimulation.
Arch Dermatol.
129
1993
433
436
4
Rook
AH
Heald
P
The immunopathogenesis of cutaneous T-cell lymphoma.
Hematol Oncol Clin North Am.
9
1995
997
1010
5
Buhl
L
Sogaard
H
Immunohistochemical expression of IL-10 in mycosis fungoides.
Exp Dermatol.
6
1997
195
198
6
Dummer
R
Heald
PW
Nestle
FO
et al
Sezary syndrome T-cell clones display T-helper 2 cytokines and express the accessory factor-1 (interferon-gamma receptor beta-chain).
Blood.
88
1996
1383
1389
7
Asadullah
K
Docke
WD
Volk
HD
Sterry
W
Cytokines and cutaneous T-cell lymphomas.
Exp Dermatol.
7
1998
314
320
8
Asadullah
K
Gellrich
S
Haeussler Quade
A
et al
Cytokine expression in primary cutaneous germinal center cell lymphomas.
Exp Dermatol.
9
2000
71
76
9
Berger
CL
Longley
BJ
Imaeda
S
Christensen
I
Heald
P
Edelson
RL
Tumor-specific peptides in cutaneous T-cell lymphoma: association with class I major histocompatibility complex and possible derivation from the clonotypic T-cell receptor.
Int J Cancer.
76
1998
304
311
10
Berger
CL
Wang
N
Christensen
I
Longley
J
Heald
P
Edelson
RL
The immune response to class I-associated tumor-specific cutaneous T-cell lymphoma antigens.
J Invest Dermatol.
107
1996
392
397
11
Eichmuller
S
Usener
D
Dummer
R
Stein
A
Thiel
D
Schadendorf
D
Serological detection of cutaneous T-cell lymphoma-associated antigens.
Proc Natl Acad Sci U S A.
98
2001
629
634
12
Rees
RC
Mian
S
Selective MHC expression in tumours modulates adaptive and innate antitumour responses.
Cancer Immunol Immunother.
48
1999
374
381
13
List
AF
Spier
CM
Miller
TP
Grogan
TM
Deficient tumor-infiltrating T-lymphocyte response in malignant lymphoma: relationship to HLA expression and host immunocompetence.
Leukemia.
7
1993
398
403
14
Medeiros
LJ
Gelb
AB
Wolfson
K
et al
Major histocompatibility complex class I and class II antigen expression in diffuse large cell and large cell immunoblastic lymphomas. Absence of a correlation between antigen expression and clinical outcome.
Am J Pathol.
143
1993
1086
1097
15
Amiot
L
Onno
M
Lamy
T
et al
Loss of HLA molecules in B lymphomas is associated with an aggressive clinical course.
Br J Haematol.
100
1998
655
663
16
Fukushima
Y
Oshika
Y
Nakamura
M
et al
Increased expression of human histocompatibility leukocyte antigen-G in colorectal cancer cells.
Int J Mol Med.
2
1998
349
351
17
Cabestre
FA
Lefebvre
S
Moreau
P
et al
HLA-G expression: immune privilege for tumour cells?
Semin Cancer Biol.
9
1999
27
36
18
Paul
P
Cabestre
FA
Le Gal
FA
et al
Heterogeneity of HLA-G gene transcription and protein expression in malignant melanoma biopsies.
Cancer Res.
59
1999
1954
1960
19
Wagner
SN
Rebmann
V
Willers
CP
Grosse-Wilde
H
Goos
M
Expression analysis of classic and non-classic HLA molecules before interferon alfa-2b treatment of melanoma.
Lancet.
356
2000
220
221
20
Khalil-Daher
I
Rouas-Freiss
N
Carosella
ED
Dausset
JB
Human leukocyte antigen-G: immunotolerant major histocompatibility complex molecule in transplantation.
World J Surg.
24
2000
819
822
21
Carosella
ED
Rouas Freiss
N
Paul
P
Dausset
J
HLA-G: a tolerance molecule from the major histocompatibility complex.
Immunol Today.
