Microarray analysis of gene expression by microdissected epidermis and dermis in mycosis fungoides and adult T-cell leukemia/lymphoma

  • Authors:
    • Keiko Hashikawa
    • Shinichiro Yasumoto
    • Kazutaka Nakashima
    • Fumiko Arakawa
    • Junichi Kiyasu
    • Yoshizo Kimura
    • Hiroshi Saruta
    • Takekuni Nakama
    • Kaori Yasuda
    • Kosuke Tashiro
    • Satoru Kuhara
    • Takashi Hashimoto
    • Koichi Ohshima
  • View Affiliations

  • Published online on: June 26, 2014     https://doi.org/10.3892/ijo.2014.2524
  • Pages: 1200-1208
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The characteristic histopathological feature of mycosis fungoides (MF) and adult T-cell leukemia/lymphoma (ATLL) is epidermotropism. To identify the mechanism for epidermotropism of lymphoma cells, total RNAs were obtained from skin biopsies of epidermis and dermis of MF and ATLL patients by means of laser capture microdissection, and used for subsequent complementary DNA (cDNA) microarray experiments. This procedure has made it possible for us to observe and evaluate the regional environment of MF and ATLL. Hierarchical cluster analysis revealed that the cDNAs could be clearly differentiated into MF and ATLL. CCL27 was expressed in the dermis generated from keratinocytes, CCR4/CCR6/CCR7/CCR10/cutaneous lymphocyte-associated antigen (CLA) lymphoma cells in the dermis, and CCL21 in the extracellular matrix (stroma). Lymphotoxin (LT) β and CCL21 expression was significantly higher and that of CCR10 relatively for MF, while CCR4 and CLA expression was relatively higher for ATLL. In the epithelium, keratinocytes expressed CCL20/CCL27, and lymphoma cells CCR4/CCR6/CCR10, while CCR4, CCR6, CCL20 and CCL27 expression was relatively higher for ATLL than MF. The dermis of MF, but not that of ATLL, showed correlation between CCR7 and CCL21. These findings support the suggestion that chemokines and chemokine receptors are involved in the pathogenesis of MF and ATLL, indicate that cutaneous homing seems to be different for MF and ATLL, and point to the possibility that cutaneous T-cell lymphomas originate in regulatory T cells, especially in the case of ATLL.

Introduction

Mycosis fungoides (MF) is the most common form of cutaneous T-cell lymphomas (CTCL), which is a heterogeneous group of non-Hodgkin’s lymphomas thought to result from malignancies of skin homing T cells (1) and presenting with patches, plaques and tumors.

While the early stage shows a wide clinical spectrum that overlaps with several inflammatory dermatoses, clinical as well as pathological findings indicate various overlaps between MF and inflammatory diseases, with epidermotropism disproportionate to the degree of spongiosis being one of the most useful pathological distinguishing features.

Adult T-cell leukemia/lymphoma (ATLL) is a human malignancy associated with human T-cell lymphotropic virus-type I (HTLV-1). Cutaneous lesions of ATLL consist of papules and nodules/tumors. While survival of ATLL cases with skin manifestations is reportedly significantly shorter than that of MF cases (2), histological findings for the two diseases are similar.

The mechanism of epidermotrophism seen in MF and ATLL has not yet been clearly identified. What is known is that chemokines regulate multiple cell functions, including cell chemotaxis, proliferation and apoptosis, and are involved in leukocyte transendothelial migration and homing to tissues. Previous studies have reported that positivity for CCR4 is significantly associated with skin involvement in MF and ATLL, and CCR10 is expressed by malignant cells in MF and Sézary syndrome (SS) (3). Malignant MF and SS cells also show high expression of CCR7. The cutaneous lymphocyte-associated antigen (CLA) recognized by the HECA-452 antibody is an adhesion molecule selectively expressed by a subset of circulating memory T-cells, normal T cells in inflamed skin and by the vast majority of CTCL (46). A previous study found that CLA was expressed on epidermotrophic lymphoma cells in all early stages MF. As for ATLL, CLA+ cases showed a significant preference for skin involvement when compared with the CLA cases as also previously reported by other investigators (7).

Complementary DNA (cDNA) microarray technology is a powerful tool for gaining insight into the molecular complexity and pathogenesis of various diseases and makes it possible to identify differences in numerous gene expressions.

Since the skin consists of epidermis [including keratinocytes and dendritic cells (DCs)], dermis (stroma and vessels) and subcutaneous tissue with or without inflamed and/or inflammatory cells, gene expression of epidermis and dermis should be analyzed separately to gain a better understanding of the contribution made by each component of a tissue to the pathogenesis of epidermotrophism. To this end, laser capture microdissection (LMD) was used in our study to allow us to focus on the differences in gene expression levels between epidermis and dermis in MF and ATLL and to subsequently determine these expressions immunohistochemically in patients.

It is believed that this technique, which combines LMD and microarray technology for the analysis of CTCL gene expression in epidermis and dermis, can improve our understanding of epidermotrophism at the molecular level.

