Recombinant programmed cell death 1 inhibits psoriatic inflammation in imiquimod‑treated mice

  • Authors:
    • Shiguang Peng
    • Mei Cao
    • Xiaoying Sun
    • Yaqiong Zhou
    • Chu‑Yen Chen
    • Tian Ma
    • Hongjin Li
    • Bin Li
    • Bo Zhu
    • Xin Li
  • View Affiliations

  • Published online on: May 21, 2020     https://doi.org/10.3892/ijmm.2020.4612
  • Pages: 869-879
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Psoriasis is a common chronic inflammatory skin disease. Programmed cell death ligand 1 (PD‑L1) and programmed cell death 1 (PD‑1) are expressed on immune cells in a number of chronic inflammatory diseases. However, a limited number of studies have investigated the expression and function of the PD‑L1 and PD‑1 pathway in psoriatic inflammation. The present study used human psoriasis samples and imiquimod‑induced murine psoriasis models to investigate the potential role of PD‑1 in the modulation of psoriatic inflammation. The results demonstrated that inhibition of PD‑1 function with antibodies promoted psoriasis development. PD‑1‑fragment crystallizable (PD‑1‑Fc) treatment inhibited psoriatic inflammation and exhibited additive effects with anti‑tumor necrosis factor α therapy in imiquimod‑induced mouse psoriasis, suggesting that PD‑1‑Fc treatment may serve as a new therapeutic strategy for psoriasis.

Introduction

Patients with psoriasis exhibit chronic skin inflammation, which is characterized by thick and scaly plaques (1,2). Psoriasis is an autoimmune disease mediated by T cells; studies using a range of immune antagonists and human skin xenografts have identified that CD8+ and CD4+ T cells infiltrate into psoriatic skin lesions (1,3-6).

Programmed cell death 1 (PD-1), a member of the superfamily of cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) immunoglobulins involved in immune regulation, has attracted increasing attention in immune disease (7). PD-1 activation functions as a rheostat to regulate immune activities, inhibiting T-cell activation (8). PD-1 is activated by binding its ligands, programmed cell death ligand 1 (PD-L1) and PD-L2 (9-11), which signal T cells to inhibit cell proliferation, cytotoxicity and cytokine production (12). PD-1 cannot normally be detected on the surface of resting T cells; however, following activation by ligand binding to T-cell receptors, activated T cells express high levels of PD-1 (13,14). PD-1 signaling also affects cytokine production, including interferon γ (IFN-γ), interleukin 2 (IL-2) and tumor necrosis factor α (TNF-α) (12). Mice lacking PD-1 are resistant to viral infections, tumor growth and metastasis (15-17). Inhibiting PD-1 or its ligand PD-L1 has demonstrated promising benefits in metastatic melanoma treatment by overcoming the immunosuppressive tumor microenvironment and, consequently, has been approved by FDA for use in patients (18-20).

Traditional therapeutics for patients with psoriasis, including topical agents, systemic therapies and phototherapy, are not always sufficiently satisfactory; topical agents frequently result in short-term outcomes, and effective phototherapy treatment demands high requirements for clinician expertise and techniques to prevent skin erythema, photoaging or burning by inadequate choice of phototherapy types, parameters or unnecessary exposure (2,21). Despite the demonstrated efficacy of biologic immune therapeutics in psoriasis treatment, which consist of two main classes that target T cells and cytokines, inconsistent efficacy is still an issue in clinical trials (22). To maximize the immunotherapeutic efficacy of psoriasis treatments, there is an urgent need to understand the predominant mechanisms of pathogenesis to provide experimental support for innovative treatments and rational combinations.

The activation of PD-1 signaling has previously been used to block T-cell activation, proliferation and cytotoxicity (8,23). In addition, a previous study has demonstrated that PD-1 upregulation occurs in mouse psoriatic inflammatory skin induced by imiquimod (IMQ) (24). Treatment with recombinant PD-L1 protein significantly suppresses IMQ-induced psoriasis (25). Additionally, PD-1-null mice exhibit severe psoriatic inflammation (24). Thus, the hypothesis of the present study was that PD-1-targeted therapies may represent a promising new therapeutic strategy in psoriasis treatment.

Materials and methods

Characteristics of patients with psoriasis

Patients with psoriasis vulgaris were diagnosed according to characteristic skin changes and histopathological features. Prior to skin biopsy collection, systemic anti-psoriasis medications or ultraviolet phototherapy were discontinued for at least 8 weeks, and topical anti-psoriasis medications were discontinued for at least 4 weeks. No evidence of autoimmune disease other than psoriasis, malignant tumor and active viral or bacterial infection was present at the time of patient recruitment. Skin biopsies were collected from 10 patients [lesion skin; 6 (60%) males, 4 (40%) females; age, 30-72 years; mean age, 49 years] for IHC staining. The inflammatory level of psoriatic lesions was evaluated using The Psoriasis Area and Severity Index (PASI; range, 0-72) (26), which included four body regions: The head (h), trunk (t), upper extremities (u) and lower extremities (l); and the levels of erythema (E), infiltration (I), desquamation (D) and body surface area (A). The degree of severity was defined as 0 (no symptoms), 1 (slight), 2 (moderate), 3 (marked), or 4 (very marked), with the surface area defined as 1 (<10%), 2 (10-29%), 3 (30-49%), 4 (50-69%), 5 (70-89%) or 6 (90-100%). The PASI was calculated using the following formula: PASI = 0.1 x (Eh + Ih + Dh) x Ah + 0.2 x (Eu + Iu + Du) x Au + 0.3 x (Et + It + Dt) x At + 0.4 x (El + Il + Dl) x Al (26). All participants signed informed consent prior to enrollment. The study was approved by the Beijing Chaoyang Hospital Scientific and ethics Committee (approval no. 2016-11-4-5) and was conducted according to the Declaration of Helsinki.