20
1999
60
62
22
Onno
M
Guillaudeux
T
Amiot
L
et al
The HLA-G gene is expressed at a low mRNA level in different human cells and tissues.
Hum Immunol.
41
1994
79
86
23
Amiot
L
Onno
M
Drenou
B
et al
Distribution of HLA-G alternative mRNAs including soluble forms in normal lymphocytes and in lymphoid cell-derived leukemia.
Eur J Immunogenet.
23
1996
311
320
24
Amiot
L
Onno
M
Renard
I
et al
HLA-G transcription studies during the different stages of normal and malignant hematopoiesis.
Tissue Antigens.
48
1996
609
614
25
Amiot
L
Onno
M
Drenou
B
Monvoisin
C
Fauchet
R
HLA-G class I gene expression in normal and malignant hematopoietic cells.
Hum Immunol.
59
1998
524
528
26
Paul
P
Adrian Cabestre
F
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.
61
2000
1138
1149
27
Hennessy
A
Pilmore
HL
Simmons
LA
Painter
DM
A deficiency of placental IL-10 in preeclampsia.
J Immunol.
163
1999
3491
3495
28
Roth
I
Corry
DB
Locksley
RM
Abrams
JS
Litton
MJ
Fisher
SJ
Human placental cytotrophoblasts produce the immunosuppressive cytokine interleukin 10.
J Exp Med.
184
1996
539
548
29
Pazmany
L
Mandelboim
O
Vales Gomez
M
et al
Human leucocyte antigen-G and its recognition by natural killer cells.
J Reprod Immunol.
43
1999
127
137
30
Rouas Freiss
N
Khalil-Daher
I
Riteau
B
et al
The immunotolerance role of HLA-G.
Semin Cancer Biol.
9
1999
3
12
31
Colonna
M
Navarro
F
Bellon
T
et al
A common inhibitory receptor for major histocompatibility complex class I molecules on human lymphoid and myelomonocytic cells.
J Exp Med.
186
1997
1809
1818
32
Colonna
M
Samaridis
J
Cella
M
et al
Human myelomonocytic cells express an inhibitory receptor for classical and nonclassical MHC class I molecules.
J Immunol.
160
1998
3096
3100
33
Cantoni
C
Verdiani
S
Falco
M
et al
p49, a putative HLA class I-specific inhibitory NK receptor belonging to the immunoglobulin superfamily.
Eur J Immunol.
28
1998
1980
1990
34
Le Gal
FA
Riteau
B
Sedlik
C
et al
HLA-G-mediated inhibition of antigen-specific cytotoxic T lymphocytes.
Int Immunol.
11
1999
1351
1356
35
Lopez-Botet
M
Navarro
F
Llano
M
How do NK cells sense the expression of HLA-G class Ib molecules?
Semin Cancer Biol.
9
1999
19
26
36
Pangault
C
Amiot
L
Caulet Maugendre
S
et al
HLA-G protein expression is not induced during malignant transformation.
Tissue Antigens.
53
4 pt 1
1999
335
346
37
Willemze
R
Kerl
H
Sterry
W
et al
EORTC classification for primary cutaneous lymphomas: a proposal from the Cutaneous Lymphoma Study Group of the European Organization for Research and Treatment of Cancer.
Blood
90
1997
354
371
38
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.
91
1994
4209
4213
39
Fujii
T
Ishitani
A
Geraghty
DE
A soluble form of the HLA-G antigen is encoded by a messenger ribonucleic acid containing intron 4.
J Immunol.
153
1994
5516
5524
40
Finnegan
MC
Goepel
JR
Hancock
BW
Goyns
MH
Investigation of the expression of housekeeping genes in non-Hodgkin's lymphoma.
Leuk Lymphoma.
10
1993
387
393
41
O'Callaghan
CA
Bell
JI
Structure and function of the human MHC class Ib molecules HLA-E, HLA-F and HLA-G.
Immunol Rev.
163
1998
129
138
42
Carosella
ED
Dausset
J
Kirszenbaum
M
HLA-G revisited.