Materials and methods

Eighteen skin samples obtained from patients treated at the Department of Dermatology, Kurume University School of Medicine consisted of five epidermal and three dermal samples each from MF and ATLL as well as two epidermal samples from dermatitis patients used as controls. Skin biopsies were performed after informed consent had been obtained from all patients or their guardians in accordance with the Declaration of Helsinki. Both paraffin-embedded and frozen tissues were used. Histopathological diagnoses were based on the World Health Organization classification (WHO) and performed by five pathologists (K.H., J.K., Y.K., H.S. and K.O.). The clinical data for all patients obtained from the medical records are summarized in Table I, which shows that both epidermal and dermal samples were harvested from two MF (E-M3/D-M1 and E-M5/D-M2) and two ATLL (E-A2/D-A1 and E-A3/D-A2) patients. The other samples were either epidermal or dermal. This study was approved by the Kurume University Institutional Review Board.

Table I

Summary of patients enrolled in this study: number, age, gender and stage.

Table I

Summary of patients enrolled in this study: number, age, gender and stage.

Sample no.Age (years)GenderTNMStage
MF
 E-M178MaleT2aN0M0IB
 E-M262FemaleT2bN0M0IB
 E-M3,D-M157MaleT1bN0M0IA
 E-M422MaleT1bN0M0IA
 E-M5, D-M264MaleT2bN0M0IB
 D-M361MaleT2bN0M0IB
ATLL
 E-A156MaleT2aN2M0IIA
 E-A2, D-A170MaleT2cN0M0IB
 E-A3, D-A232FemaleT2cN0M0IB
 E-A463MaleT3aNxM0IIB
 E-A579MaleT3bN0M0IIB
 D-A354MaleT3aN1M0IIB
Control
 N166Male
 N271Female

[i] The samples were obtained from six MF and ATLL patients each, and from two dermatitis patients as controls. The five epidermal (E) samples of MF (−M) patients were numbered E-M1 through E-M5, and the three dermal (D) samples of MF (−M) patients were numbered D-M1 through D-M3. Similarly, the ATLL (−A) samples were numbered E-A1 through E-A5 and D-A1 through D-A3. The two epidermal samples from dermatitis patients used as controls were numbered N1 and N2.

Laser microdissection

The samples were embedded in an optical cutting temperature compound (Sakura Finetechnical, Tokyo, Japan), immediately frozen in liquid nitrogen, and stored at −80°C for microdissection. A series of 10-μm thick sections were cut from frozen tissue specimens at −20°C, and mounted on 2.0-μm-thick PEN-Membrane slides (MicroDissect GmbH, Herborn, Germany). The epidermal and/or dermal regions were microdissected from the about 70 cryosections by means of LMD. After fixation in 100% ethanol, the slides were stained in turn with Mayer hematoxylin and eosin, washed in diethylpyrocarbonate-treated water at each phase of the process and then air-dried with a fan. The frozen sections were microdissected with a Leica LMD6000 laser microdissection system (Leica, Wetzlar, Germany) in accordance with the company’s protocol. Finally, the microdissected fragments were dropped into 0.5 ml tube caps containing 50 μl lysis buffer for RNA extraction (8).

RNA extraction and biotinylated cRNA amplification

Total RNA was extracted from the samples collected by means of LMD and with an RNAqueous-Micro kit (AM1931; Ambion, Austin, TX) according to the manufacturer’s instructions. Complementary RNA (cRNA) amplification and labeling with biotin were used for gene expression profiling by microarray analysis. Briefly, 500 ng total RNA was amplified overnight (14 h) with the Illumina Total Prep RNA Amplification kit (AMIL1791; Ambion) according to the manufacturer’s protocol. Reaction cRNA was biotinylated during in vitro transcription.

Illumina BeadChips microarray

Sentrix Human WG-6 v3.0 Expression BeadChips were purchased from Illumina, Inc. (San Diego, CA). More than 48,000 different bead types, each with a 50-base gene-specific probe, are represented on a single Beadchip. For each probe represented on the array, beads are assembled with an average 30-fold redundancy. A hybridization mixture containing 1.5 μg biotinylated cRNA was hybridized to the beadchips at 58°C overnight (18 h) before being washed and stained with streptavidin-Cyanine-3 (Cy3) (PA23031; GE Healthcare, Buckinghamshire, UK) according to the manufacturer’s protocol. Beadchips were scanned on Illumina BeadStation 500 and fluorescent hybridization signals were assessed with Illumina Beadstudio software.

The data discussed in this publication have been deposited in National Center for Biotechnology Information (NCBIs) Gene Expression Omnibus (GEO, http://www.ncbi.nlm.nih.gov/geo/) and are accessible through GEO Series accession number GSE40639.