Animal experiments

For all the mouse experiments, C57BL/6 mice (8-12 weeks old; Jackson Laboratory) were treated every day with a topical IMQ cream (62.5 mg in 5%; 3M Company) on the backs and ears, whereas the control mice were treated with Vaseline Lanette cream (Fagron, Inc.), and all mice were observed for the following 8 consecutive days as previously described (27). A total of 180 mice were used in this experiment. PASI scoring was used to quantify the erythema, thickness and scaling independently (score range, 0-4). The total score was calculated as previously described (27). The experimental mice were maintained under pathogen-free conditions, with ad libitum food and water. Mice were euthanized by inhalation of CO2 in a controllable manner at a flow rate of 15% volume displacement per minute, and the skin tissues were obtained and analyzed on the indicated days as described below.

Mouse in vivo experimental procedures

In the neutralizing anti-PD-1 mAb experiment, mice (8-12 weeks old) were intraperitoneally injected with 200 µg anti-PD-1 (RMP1-14) (n=20) or Igg (n=20) daily for 7 days starting when IMQ treatment was initiated. A total of 40 mice were used for this experiment. In the PD-1-Fc and anti-TNF-α intervention experiment, mice of the same age were intraperitoneally injected with 50 µg PD-1-Fc and/or anti-TNF-α on day 0 and day 3, starting at IMQ treatment initiation. each individual group had 20 mice (5 mice per time point), with 80 mice in total used for this experiment. All mice were monitored daily, and ear thickness was measured with a dial thickness gauge caliper daily. Mice were euthanized by inhalation of CO2, and the tissues were obtained and analyzed on the indicated day as described. All mouse experiments were approved by the Institutional Animal Care and Use Committee at Beijing Chaoyang Hospital in Capital Medical University in Beijing China (approval no. 2016-A-177).

Single-cell suspension preparation and flow cytometry

The isolated skin-draining inguinal lymph nodes from the mice were mashed with frosted glass slides and filtered through a 70-µm cell strainer to produce a single-cell suspension (1×107 cells/ml). The single-cell suspension was fixed with 2% paraformaldehyde for 15 min at room temperature prior to blocking with mouse Igg for 30 min on ice. Subsequently, the cells (1×106 in 100 µl) were incubated with anti-CD45 PerCP-Cy5.5 (cat. no. 45-0451-82; Invitrogen; Thermo Fisher Scientific, Inc.), anti-CD4 FITC (cat. no. RM4-5; BD Biosciences), anti-CD8 PE (cat. no. 553032; BD Biosciences) and anti-PD-1 Pe-Cy7 (cat. no. 25-9985-82; Invitrogen; Thermo Fisher Scientific, Inc.) antibodies on ice for 1 h. The cells were analyzed with an LSRII flow cytometer (BD Biosciences), and the data were analyzed with FlowJo software v10 (FlowJo, LLC).

Reverse transcription-quantitative (RT-q)PCR

Mouse skin was isolated for the extraction of total RNA using the RNeasy Mini kit (cat. no. 74104; Qiagen GmbH). The cDNA was synthesized with SuperScript II Reverse Transcriptase (Invitrogen; Thermo Fisher Scientific, Inc.) with dNTPs and poly(A) oligo(dT)25 as a template primer at 42°C for 50 min. A total of 40 ng cDNA was used as the template for the RT-qPCR assay with SyBR® green PCR master mix (Applied Biosystems; Thermo Fisher Scientific, Inc.) in a 25-µl reaction system using a Quantstudio 12K Flex qPCR System (Thermo Fisher Scientific, Inc.) under the following thermocycling conditions: Denaturation at 95°C for 15 sec and annealing/extension at 60°C for 1 min for total 40 cycles. Control RT-qPCR was performed using the master mix without reverse transcriptase. The relative gene expression levels were normalized to the expression of Gapdh. The comparative Ct method used was previously described (28). qPCR was performed three times.

The relative transcript levels of IL17, IFN-γ, IL-22, and TNFα were determined by RT-qPCR as previously described (29,30). The primers used were as follows: Mouse IL17 forward, 5′-TTT AACTCCCTTGGCGCAAAA-3′ and reverse, 5′-CTTTCCCTCCGCATTGACAC-3′; mouse IFN-γ forward, 5′-ATGAACGCTACACACTGC ATC-3′ and reverse, 5′-CCATCCTTTTGC CAGTTC CTC-3′; mouse TNFα forward, 5′-CCCTCACACTCAGATCATCTTCT-3′ and reverse, 5′-GTCACGACGTGG GCTACAG-3′; mouse IL22 forward, 5′-ATGAGTTTTTCCCTTATGGGGAC-3′ and reverse, 5′-GCTGGAAGTTGGACACCTCAA-3′; and mouse Gapdh forward, 5′-TGTGTCCGTCGTGGATCTGA-3′ and reverse, 5′-TTGCTGTTGAAGTCGCAGGAG-3′.