Immunol Today.
17
1996
407
409
43
Paul
P
Rouas-Freiss
N
Khalil-Daher
I
et al
HLA-G expression in melanoma: a way for tumor cells to escape from immunosurveillance.
Proc Natl Acad Sci U S A.
95
1998
4510
4515
44
Ulbrecht
M
Rehberger
B
Strobel
I
et al
HLA-G: expression in human keratinocytes in vitro and in human skin in vivo.
Eur J Immunol.
24
1994
176
180
45
Castro
MJ
Morales
P
Rojo-Amigo
R
et al
Homozygous HLA-G*0105N healthy individuals indicate that membrane-anchored HLA-G1 molecule is not necessary for survival.
Tissue Antigens.
56
2000
232
239
46
Moreau
P
Adrian Cabestre
F
Menier
C
et al
IL-10 selectively induces HLA-G expression in human trophoblasts and monocytes.
Int Immunol.
11
1999
803
811
47
Lefebvre
S
Adrian
F
Moreau
P
et al
Modulation of HLA-G expression in human thymic and amniotic epithelial cells.
Hum Immunol.
61
2000
1095
1101
48
Girdlestone
J
Synergistic induction of HLA class I expression by RelA and CIITA.
Blood.
95
2000
3804
3808
49
Saed
G
Fivenson
DP
Naidu
Y
Nickoloff
BJ
Mycosis fungoides exhibits a Th1-type cell-mediated cytokine profile whereas Sezary syndrome expresses a Th2-type profile [see comments].
J Invest Dermatol.
103
1994
29
33
Comment in J Invest Dermatol. 1995;105:855-856.
50
Harwix
S
Zachmann
K
Neumann
C
T-cell clones from early-stage cutaneous T-cell lymphoma show no polarized Th-1 or Th-2 cytokine profile.
Arch Dermatol Res.
292
2000
1
8
51
Yang
Y
Chu
W
Geraghty
DE
Hunt
JS
Expression of HLA-G in human mononuclear phagocytes and selective induction by IFN-gamma.
J Immunol.
156
1996
4224
4231
52
Wiendl
H
Behrens
L
Maier
S
Johnson
MA
Weiss
EH
Hohlfeld
R
Muscle fibers in inflammatory myopathies and cultured myoblasts express the nonclassical major histocompatibility antigen HLA-G.
Ann Neurol.
48
2000
679
684
53
Riteau
B
Menier
C
Khalil-Daher
I
et al
HLA-G1 co-expression boosts the HLA class I-mediated NK lysis inhibition.
Int Immunol.
13
2001
193
201
54
Riteau
B
Rouas-Freiss
N
Menier
C
Paul
P
Dausset
J
Carosella
ED
HLA-G2, -G3, and -G4 isoforms expressed as nonmature cell surface glycoproteins inhibit NK and antigen-specific CTL cytolysis.
J Immunol.
166
2001
5018
5026
55
Asadullah
K
Sabat
R
Wiese
A
Docke
WD
Volk
HD
Sterry
W
Interleukin-10 in cutaneous disorders: implications for its pathophysiological importance and therapeutic use.
Arch Dermatol Res.
291
1999
628
636
56
Khatri
VP
Caligiuri
MA
A review of the association between interleukin-10 and human B-cell malignancies.
Cancer Immunol Immunother.
46
1998
239
244
57
Diehl
M
Munz
C
Keilholz
W
et al
Nonclassical HLA-G molecules are classical peptide presenters.
Curr Biol.
6
1996
305
314
58
Lee
N
Malacko
AR
Ishitani
A
et al
The membrane-bound and soluble forms of HLA-G bind identical sets of endogenous peptides but differ with respect to TAP association.
Immunity.
3
1995
591
600
59
Hamai
Y
Fujii
T
Yamashita
T
et al
Peripheral blood mononuclear cells from women with recurrent abortion exhibit an aberrant reaction to release cytokines upon the direct contact of human leukocyte antigen-G-expressing cells.