Statistical analysis

For the pre-processing step, variance in the data was first stabilized with the variance stabilizing transform method (9) and then normalized with a robust spline normalization method, both of which are used with the Lumi BioConductor package (Illumina) (10). Effectively absent transcripts were filtered out to reduce false positives. Detection of transcripts was considered to be achieved if the detection p-value calculated from the background with the Illumina BeadStudio was <0.05 for all hybridizations. The Significance Analysis of the Microarrays statistical test (11), which is used as part of the bioconductor ‘samr’ package and takes multiple testing into account by estimating the false discovery rate, was used to identify differentially expressed transcripts for a comparison of MF and ATLL patients. If a transcript was up- or downregulated by a factor of ≥2 and had a Significance Analysis of Microarrays q-value (false discovery rate) of <0.001, we regarded the expression of this transcript in MF patients as different from that in ATLL patients. To obtain reproducible clusters for classifying the 16 samples (all samples detailed above except for the two control samples), expression data were analyzed with GeneSpring 7.2 software (Silicon Genetics, Redwood City, CA), which was also used to generate heatmaps of certain genes of interest.

Immunohistochemistry

The paraffin-embedded specimens were used for manual immunohistochemical analysis of CCR4 (Pharmingen, San Diego, CA), CCR7, CCR6 and CCR10 (all from Medical and Biological Laboratories Co., Ltd., Nagoya, Japan), CCL20, CCL27, lymphotoxin (LT) β and TNF receptor (TNFR) 2 (all from R&D Systems, Minneapolis, MN), CCL21 (Santa Cruz Biotechnology, Santa Cruz, CA), β-defensin (BDF) 1 (Phoenix Pharmaceuticals, Inc., Burlingame, CA) as previously described (12). Appropriate positive and negative control experiments were run simultaneously. Heat-mediated antigen retrieval was used for all analyses except those of CCR6 and BDF1. The staining results were evaluated semiquantitatively by three independent observers, and scored comprehensively in view of intensity, expression pattern and number of positive cells, and so on. Images were visualized with an Olympus AX80 microscope (Olympus, Tokyo, Japan), equipped with an Olympus Planapo 40x/0.95 numerical aperture objective. Images were captured with an Olympus DP70 camera and Olympus DP controller software, and were processed with Olympus DP manager software.

Results

Following appropriate normalization and standardization procedures, the data on each chip were compared with each other by using a hierarchical clustering method (Fig. 1). Genes differentially expressed in MF and ATLL were organized by means of Ingenuity Pathway Analysis into an interactome network, which was then used for an comparison of the gene expression (transcriptional profile) of epidermal or dermal MF with that of ATLL. In the epidermis, 580 probe sets were identified as upregulated or downregulated on a heat map (Fig. 1A), while in the dermis, 234 probe sets were found to be up- or downregulated (Fig. 1B). The first 10 genes showing the highest upregulation are listed in Table II. In the dermis, LTβ and CCL21 in particular showed different expressions in MF and ATLL.

Table II

Genes differentially regulated in MF and ATLL.

Table II

Genes differentially regulated in MF and ATLL.

A, Upregulated genes in MF compared with genes in ATLL

EpidermisGene symbolGene nameExp. value
1ZSCAN18Zinc finger and SCAN domain containing 181.392
2COL16A1Collagen1.217
3EID2BEP300 interacting inhibitor of differentiation 2B1.167
4HSPA1AHeat shock 70 kDa protein 1A1.046
5GGA1Golgi associated1.038
6LRIG1Leucine-rich repeats and immunoglobulin-like domains 11.010
7XPNPEP3X-prolyl aminopeptidase (aminopeptidase P) 31.003
8SMARCA2SWI/SNF related0.979
9ID2Inhibitor of DNA binding 20.968
10ARL6IP5 ADP-ribosylation-like factor 6 interacting protein 50.950

DermisGene symbolGene nameExp. value

1LTBLymphotoxin β2.946
2CCL21Chemokine (C-C motif) ligand 212.189
3IFITM3Interferon induced transmembrane protein 31.666
4THY1Thy-1 cell surface antigen1.594
5ACTN1Actinin1.585
6CHST15Carbohydrate1.449
7PON2Paraoxonase 21.447
8CYR61Cysteine-rich1.436
9TAGLNTransgelin1.436
10PRRX1Paired related homeobox 11.412

B, Downregulated genes in MF compared with genes in ATLL

EpidermisGene symbolGene nameExp. value

1GPX3Glutathione peroxidase 3−2.097
2PSMC1Proteasome−1.668
3CALML3Calmodulin-like 3−1.587
4NDRG4NDRG family member 4−1.169
5PRSS3Protease−1.113
6ADMAdrenomedullin−1.063
7SMOXSpermine oxidase−1.056
8CBX3Chromobox homolog 3−1.044
9WBP5WW domain binding protein 5−0.970
10LRRC20Leucine rich repeat containing 20−0.967

DermisGene symbolGene nameExp. value

1TOX2TOX high mobility group box family member 2−2.925
2ADAP1ArfGAP with dual PH domains 1−2.848
3AKAP7A kinase anchor protein 7−2.449
4CADM1Cell adhesion molecule 1−2.293
5ZNF365Zinc finger protein 365−2.157
6RGS2Regulator of G-protein signaling 2−2.129
7MTX3Metaxin 3−2.080
8SH3KBP1SH3-domain kinase binding protein 1−1.871
9CHST7Carbohydrate sulfotransferase 7−1.792
10CST7Cystatin F (leukocystatin)−1.739

[i] The 10 most significant signatures of gene expression are shown. LTβ and CCL21 in the dermis showed higher expressions in MF than in ATLL.