Antibodies, cytokines and in vivo treatment fusion proteins

The neutralizing monoclonal antibody (mAb) against PD-1 (clone RMP1-14; cat. no. BE0146) and Rat IgG2a (clone 2A3; cat. no. BE0089) in vivo were purchased from Bio X Cell. The PD-1-Fc protein (cat. no. 50124-M03H) was obtained from Sino Biological, Inc. Anti-mouse TNF-α (clone XT3.11; cat. no. BE0058) mAb was purchased from Bio X Cell. Anti-CD3 (cat. no. sc-20047), anti-CD8 (cat. no. sc-7188) and anti-CD4 (cat. no. sc-7219) antibodies were purchased from Santa Cruz Biotechnology, Inc. Anti-PD-1 and CD279 (cat. nos. PA5-2035 and PA5-32543) antibodies were purchased from Thermo Fisher Scientific, Inc., and the anti-PD-1 (clone J43; cat. no. 11-9985-81) antibody was purchased from eBioscience.

Histology and immunohistochemical (IHC) staining

Patient biopsies and mouse skin samples were fixed in formalin at room temperature for 24 h and embedded in paraffin. Hematoxylin and eosin (H&E) staining was performed as previously described (29). IHC staining was performed as previously described (29) with anti-CD4, anti-CD8 and anti-PD-1 antibodies. An olympus light microscope was used to examine the slides. Two independent researchers analyzed the H&E and IHC staining slides. epidermal thickness was assessed by quantification of keratinocyte numbers in the epidermis. Cells positive for CD4, CD8 and PD-1 were quantified as the mean number of positive cells in five high power fields (original magnification, ×400).

Western blotting and ELISA

Mouse back skin with psoriatic lesions after IMQ treatment for 6 days was isolated and lysed with RIPA buffer and protease inhibitor cocktail (Thermo Fisher Scientific, Inc.), followed by homogenization with a D1000 Handheld Homogenizer (Benchmark Scientific, Inc.) for ELISA with the IL-17 Mouse ELISA kit (cat. no. BMS6001; Thermo Fisher Scientific, Inc.) and IL-23 Mouse ELISA kit (cat. no. BMS6017; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions.

The skin lysis protein concentration was measured with the Braford Protein Assay kit I (Bio-Rad Laboratories, Inc.), and 50 µg total protein of each sample was loaded into each lane and separated with 12% SDS-PAGE. After transfer onto a PVDF membrane, 5% (weight/volume) 5% BSA (Sigma-Aldrich; Merck KGaA) was used for blocking at room temperature for 1 h, and then anti-IL17 (1:1,000, cat. no. ab218013; Abcam), anti-IL23 (1:400, cat. no. ab45420; Abcam), and the control horseradish peroxidase (HRP)-conjugated anti-β-actin (1:4,000, cat. no. ab49900, Abcam) antibodies were used to blot the membranes at 4°C overnight. After the 3 washes with 1X TBS buffer with 1% Tween-20 (TBST), a goat anti-rabbit Igg secondary antibody (cat. no. 1706515; Bio-Rad Laboratories, Inc.) was used to blot the membrane at room temperature for 2 h. Subsequently, 1X TBST was used to wash the membranes again prior to incubation with Pierce SuperSignal chemiluminescent substrate (Thermo Fisher Scientific, Inc.). The membranes were visualized by autoradiography into clear-blue X-ray film (cat. no. 34089; Thermo Fisher Scientific, Inc.), and the densitometry analysis was performed with Imagej 1.8.0 (national Institutes of Health).

Statistical analysis

The experiments were repeated three times. Data are presented as the mean ± SD. Statistical analysis was performed using GraphPad Prism 7 (graphPad Software, Inc.). Statistical significance was analyzed by a two-tailed unpaired Student's t-test. Correlation was determined with Pearson's correlation coefficient. For comparing multiple groups, one-way ANOVA and Tukey's post hoc test was performed. P<0.05 was considered to indicate a statistically significant difference.

Results

PD-1 is upregulated and correlates with markers of psoriatic inflammation

Immunohistochemical staining was performed on patient psoriatic tissue samples using specific anti-PD-1 antibodies. The expression of PD-1 was detectable in all psoriatic samples and was enhanced with increasing psoriatic inflammation (Fig. 1A). In addition, the numbers of CD4+ (Fig. 1B) and CD8+ (Fig. 1C) T cells increased in patients with high levels of psoriatic lesions. These results indicated an inflammation-associated role of PD-1 in psoriasis.

To identify the associations among epidermal hyperplasia, inflammation and PD-1 expression, epidermal thickness was measured by quantifying the keratinocyte number in the epidermis in five different regions to indicate psoriatic inflammation (31). The number of Pd-1+ cells was significantly correlated with the number of keratinocytes (Fig. 1D), which suggested that Pd-1+ cells were associated with the progression of skin inflammation. Furthermore, the number of PD-1+ cells was significantly correlated with the number of CD4+ and CD8+ T cells (Fig. 1E and F). Collectively, these results indicated that PD-1 expression levels were associated with skin inflammation in psoriasis.

PD-1 is upregulated in IMQ-treated mouse T cells

An IMQ-induced mouse psoriasis model was used to determine PD-1 expression. The representative skin inflammatory lesion was observed between days 0 and 8 (Fig. 2A). Inflammation was also indicated by the thickness of skin determined by H&E staining of epidermal samples from the backs and ears of the mice (Fig. 2B).