Am J Reprod Immunol.
40
1998
408
413
60
Maejima
M
Fujii
T
Kozuma
S
Okai
T
Shibata
Y
Taketani
Y
Presence of HLA-G-expressing cells modulates the ability of peripheral blood mononuclear cells to release cytokines.
Am J Reprod Immunol.
38
1997
79
82
61
Gilliam
AC
Wood
GS
Primary cutaneous lymphomas other than mycosis fungoides.
Semin Oncol.
26
1999
290
306
62
Dummer
R
Schwarz
T
Cytokines as regulatory proteins in lymphoproliferative skin infiltrates.
Dermatol Clin.
12
1994
283
294
63
Siegel
RS
Pandolfino
T
Guitart
J
Rosen
S
Kuzel
TM
Primary cutaneous T-cell lymphoma: review and current concepts.
J Clin Oncol.
18
2000
2908
2925
64
Diamandidou
E
Colome Grimmer
M
Fayad
L
Duvic
M
Kurzrock
R
Transformation of mycosis fungoides/Sezary syndrome: clinical characteristics and prognosis.
Blood.
92
1998
1150
1159
65
Kamarashev
J
Burg
G
Mingari
MC
Kempf
W
Hofbauer
G
Dummer
R
Differential expression of cytotoxic molecules and killer cell inhibitory receptors in CD8+ and CD56+ cutaneous lymphomas.
Am J Pathol.
158
2001
1593
1598
66
Saverino
D
Fabbi
M
Ghiotto
F
et al
The CD85/LIR-1/ILT2 inhibitory receptor is expressed by all human T lymphocytes and down-regulates their functions.
J Immunol.
165
2000
3742
3755
67
Blaschitz
A
Lenfant
F
Mallet
V
et al
Endothelial cells in chorionic fetal vessels of first trimester placenta express HLA-G.
Eur J Immunol.
27
1997
3380
3388
68
Le Bouteiller
P
Solier
C
Is antigen presentation the primary function of HLA-G?
Microbes Infect.
3
2001
323
332
69
Beljaards
RC
Meijer
CJ
Scheffer
E
et al
Prognostic significance of CD30 (Ki-1/Ber-H2) expression in primary cutaneous large-cell lymphomas of T-cell origin. A clinicopathologic and immunohistochemical study in 20 patients.
Am J Pathol.
135
1989
1169
1178
70
van Haselen
CW
Toonstra
J
van der Putte
SJ
van Dongen
JJ
van Hees
CL
van Vloten
WA
Granulomatous slack skin. Report of three patients with an updated review of the literature.
Dermatology.
196
1998
382
391
71
Onno
M
Pangault
C
Le Friec
G
Guilloux
V
Andre
P
Fauchet
R
Modulation of HLA-G antigens expression by human cytomegalovirus: specific induction in activated macrophages harboring human cytomegalovirus infection.
J Immunol.
164
2000
6426
6434
72
Onno
M
Le Friec
G
Pangault
C
et al
Modulation of HLA-G antigens expression in myelomonocytic cells.
Hum Immunol.
61
2000
1086
1094
73
Lopez Botet
M
Llano
M
Navarro
F
Bellon
T
NK cell recognition of non-classical HLA class I molecules.
Semin Immunol.
12
2000
109
119
74
Allan
DS
Colonna
M
Lanier
LL
et al
Tetrameric complexes of human histocompatibility leukocyte antigen (HLA)-G bind to peripheral blood myelomonocytic cells.
J Exp Med.
189
1999
1149
1156
75
Borges
L
Cosman
D
LIRs/ILTs/MIRs, inhibitory and stimulatory Ig-superfamily receptors expressed in myeloid and lymphoid cells.
Cytokine Growth Factor Rev.
11
2000
209
217

Author notes

Reinhard Dummer, UniversitätsSpital Zürich, Dermatologische Klinik, Gloriastrasse 31, CH-8091 Zürich, Switzerland; e-mail:dummer@derm.unizh.ch.

Sign in via your Institution