Next, we focused on the array data that could identify the possible involvement of specific chemokine receptors and ligands, including CCR4, CCR6, CCR7 and CCR10 and others, in the pathophysiology of CTCL, MF and ATLL. CCR4 expression was especially high in ATLL, while CCL27 expression in the epidermis was high in both MF and ATLL, and CCR10, a receptor of CCL27 detected in the dermis, was more highly expressed in MF than in ATLL. Also in the dermis, expression of the LN homing molecule CCR7 was high in both MF and ATLL, while a close correlation between CCR7 and CCL21, a chemokine receptor and its ligand, was observed in MF. CCR6 expression was elevated in the epidermis and dermis of both MF and ATLL, while expression of CCL20, a CCR6 ligand, showed no correlation with CCR6. BDFs were identified as not only antibacterial peptides but also as CCR6 ligands. BDF 1 expression was reduced in the epidermis of MF and ATLL, while BDF 3 expression in the epidermis of MF was higher than in controls, which was similar to the finding of a previous study (13).

We also focused on CLA, which is recognized as an adhesion molecule selectivity expressed by a subset of circulating memory T-cells, normal T cells in inflamed skin and by the vast majority of CTCL (46). CLA was expressed in the dermis of both MF and ATLL, with a particularly high expression in ATLL.

In order to confirm the expression levels of the proteins, CCR6, CCR7, CCR10, CCL20, CCL21, CCL27, BDF1, LTβ and TNFR2 on MF and ATLL tissue sections were subjected to immunohistochemistry. The findings of the immunohistochemical analysis were basically consistent with the results of the microarray analysis. A representative stain is shown in Fig. 2 and the immunohistochemical scores are presented graphically in Fig. 3.

In principle, CCR4, CCR6, CCR7, CCR10 and/or CLA were all expressed in the lymphoma cells. CCR4, CCR6 and CCR10 were identified in both dermis and epidermis, while CCL27 and CCL20 were expressed in keratinocytes and CCL21 was expressed in the extracellular matrix (stroma). In addition, CCL27 was also stained in dermis released from keratinocytes.

In controls, epidermal keratinocytes weakly expressed CCL27 within their cytoplasm, and mainly in the spinous layers of the epidermis. However, positive lesions of epidermis keratinocytes were stronger in thicker layers in most MF and ATLL patients than in controls. Moreover, CCL27 was expressed not only in cytoplasm but also occasionally in membrane of MF and ATLL, so that there was in fact not much difference between these two types of CTCL. Faint CCL27 expression in dermis was detected in MF, ATLL and in one of the controls. The differences in CCL27 expression levels in dermis were immunohistochemically imperceptible. CCR10 was stained weakly in both epidermis and dermis, but in the dermis usually more strongly in MF than in ATLL cells, which correlated moderately with the overall gene expression profile. CCR6 expression was more elevated in the epidermis and dermis of both MF and ATLL than in controls, and the immunohistochemical score correlated with the result of the microarray analysis. CCL20 was expressed very weakly in the cytoplasm of keratinocytes, stained in the granular layer of all samples and in all layers of the epidermis of some samples.

Although each one of the controls and MF samples showed high levels of CCL20, all ATLL samples displayed high expression in the epidermis. Expression of BDF1 in nucleus and/or nuclear membrane was diminished in the epidermis of MF and ATLL. Because the stain was quite faint, it was difficult to determine even whether CCL20 and BDF1 in the dermis were positive or negative. The cell membrane of infiltrating lymphoma cells stained for CCR7, while staining for CCL21 was observed within the cytoplasm of epidermal keratinocytes and was diffusely distributed on the dermal extracellular matrix. The intensity and density of CCR7+ infiltrating lymphoma cells correlated with the intensity of CCL21 in stroma in the dermis of MF cases. LTβ positive cells were scattered throughout the dermis and their expression was homogeneous in the nucleus or the surface of the nucleus of lymphoma cells, while LTβ expression levels in the dermis were higher for MF than for ATLL. No correlation with LTβ was observed for TNFR2, which is one of the soluble secreted LT homotrimers (LTα3) that trigger TNFRs.

Discussion

The microarray and immunohistochemistry findings of our study showed high expression of LTβ and CCL21, and correltion between CCL21 and CCR7, a receptor of CCL21 which is the LN homing molecule, was observed in the dermis of MF. CCR7 expression was also high in the dermis of ATLL.

LTβ, which was indentified in the dermis of MF samples in our study, is a member of the tumor necrosis factor superfamily. LTβ in human cells contains a transmembrane domain, and the surface LT form is most likely a trimer with an LTα1β2 stoichiometry, while the LTα1β2 heterotrimer binds the LTβ receptor (LTβR) (14).

LTs are expressed by activated T-, B-, NK- and lymphoid tissue inducer cells (1517) and are crucial for organogenesis and maintenance of lymphoid tissues (18,19). LTβR, on the other hand, is expressed on many different cell types including cells of epithelial lineages, while ligation of LTβR results in NF-κB activation (2022) and leads to secondary lymphoid organogenesis and homeostasis. Signaling via LTβR is involved not only in host defense and autoimmune diseases, but also in tumor cell proliferation (23). Furthermore, NF-κB stimulates proliferation and blocks programmed cell death (apoptosis) in various cell types (24,25).