To analyze immune cell population changes, skin-draining lymph nodes were collected to detect the CD4+ and CD8+ T-cell populations in the IMQ-induced psoriasis mouse model using flow cytometry. The results demonstrated that the percentages of CD4+ and CD8+ T cells within the CD45+ cells increased following treatment with IMQ compared with those from mice treated with DMSO (Fig. 2C and D). In addition, the PD-1+ CD4+ population was significantly increased in IMQ-treated mice compared with those treated with DMSO (Fig. 2E and F). These results indicated that the level of PD-1 increased in psoriatic lesions, suggesting that PD-1 may serve a role in the modulation of psoriatic inflammation.

Anti-PD-1 treatment enhances psoriatic inflammation

To analyze PD-1 signaling in psoriasis progression in vivo, a PD-1-targeting intervention was used to treat IMQ-induced mouse psoriasis. Specifically, 200 µg/day neutralizing mAb against PD-1 was used to treat the IMQ-induced mice. Isotype-matched IgG was used as a negative control. PASI was determined every day to quantify the psoriatic inflammation and evaluate the progression of psoriasis (27). The results demonstrated that skin inflammation increased following IMQ treatment for 6 days and decreased after IMQ treatment for 8 days, and that anti-PD-1-treated mice exhibited exacerbated psoriatic lesions and higher cumulative PASI sores compared with those in the control group (Fig. 3A and B). Epidermal thickness for the PASI score, which indicates the level of psoriatic inflammation, was assessed using histopathological staining of the back skin psoriatic lesions (Fig. 3B and C). The results demonstrated increased inflammatory immune cell populations in mice treated with anti-PD-1 compared with those in the IgG group. In addition, compared with mice treated with IgG, mice treated with anti-PD-1 exhibited increased percentages of CD4+ and CD8+ T cells in the skin-draining lymph nodes on day 6 (Fig. 3D and E). The levels of cytokines were measured in psoriatic back skin isolated from mice treated with IgG or anti-PD-1 using RT-qPCR. The results demonstrated that enhanced cytokine expression, including that of IL-17, IL-22, IFN-γ and TNFα, was detected in mice with IMQ-induced psoriasis treated with anti-PD-1 compared with that in mice treated with IgG (Fig. 3F).

PD-1-Fc inhibits inflammation in the IMQ-induced psoriatic mouse model

The present study investigated the role of PD-1 in psoriasis inflammation and its therapeutic potential by determining the benefits of recombinant PD-1-Fc therapy on psoriasis progression in IMQ-induced psoriatic mice. The recombinant PD-1-Fc protein (50 µg per injection) exhibited a weak inflammatory response in IMQ-treated mouse (Fig. 4A). The PASI score was measured each day, and the results demonstrated that the PD-1-Fc-treated mice exhibited lower cumulative PASI scores compared with those in the control group (P<0.001 after treatment for 6 days; Fig. 4B). Histopathological examination of skin lesions confirmed that treatment with PD-1-Fc decreased the thickness of the epidermis of mouse back and ears (Fig. 4C). This result suggested that treatment with PD-1-Fc reduced psoriatic inflammation. Compared with the control mice, mice treated with PD-1-Fc exhibited decreased percentages of CD4+ and CD8+ T cells in the skin-draining lymph nodes (Fig. 4D and E). Cytokine levels were detected in mouse back psoriatic skin using RT-qPCR; the results revealed that PD-1-Fc treatment inhibited cytokine expression, including IL-17, IL-22, IFN-γ and TNFα, in psoriatic skin compared with that in the control mice (Fig. 4F).

PD-1-Fc and anti-TNF-α exert an additive effect to alleviate psoriatic inflammation

Anti-TNF-α therapy is used in psoriasis treatment to reduce psoriatic inflammation (32). The IMQ-induced psoriasis model was used to confirm the benefits of anti-TNF-α, and the resulted demonstrated that anti-TNF-α decreased the epidermal thickness of the mouse psoriatic ear, which is one of the key epidermal parameters to evaluate psoriasis development (Fig. 5A). In addition, the effects of co-treatment with anti-TNFα and PD-1-Fc were assessed. PD-1-Fc (50 µg per injection) alone or together with anti-TNFα (50 µg per injection) were administered to IMQ-treated mice with intraperitoneal injections on days 0 and 3, starting at IMQ treatment initiation; PD-1-Fc treatment enhanced the benefits of anti-TNF-α therapy, and the co-treatment resulted in weaker skin inflammation compared with that of anti-TnF-α treatment alone (Fig. 5B and C).

Flow cytometry was performed to analyze the percentage of CD4+ and CD8+ T cells in the skin-draining lymph nodes, and the results revealed that compared with anti-TNFα treatment alone, co-treatment with PD-1-Fc and anti-TNFa reduced the percentages of CD4+ and CD8+ T-cell in total CD45+ cells (Fig. 5D and E). To determine the potential function of PD-1-Fc in microenvironmental cytokine production, mouse back skin tissues were homogenized after 6 days of IMQ treatment for western blotting and ELISAs. The results demonstrated that the levels of IL-17 and IL-23 were reduced by co-treatment with Pd-1-Fc and anti-TNF-α compared with anti-TNF-α treatment alone (Fig. 5F). The ELISA results revealed that PD-1-Fc or anti-TNF-α alone significantly suppressed cytokine IL-17 and IL-23 production (Fig. 5G), which was in agreement with the previous conclusion. In addition, co-treatment with PD-1-Fc and anti-TNF-α further inhibited IL-17 and IL-23 production (Fig. 5G). These results suggested that recombinant PD-1-Fc may be a potential candidate for co-treatment with anti-TNFα in patients with psoriasis.