Previous reports have suggested that the activation of the NF-κB signaling pathway via LTβR results in the proliferation of melanoma (26) and hepatocellular carcinoma (27). NF-κB activation was also detected in lymphoid malignancies (28,29). In this connection, it was found that peripheral T cell lymphomas/not otherwise specified (PTCLs/NOS) with NF-κB inactivity showed better survival (3032).

Another study reported that LTβR organizes the recruitment and survival of CD4 T cells through the induction of CCL21 expression which is produced exclusively by stroma (33). In our study we found that expression levels of both the LTβ and CCL21 gene are high in the dermis of MF. These findings seem to suggest that LTβR, which can be expressed in epidermis, organizes the recruitment and survival of CD4-positive MF cells through the induction of CCL21.

CCR7 (the receptor of CCL21) physiologically regulates the lymphoid homing of T-cells and probably plays an important role in the tropism of CTCL cells to peripheral LNs, which constitutively synthesize CCR7 ligands, CCL19 and CCL21 (34). It has been further hypothesized that CCR7, which is expressed at fairly high levels in SS cells (35), may function as a mediator of LN infiltration of CTCL. In our study, CCR7 expression was detected in the dermis of MF and ATLL. Interaction between CCR7 and CCL21 in the dermis of MF probably functions to maintain localization to the skin for a long time. On the other hand, T cells infected by HTLV-1 circulate in the bloodstream from the outset, while ATLL cells, expressing the skin-homing properties of CCR4, CLA, CCR6 and CCR10, may affect cutaneous involvement in smorderling or cutaneous type of ATLL patients.

While T cells infected by HTLV-1 are circulating in the peripheral blood, they can spontaneously encounter CCL21 produced by LNs, and CCR7+ ATLL cells can be trapped in the LNs. Migration to the LNs can thus occur at an earlier stage in ATLL than in MF.

CTCL is characterized by clonal expansion of a mature CD4-positive clone of the Th phenotype, putatively from a skin homing subset of memory T cells (36) whose migration to respective tissues is tightly regulated by adhesion molecules and chemokine receptors. For example, memory T cells that infiltrate the skin express a unique adhesion molecule known as CLA. Chemokines and their receptors have also been associated with tumor metastasis (3739) and possibly trafficking of lymphoma cells (40). In addition, chemokines are produced by not only malignant T cells but also epidermal keratinocytes, DCs and dermal vessels. We therefore did not use fractionated malignant T cells, but the microdissected epidermal and dermal affected regions in MF and ATLL to investigate the expression profiles of CCR4, CCR6, CCR7, CCR10, CCL20, CCL21, CCL27, BDFs and CLA by means of microarray analysis and/or immunohistochemistry. We found that CCL27 expression in the epidermis was high in both MF and ATLL. CCR10 expression observed in the dermis was higher in MF than ATLL, and the microarray analysis showed that CCR4 expression was especially high in ATLL. Previous studies have reported that CCR4 expression was detected much more frequently in both MF and ATLL than in healthy controls (41,42).

In addition, findings of a previous study indicated that DCs synthesize CCR4 ligands, which rapidly stimulate chemotaxis of, or conjugate formation with, normal T cells (43,44). A characteristic histopathological marker, Pautrier’s microabscesses of CTCL, which are known to be DC-malignant T cell conjugates (45), may be initiated by DC-derived chemokines. Other data indicated that preferential expression of CCR4 constitutes a sign of worse prognosis, and CCR4 expression was especially high in ATLL in our study. This finding corresponds to the pathological feature of more visible and larger Pautrier’s microabscesses as well as the clinical feature of worse prognosis for ATLL than for MF.

CCR10 is enriched in CLA+ skin-homing T cells of psoriasis, atopic dermatitis and CTCL patients, while it is only rarely expressed on peripheral blood T cells and skin samples from healthy persons (3,46). Moreover, CCL27 (CCR10 ligand) is constitutively present in epidermal keratinocytes (basal layer) under basal, non-inflammatory conditions and can be rapidly released from activated keratinocytes (46). CCL27-CCR10 interactions thus play an early role in the pathophysiology of MF from the patch stage (47), and an increase in CCL27 in the serum of MF patients was observed in previous studies (48). Our data for CCL27-CCR10 interactions agreed with the pathophysiology outlined above.

Our findings also indicate that CCR4 in ATLL as well as CCR10 in MF may play a much more prominent role in epidermotrophism. However, the origin of malignant T cells in MF and ATLL remains to be fully elucidated. High-level expressions of CCR4, CLA and CCR6 indicate the possibility of not only Th but also Treg origin especially in ATLL. Treg action suppresses activation of the immune system and tolerance for self-antigens. The phenotype of CD4+CD25+ and the expression of Foxp3, a master transcriptional regulator of Treg development and function, suggest that human skin-resident T cells appear to represent Tregs. Tregs also express functional skin-homing receptors. Although CCR6 does not seem to be a functional marker for Treg, it may facilitate CLA+ Treg migration to skin. As for Th, there are conflicting reports about Th1 and Th2 profiles of CCR4 in MF. Berger et al demonstrated that malignant CD4+ T cells can be induced to express a CD25+ Treg phenotype (49). Our analyses, however, disclosed that CCR4 and CCR6 in both epidermis and dermis and CLA in dermis were expressed in MF and ATLL, with the expression being especially high in ATLL. Our data therefore support the possibility that the origin of CTCL is Treg (particularly in ATLL) as also previously reported (50). Further investigation will be required to determine whether CTCL shows Th1/Th2/Treg-type polarization in lesional skin.

While the mechanisms involved in tumor homing have not yet been fully identified, it has been suggested that chemokines and chemokine receptors are involved in the pathogenesis of tumor homing. There are no previous reports on the use of the microdissection method for a comparison of epidermal and dermal gene expression levels in MF and ATLL by means of microarray analysis, a procedure which has made it possible to observe and evaluate the regional environment of MF and ATLL, including malignant and inflamed T-cells, DCs, keratinocytes and dermal vessels. DNA microarray analysis enabled us to comprehensively identify differences and patterns in gene expression in MF and ATLL, while our findings support the notion that CCR10 and its ligand CCL27 may contribute to the skin infiltration of malignant T-cells in MF and ATLL. However, further studies are needed to clarify the mechanism for epidermotrophism because of the complexity of its interactions.

Acknowledgements

The authors would like to thank Konomi Takasu, Mayumi Miura and Kanoko Miyazaki for their technical support.

References

1 

Willemze R, Jaffe ES, Burg G, et al: WHO-EORTC classification for cutaneous lymphomas. Blood. 105:3768–3785. 2009. View Article : Google Scholar

2 

Kikuchi A, Ohata Y, Matsumoto H, Sugiura M and Nishikawa T: Anti-HTLV-1 antibody positive cutaneous T-cell lymphoma. Cancer. 79:269–274. 1997. View Article : Google Scholar : PubMed/NCBI

3 

Notohamiprodjo M, Segerer S, Huss R, et al: CCR10 is expressed in cutaneous T-cell lymphoma. Int J Cancer. 115:641–647. 2005. View Article : Google Scholar : PubMed/NCBI

4 

Picker LJ, Terstappen LW, Rott LS, Streeter PR, Stein H and Butcher EC: Differential expression of homing-associated adhesion molecules by T-cell subsets in man. J Immunol. 145:3247–3255. 1990.PubMed/NCBI

5 

Picker LJ, Michie SA, Rott LS and Butcher EC: A unique phenotype of skin-associated lymphocytes in humans: preferential expression of the HECA-452 epitope by benign and malignant T cells at cutaneous sites. Am J Pathol. 136:1053–1068. 1990.PubMed/NCBI

6 

Berg EL, Yoshino T, Rott LS, et al: The cutaneous lymphocyte antigen is a skin lymphocyte homing receptor for the vascular lectin endothelial cell leukocyte adhesion molecule 1. J Exp Med. 174:1461–1466. 1991. View Article : Google Scholar : PubMed/NCBI

7 

Tanaka Y, Wake A, Horgan KJ, et al: Distinct phenotype of leukemic T cells with various tissues tropisms. J Immunol. 158:3822–3829. 1997.PubMed/NCBI

8 

Rubin MA: Use of laser capture microdissection, cDNA microarrays, and tissue microarrays in advancing our understanding of prostate cancer. J Pathol. 195:80–86. 2001. View Article : Google Scholar : PubMed/NCBI

9 

Lin SM, Du P, Huber W and Kibbe WA: Model-based variance-stabilizing transformation for Illumina microarray data. Nucleic Acids Res. 36:e112008. View Article : Google Scholar : PubMed/NCBI

10 

Du P, Kibbe WA and Lin SM: lumi: a pipeline for processing Illumina microarray. Bioinformatics. 24:1547–1548. 2008. View Article : Google Scholar : PubMed/NCBI

11 

Tusher VG, Tibshirani R and Chu G: Significance analysis of microarrays applied to the ionizing radiation response. Proc Natl Acad Sci USA. 98:5116–5121. 2001. View Article : Google Scholar : PubMed/NCBI

12 

Ohshima K, Kawasaki C, Muta H, et al: CD10 and Bcl10 expression in diffuse large B-cell lymphoma: CD10 is a marker of improved prognosis. Histopathology. 39:156–162. 2001. View Article : Google Scholar : PubMed/NCBI

13 

Gambichler T, Skrygan M, Appelhans C, et al: Expression of human beta-defensins in patients with mycosis fungoides. Arch Dermatol Res. 299:221–224. 2007. View Article : Google Scholar : PubMed/NCBI

14 

Browning JL, Dougas I, Ngam-ek A, et al: Characterization of surface lymphotoxin forms. Use of specific monoclonal antibodies and soluble receptors. J Immunol. 154:33–46. 1995.PubMed/NCBI

15 

Ware CF, Crowe PD, Grayson MH, Androlewicz MJ and Browning JL: Expression of surface lymphotoxin and tumor necrosis factor on activated T, B, and natural killer cells. J Immunol. 149:3881–3888. 1992.PubMed/NCBI