Discussion

The results of the present study identified a potential therapeutic strategy for patients with psoriasis. Traditional treatments are insufficient as topical agents usually function in the short-term; phototherapy treatment has high demands for physicians; and methotrexate, PUVA therapy, retinoids and cyclosporine are highly toxic to patients (2). Targeted biologic therapies are considered to be safer and more effective compared with generalized therapies (33). Through the analysis of clinical samples from patients with psoriasis, the present study identified that PD-1 was expressed in human psoriatic lesions, and upregulated PD-1 was associated with the level of psoriatic inflammation, suggesting a potential role for PD-1 in psoriasis development and progression. The results of the present study also demonstrated that Pd-1-Fc treatment effectively alleviated IMQ-induced psoriatic inflammation in mice.

T-lymphocyte activation is essential for the maintenance of psoriasis, and multiple mechanisms are involved in T-cell activation (34). PD-1 is bound by its ligands PD-L1 and PD-L2 and inhibits T-cell activity to prevent autoimmunity (23). However, it remains unclear whether deregulation of PD-1 expression is a hallmark of the progression of the autoimmune disease psoriasis (23). The PD-1/PD-L1 mechanism has been extensively studied in the context of understanding T-cell activation and immune checkpoint-targeted therapy (35-37). Immune checkpoint inhibition with anti-PD1 or anti-PD-L1 therapies results in psoriasis exacerbation (38,39). PD-1 expression is detected on the surface of several types of immune cells, including T cells, monocytes, dendritic cells (DCs), natural killer cells, B cells and macrophages (12,40). PD-L1 serves an important role in the regulation of T cell-mediated immunity (41,42). DCs express both the PD-1 receptor and PD-L1 to interact with cells expressing either PD-1 or PD-L1 (43). Pro-inflammatory cytokines and TLR ligands induce DC activation, during which high levels of PD-L1 expression are observed (44,45). PD-L1 is also expressed on the surface of CD4+ and CD8+ T cells to inhibit their activities (46). However, there is also a co-stimulatory interaction between PD-1 and PD-L1 to promote the development of memory CD4+ T cells (43). In addition, the function of Pd-1 signaling in the crosstalk between DCs and other effector cells, including γδ T cells, MDSCs and tumor-associated macrophages, is still unclear (43). The results of the present study demonstrated that PD-1-Fc negatively modulated psoriatic inflammation; the specific signaling pathway involved in this modulation needs to be explored in future studies.

Psoriasis can be induced or exacerbated by certain drugs, such as immune checkpoint inhibitors anti-PD-1 and small molecule TNF-α antagonists in cancer immunotherapy (47). A meta-analysis has reported that patients with psoriasis have a high risk of cancer incidence, as well as cancer-related death (48). The association between cancer and psoriasis may be related to inflammation; psoriasis is a chronic inflammatory skin disease, and chronic inflammation is associated with increased cancer risk (49). Immunomodulatory therapy for psoriasis treatment, which suppresses immunity and helps to reduce psoriasis symptoms, therefore, may decrease the risk of developing cancers (48). PD-L1 has been demonstrated to alleviate psoriatic inflammation, and PD-L1-Fc has exhibited promising benefits in psoriasis treatment (25). Studies have reported that PD-L1 levels serve a key role in the development of effective T cells (41,42).

PD-1 is a T-cell regulator that belongs to the CD28/CTLA-4 superfamily and negatively modulates T-cell activity (7). Treatment with soluble CTLA-4-Ig resulted in ≥50% improvement of Physician's global Assessment in clinical studies of psoriasis vulgaris (50). The mouse psoriasis model used in the present study was induced with IMQ, a ligand for Toll-like receptor (TLR) 7 and TLR8, which is used to induce immune activity and leads to mouse psoriasis; this mouse model is the most widely used inducible psoriasis model (27,51). Using this model, a previous study has revealed that PD-1 deficiency leads to enhanced dermal inflammation and increased expression of inflammatory cytokines, including IL-17 and IL-22, by innate γδ-low T cells in the IMQ-induced psoriasis mouse model (24). The results of the present study revealed that PD-1-Fc treatment effectively alleviated psoriatic inflammation in the IQM-induced mouse model and has the potential to exhibit synergistic effects with anti-TnF-α treatment.