16 

Fu YX, Huang G, Wang Y and Chaplin DD: B lymphocytes induce the formation of follicular dendritic cell clusters in a lymphotoxin alpha-dependent fashion. J Exp Med. 187:1009–1018. 1998. View Article : Google Scholar : PubMed/NCBI

17 

Ware CF: Network communications: lymphotoxins, LIGHT, and TNF. Annu Rev Immunol. 23:787–819. 2005. View Article : Google Scholar : PubMed/NCBI

18 

Rennert PD, Browning JL, Mebius R, Mackay F and Hochman PS: Surface lymphotoxin alpha/beta complex is required for the development of peripheral lymphoid organs. J Exp Med. 184:1999–2006. 1996. View Article : Google Scholar : PubMed/NCBI

19 

Tumanov AV, Kuprash DV and Nedospasov SA: The role of lymphotoxin in development and maintenance of secondary lymphoid tissues. Cytokine Growth Factor Rev. 14:275–288. 2003. View Article : Google Scholar : PubMed/NCBI

20 

Dejardin E, Droin NM, Delhase M, et al: The lymphotoxin-beta receptor induces different patterns of gene expression via two NF-kappaB pathways. Immunity. 17:525–535. 2002. View Article : Google Scholar : PubMed/NCBI

21 

Dejardin E: The alternative NF-kappaB pathway from biochemistry to biology: pitfalls and promises for future drug development. Biochem Pharmacol. 72:1161–1179. 2006. View Article : Google Scholar : PubMed/NCBI

22 

Matsushima A, Kaisho T, Rennert PD, et al: Essential role of nuclear factor (NF)-kappaB-inducing kinase and inhibitor of kappaB (IkappaB) kinase alpha in NF-kappaB activation through lymphotoxin beta receptor, but not through tumor necrosis factor receptor I. J Exp Med. 193:631–636. 2001. View Article : Google Scholar

23 

Hehlgans T, Stoelcker B, Stopfer P, et al: Lymphotoxin-beta receptor immune interaction promotes tumor growth by inducing angiogenesis. Cancer Res. 62:4034–4040. 2002.PubMed/NCBI

24 

Barkett M and Gilmore TD: Control of apoptosis by Rel/NF-kappaB transcription factors. Oncogene. 18:6910–6924. 1999. View Article : Google Scholar : PubMed/NCBI

25 

Biswas DK, Martin KJ, McAlister C, et al: Apoptosis caused by chemotherapeutic inhibition of nuclear factor-kappaB activation. Cancer Res. 63:290–295. 2003.PubMed/NCBI

26 

Dhawan P, Su Y, Thu YM, et al: The lymphotoxin-beta receptor is an upstream activator of NF-kappaB-mediated transcription in melanoma cells. J Biol Chem. 283:15399–15408. 2008. View Article : Google Scholar : PubMed/NCBI

27 

Haybaeck J, Zeller N, Wolf MJ, et al: A lymphotoxin-driven pathway to hepatocellular carcinoma. Cancer Cell. 16:295–308. 2009. View Article : Google Scholar : PubMed/NCBI

28 

Gilmore TD: Multiple mutations contribute to the oncogenicity of the retroviral oncoprotein v-Rel. Oncogene. 18:6925–6937. 1999. View Article : Google Scholar : PubMed/NCBI

29 

Häcker H and Karin M: Is NF-kappaB2/p100 a direct activator of programmed cell death? Cancer Cell. 2:431–433. 2002.PubMed/NCBI

30 

Martinez-Delgado B, Meléndez B, Cuadros M, et al: Expression profiling of T-cell lymphomas differentiates peripheral and lymphoblastic lymphomas and defines survival related genes. Clin Cancer Res. 10:4971–4982. 2004. View Article : Google Scholar

31 

Martinez-Delgado B, Cuadros M, Honrado E, et al: Differential expression of NF-kappaB pathway genes among peripheral T-cell lymphomas. Leukemia. 19:2254–2263. 2005. View Article : Google Scholar : PubMed/NCBI

32 

Ballester B, Ramuz O, Gisselbrecht C, et al: Gene expression profiling identifies molecular subgroups among nodal peripheral T-cell lymphomas. Oncogene. 25:1560–1570. 2006. View Article : Google Scholar : PubMed/NCBI

33 

Bekiaris V, Gaspal F, Kim MY, et al: CD30 is required for CCL21 expression and CD4 T cell recruitment in the absence of lymphotoxin signals. J Immunol. 182:4771–4775. 2009. View Article : Google Scholar : PubMed/NCBI

34 

Kallinich T, Muche JM, Qin S, Sterry W, Audring H and Kroczek RA: Chemokine receptor expression on neoplastic and reactive T cells in the skin at different stages of mycosis fungoides. J Invest Dermatol. 121:1045–1052. 2003. View Article : Google Scholar : PubMed/NCBI

35 

Sokolowska-Wojdylo M, Wenzel J, Gaffal E, et al: Circulating clonal CLA(+) and CD4(+) T cells in Se’zary syndrome express the skin-homing chemokine receptors CCR4 and CCR10 as well as the lymph node-homing chemokine receptor CCR7. Br J Dermatol. 152:258–264. 2005.PubMed/NCBI

36 

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. 90:354–371. 1997.