Funding

This study was supported by the national natural Science Foundation of China (grant nos. 81874470 and 81973860), the national Key Research and Development Program of China (grant no. 2018yFC1705301) and the Shanghai natural Science Foundation of China (grant no. 19ZR1458700).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

SGP, BZ and XL conceived and designed the study. SGP, MC, XYS, YQZ, TM and HJL performed experimental work, and collected and analyzed the statistical data. SGP, BL, BZ and XL interpreted the results. SGP, MC, CYC, BZ and XL wrote and edited the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

This study was approved by the Institutional Review Boards of the Yueyang Integrated Traditional Chinese and Western Medicine Hospital (approval no. 2016-016). All mouse experiments were performed following procedures approved by the Institutional Animal Care and Use Committee at Beijing Chaoyang Hospital in Capital Medical university in Beijing China (approval no. 2016-A-177).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

References

1 

Bowcock AM and Krueger JG: Getting under the skin: The immunogenetics of psoriasis. Nat Rev Immunol. 5:699–711. 2005. View Article : Google Scholar : PubMed/NCBI

2 

Gottlieb AB: Psoriasis: emerging therapeutic strategies. Nat Rev Drug Discov. 4:19–34. 2005. View Article : Google Scholar : PubMed/NCBI

3 

Nickoloff BJ, Bonish B, Huang BB and Porcelli SA: Characterization of a T cell line bearing natural killer receptors and capable of creating psoriasis in a SCID mouse model system. j Dermatol Sci. 24:212–225. 2000. View Article : Google Scholar : PubMed/NCBI

4 

Chang JC, Smith LR, Froning KJ, Schwabe BJ, Laxer JA, Caralli LL, Kurland HH, Karasek MA, Wilkinson DI, Carlo DJ, et al: CD8+ T cells in psoriatic lesions preferentially use T-cell receptor V beta 3 and/or V beta 13.1 genes. Proc Natl Acad Sci USA. 91:9282–9286. 1994. View Article : Google Scholar

5 

Prinz JC, Vollmer S, Boehncke WH, Menssen A, Laisney I and Trommler P: Selection of conserved TCR VDJ rearrangements in chronic psoriatic plaques indicates a common antigen in psoriasis vulgaris. Eur J Immunol. 29:3360–3368. 1999. View Article : Google Scholar : PubMed/NCBI

6 

Vollmer S, Menssen A and Prinz JC: Dominant lesional T cell receptor rearrangements persist in relapsing psoriasis but are absent from nonlesional skin: evidence for a stable antigen-specific pathogenic T cell response in psoriasis vulgaris. J Invest Dermatol. 117:1296–1301. 2001. View Article : Google Scholar : PubMed/NCBI

7 

Ishida Y, Agata Y, Shibahara K and Honjo T: Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J. 11:3887–3895. 1992. View Article : Google Scholar : PubMed/NCBI

8 

Riley JL: PD-1 signaling in primary T cells. Immunol Rev. 229:114–125. 2009. View Article : Google Scholar : PubMed/NCBI

9 

Freeman GJ, Long AJ, Iwai Y, Bourque K, Chernova T, Nishimura H, Fitz LJ, Malenkovich N, Okazaki T, Byrne MC, et al: Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med. 192:1027–1034. 2000. View Article : Google Scholar : PubMed/NCBI

10 

Latchman Y, Wood CR, Chernova T, Chaudhary D, Borde M, Chernova I, Iwai Y, Long AJ, Brown JA, nunes R, et al: PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nat Immunol. 2:261–268. 2001. View Article : Google Scholar : PubMed/NCBI

11 

Sun X, Roudi R, Dai T, Chen S, Fan B, Li H, Zhou Y, Zhou M, Zhu B, Yin C, et al: Immune-related adverse events associated with programmed cell death protein-1 and programmed cell death ligand 1 inhibitors for non-small cell lung cancer: A PRISMA systematic review and meta-analysis. BMC Cancer. 19:5582019. View Article : Google Scholar : PubMed/NCBI

12 

Keir ME, Butte MJ, Freeman GJ and Sharpe AH: PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol. 26:677–704. 2008. View Article : Google Scholar : PubMed/NCBI

13 

Nishimura H, Agata Y, Kawasaki A, Sato M, Imamura S, Minato N, Yagita H, Nakano T and Honjo T: Developmentally regulated expression of the PD-1 protein on the surface of double-negative (CD4-CD8-) thymocytes. Int Immunol. 8:773–780. 1996. View Article : Google Scholar : PubMed/NCBI

14 

Agata Y, Kawasaki A, Nishimura H, Ishida Y, Tsubata T, Yagita H and Honjo T: expression of the PD-1 antigen on the surface of stimulated mouse T and B lymphocytes. Int Immunol. 8:765–772. 1996. View Article : Google Scholar : PubMed/NCBI

15 

Iwai Y, Ishida M, Tanaka Y, okazaki T, Honjo T and Minato N: Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc Natl Acad Sci USA. 99:12293–12297. 2002. View Article : Google Scholar : PubMed/NCBI

16 

Okazaki T, Chikuma S, Iwai Y, Fagarasan S and Honjo T: A rheostat for immune responses: The unique properties of PD-1 and their advantages for clinical application. Nat Immunol. 14:1212–1218. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Fourcade J, Sun Z, Benallaoua M, Guillaume P, Luescher IF, Sander C, Kirkwood JM, Kuchroo V and Zarour HM: upregulation of Tim-3 and PD-1 expression is associated with tumor antigen-specific CD8+ T cell dysfunction in melanoma patients. J Exp Med. 207:2175–2186. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Ott PA, Hodi FS and Robert C: CTLA-4 and PD-1/PD-L1 blockade: new immunotherapeutic modalities with durable clinical benefit in melanoma patients. Clin Cancer Res. 19:5300–5309. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, Powderly JD, Carvajal RD, Sosman JA, Atkins MB, et al: Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. New Engl J Med. 366:2443–2454. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Brahmer JR, Tykodi SS, Chow LG, Hwu WJ, Topalian SL, Hwu P, Drake CG, Camacho LH, Kauh J, odunsi K, et al: Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 366:2455–2465. 2012. View Article : Google Scholar : PubMed/NCBI

21 

Zhang P and Wu MX: A clinical review of phototherapy for psoriasis. Lasers Med Sci. 33:173–180. 2018. View Article : Google Scholar :

22 

Saurat JH, Stingl G, Dubertret L, Papp K, Langley RG, ortonne JP, Unnebrink K, Kaul M and Camez A; CHAMPIon Study Investigators: Efficacy and safety results from the randomized controlled comparative study of adalimumab vs. methotrexate vs placebo in patients with psoriasis (CHAMPIon). Br J Dermatol. 158:558–566. 2008. View Article : Google Scholar

23 

Francisco LM, Sage PT and Sharpe AH: The PD-1 pathway in tolerance and autoimmunity. Immunol Rev. 236:219–242. 2010. View Article : Google Scholar : PubMed/NCBI

24 

Imai Y, Ayithan N, Wu X, yuan Y, Wang L and Hwang ST: Cutting edge: PD-1 regulates imiquimod-induced psoriasiform dermatitis through inhibition of IL-17A expression by innate gammadelta-low T cells. J Immunol. 195:421–425. 2015. View Article : Google Scholar : PubMed/NCBI

25 

Kim JH, Choi YJ, Lee BH, Song My, Ban CY, Kim J, Park J, Kim Se, Kim TG, Park SH, et al: Programmed cell death ligand 1 alleviates psoriatic inflammation by suppressing IL-17A production from programmed cell death 1-high T cells. J Allergy Clin Immunol. 137:1466–1476.e3. 2016. View Article : Google Scholar

26 

Langley RG and Ellis CN: evaluating psoriasis with psoriasis area and severity index, psoriasis global assessment, and lattice system physician's global assessment. J Am Acad Dermatol. 51:563–569. 2004. View Article : Google Scholar : PubMed/NCBI

27 

van der Fits L, Mourits S, Voerman JS, Kant M, Boon L, Laman JD, Cornelissen F, Mus AM, Florencia E, Prens EP and Lubberts E: Imiquimod-induced psoriasis-like skin inflammation in mice is mediated via the IL-23/IL-17 axis. J Immunol. 182:5836–5845. 2009. View Article : Google Scholar : PubMed/NCBI

28 

Zhu B, Zhang M, Williams EM, Keller C, Mansoor A and Davie JK: TBX2 represses PTEN in rhabdomyosarcoma and skeletal muscle. Oncogene. 35:4212–4224. 2016. View Article : Google Scholar :

29 

Cui R, Widlund HR, Feige E, Lin JY, Wilensky DL, Igras VE, D'orazio J, Fung CY, Schanbacher CF, Granter SR and Fisher DE: Central role of p53 in the suntan response and pathologic hyperpigmentation. Cell. 128:853–864. 2007. View Article : Google Scholar : PubMed/NCBI

30 

D'orazio JA, nobuhisa T, Cui R, Arya M, Spry M, Wakamatsu K, Igras V, Kunisada T, granter SR, Nishimura EK, et al: Topical drug rescue strategy and skin protection based on the role of Mc1r in UV-induced tanning. Nature. 443:340–344. 2006. View Article : Google Scholar : PubMed/NCBI

31 

Works MG, Yin F, Yin CC, Yiu Y, Shew K, Tran TT, Dunlap N, Lam J, Mitchell T, Reader J, et al: Inhibition of TYK2 and JAK1 ameliorates imiquimod-induced psoriasis-like dermatitis by inhibiting IL-22 and the IL-23/IL-17 axis. J Immunol. 193:3278–3287. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Lubrano E, Scriffignano S and Perrotta FM: TNF-alpha inhibitors for the six treatment targets of psoriatic arthritis. Expert Rev Clin Immunol. 15:1303–1312. 2019. View Article : Google Scholar : PubMed/NCBI

33 

Ronholt K and Iversen L: old and new biological therapies for psoriasis. Int J Mol Sci. 18:E22972017. View Article : Google Scholar : PubMed/NCBI

34 

Sibaud V: Dermatologic reactions to immune checkpoint inhibitors: Skin toxicities and immunotherapy. Am J Clin Dermatol. 19:345–361. 2018. View Article : Google Scholar

35 

Muenst S, Soysal SD, Tzankov A and Hoeller S: The PD-1/ PD-L1 pathway: Biological background and clinical relevance of an emerging treatment target in immunotherapy. Expert Opin Ther Targets. 19:201–211. 2015. View Article : Google Scholar

36 

Okazaki T and Honjo T: The PD-1-PD-L pathway in immunological tolerance. Trends Immunol. 27:195–201. 2006. View Article : Google Scholar : PubMed/NCBI

37 

Li X, Xiang Y, Li F, yin C, Li B and Ke X: WnT/β-catenin signaling pathway regulating T cell-inflammation in the tumor microenvironment. Front Immunol. 10:22932019. View Article : Google Scholar

38 

Voudouri D, Nikolaou V, Laschos K, Charpidou A, Soupos N, Triantafyllopoulou I, Panoutsopoulou I, Aravantinos G, Syrigos K and Stratigos A: Anti-PD1/PDL1 induced psoriasis. Curr Probl Cancer. 41:407–412. 2017. View Article : Google Scholar : PubMed/NCBI