37 

Müller A, Homey B, Soto H, et al: Involvement of chemokine receptors in breast cancer metastasis. Nature. 410:50–56. 2001.PubMed/NCBI

38 

von Andrian UH and Mempel TR: Homing and cellular traffic in lymph nodes. Nat Rev Immunol. 3:867–878. 2003.PubMed/NCBI

39 

Balkwill F: Cancer and the chemokine network. Nat Rev Cancer. 4:540–550. 2004. View Article : Google Scholar

40 

Jones D, O’Hara C, Kraus MD, et al: Expression pattern of T-cell-associated chemokine receptors and their chemokines correlates with specific subtypes of T-cell non-Hodgkin lymphoma. Blood. 96:685–690. 2000.PubMed/NCBI

41 

Ferenczi K, Fuhlbrigge RC, Pinkus J, Pinkus GS and Kupper TS: Increased CCR4 expression in cutaneous T cell lymphoma. J Invest Dermatol. 119:1405–1410. 2002. View Article : Google Scholar : PubMed/NCBI

42 

Narducci MG, Scala E, Bresin A, et al: Skin homing of Se’zary cells involves SDF-1-CXCR4 signaling and down-regulation of CD26/dipeptidylpeptidase IV. Blood. 107:1108–1115. 2006.

43 

Tang HL and Cyster JG: Chemokine up-regulation and activated T cell attraction by maturing dendritic cells. Science. 284:819–822. 1999. View Article : Google Scholar : PubMed/NCBI

44 

Wu M, Fang H and Hwang ST: Cutting edge: CCR4 mediates antigen-primed T cell binding to activated dendritic cells. J Immunol. 167:4791–4795. 2001. View Article : Google Scholar : PubMed/NCBI

45 

Edelson RL: Cutaneous T cell lymphoma: the helping hand of dendritic cells. Ann N Y Acad Sci. 941:1–11. 2001. View Article : Google Scholar : PubMed/NCBI

46 

Homey B, Alenius H, Müller A, et al: CCL27-CCR10 interactions regulate T cell-mediated skin inflammation. Nat Med. 8:157–165. 2002. View Article : Google Scholar : PubMed/NCBI

47 

Fujita Y, Abe R, Sasaki M, et al: Presence of circulating CCR10+ T cells and elevated serum CTACK/CCL27 in the early stage of mycosis fungoides. Clin Cancer Res. 12:2670–2675. 2006.PubMed/NCBI

48 

Kagami S, Sugaya M, Minatani Y, et al: Elevated serum CTACK/CCL27 levels in CTCL. J Invest Dermatol. 126:1189–1191. 2006. View Article : Google Scholar : PubMed/NCBI

49 

Berger CL, Tigelaar R, Cohen J, et al: Cutaneous T-cell lymphoma: malignant proliferation of T-regulatory cells. Blood. 105:1640–1647. 2005. View Article : Google Scholar : PubMed/NCBI

50 

Karube K, Aoki R, Sugita Y, et al: The relationship of FOXP3 expression and clinicopathological characteristics in adult T-cell leukemia/lymphoma. Mod Pathol. 21:617–625. 2008. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2014
Volume 45 Issue 3

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Hashikawa K, Yasumoto S, Nakashima K, Arakawa F, Kiyasu J, Kimura Y, Saruta H, Nakama T, Yasuda K, Tashiro K, Tashiro K, et al: Microarray analysis of gene expression by microdissected epidermis and dermis in mycosis fungoides and adult T-cell leukemia/lymphoma. Int J Oncol 45: 1200-1208, 2014
APA
Hashikawa, K., Yasumoto, S., Nakashima, K., Arakawa, F., Kiyasu, J., Kimura, Y. ... Ohshima, K. (2014). Microarray analysis of gene expression by microdissected epidermis and dermis in mycosis fungoides and adult T-cell leukemia/lymphoma. International Journal of Oncology, 45, 1200-1208. https://doi.org/10.3892/ijo.2014.2524
MLA
Hashikawa, K., Yasumoto, S., Nakashima, K., Arakawa, F., Kiyasu, J., Kimura, Y., Saruta, H., Nakama, T., Yasuda, K., Tashiro, K., Kuhara, S., Hashimoto, T., Ohshima, K."Microarray analysis of gene expression by microdissected epidermis and dermis in mycosis fungoides and adult T-cell leukemia/lymphoma". International Journal of Oncology 45.3 (2014): 1200-1208.
Chicago
Hashikawa, K., Yasumoto, S., Nakashima, K., Arakawa, F., Kiyasu, J., Kimura, Y., Saruta, H., Nakama, T., Yasuda, K., Tashiro, K., Kuhara, S., Hashimoto, T., Ohshima, K."Microarray analysis of gene expression by microdissected epidermis and dermis in mycosis fungoides and adult T-cell leukemia/lymphoma". International Journal of Oncology 45, no. 3 (2014): 1200-1208. https://doi.org/10.3892/ijo.2014.2524