39 

De Bock M, Hulstaert E, Kruse V and Brochez L: Psoriasis vulgaris exacerbation during treatment with a PD-1 checkpoint inhibitor: Case report and literature review. Case Rep Dermatol. 10:190–197. 2018. View Article : Google Scholar : PubMed/NCBI

40 

He J, Hu Y, Hu M and Li B: Development of PD-1/PD-L1 pathway in tumor immune microenvironment and treatment for non-small cell lung cancer. Sci Rep. 5:131102015. View Article : Google Scholar : PubMed/NCBI

41 

Karunarathne DS, Horne-Debets JM, Huang JX, Faleiro R, Leow CY, Amante F, Watkins TS, Miles JJ, Dwyer PJ, Stacey KJ, et al: Programmed death-1 ligand 2-mediated regulation of the PD-L1 to PD-1 axis is essential for establishing CD4(+) T Cell Immunity. Immunity. 45:333–345. 2016. View Article : Google Scholar : PubMed/NCBI

42 

Ellis JS, guloglu FB, Tartar DM, Hoeman CM, Haymaker CL, Cascio JA, Wan X, Dhakal M, VanMorlan A, Yahng SH and Zaghouani H: APCs expressing high levels of programmed death ligand 2 sustain the development of CD4 T cell memory. J Immunol. 185:3149–3157. 2010. View Article : Google Scholar : PubMed/NCBI

43 

Versteven M, Van den Bergh JMJ, Marcq E, Smits ELJ, Van Tendeloo VFI, Hobo W and Lion E: Dendritic cells and programmed death-1 blockade: A joint venture to combat cancer. Front Immunol. 9:3942018. View Article : Google Scholar : PubMed/NCBI

44 

Ritprajak P and Azuma M: Intrinsic and extrinsic control of expression of the immunoregulatory molecule PD-L1 in epithelial cells and squamous cell carcinoma. Oral Oncol. 51:221–228. 2015. View Article : Google Scholar

45 

Pulko V, Liu X, Krco CJ, Harris KJ, Frigola X, Kwon ED and Dong H: TLR3-stimulated dendritic cells up-regulate B7-H1 expression and influence the magnitude of CD8 T cell responses to tumor vaccination. J Immunol. 183:3634–3641. 2009. View Article : Google Scholar : PubMed/NCBI

46 

Diskin B, Adam S, Cassini MF, Sanchez G, Liria M, Aykut B, Buttar C, Li E, Sundberg B, Salas RD, et al: PD-L1 engagement on T cells promotes self-tolerance and suppression of neighboring macrophages and effector T cells in cancer. Nat Immunol. 21:442–454. 2020. View Article : Google Scholar : PubMed/NCBI

47 

Balak DM and Hajdarbegovic E: Drug-induced psoriasis: Clinical perspectives. Psoriasis (Auckl). 7:87–94. 2017.

48 

Trafford AM, Parisi R, Kontopantelis E, Griffiths CEM and Ashcroft DM: Association of psoriasis with the risk of developing or dying of cancer: A systematic review and meta-analysis. JAMA Dermatol. 2019.Epub ahead of print. View Article : Google Scholar : PubMed/NCBI

49 

Coussens LM and Werb Z: Inflammation and cancer. Nature. 420:860–867. 2002. View Article : Google Scholar : PubMed/NCBI

50 

Abrams JR, Lebwohl MG, Guzzo CA, Jegasothy BV, Goldfarb MT, Goffe BS, Menter A, Lowe NJ, Krueger G, Brown MJ, et al: CTLA4Ig-mediated blockade of T-cell costimulation in patients with psoriasis vulgaris. J Clin Invest. 103:1243–1252. 1999. View Article : Google Scholar : PubMed/NCBI

51 

El Malki K, Karbach SH, Huppert J, Zayoud M, Reissig S, Schüler R, Nikolaev A, Karram K, Münzel T, Kuhlmann CR, et al: An alternative pathway of imiquimod-induced psoriasis-like skin inflammation in the absence of interleukin-17 receptor a signaling. J Invest Dermatol. 133:441–451. 2013. View Article : Google Scholar

Related Articles

Journal Cover

August-2020
Volume 46 Issue 2

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Peng S, Cao M, Sun X, Zhou Y, Chen CY, Ma T, Li H, Li B, Zhu B, Li X, Li X, et al: Recombinant programmed cell death 1 inhibits psoriatic inflammation in imiquimod‑treated mice. Int J Mol Med 46: 869-879, 2020
APA
Peng, S., Cao, M., Sun, X., Zhou, Y., Chen, C., Ma, T. ... Li, X. (2020). Recombinant programmed cell death 1 inhibits psoriatic inflammation in imiquimod‑treated mice. International Journal of Molecular Medicine, 46, 869-879. https://doi.org/10.3892/ijmm.2020.4612
MLA
Peng, S., Cao, M., Sun, X., Zhou, Y., Chen, C., Ma, T., Li, H., Li, B., Zhu, B., Li, X."Recombinant programmed cell death 1 inhibits psoriatic inflammation in imiquimod‑treated mice". International Journal of Molecular Medicine 46.2 (2020): 869-879.
Chicago
Peng, S., Cao, M., Sun, X., Zhou, Y., Chen, C., Ma, T., Li, H., Li, B., Zhu, B., Li, X."Recombinant programmed cell death 1 inhibits psoriatic inflammation in imiquimod‑treated mice". International Journal of Molecular Medicine 46, no. 2 (2020): 869-879. https://doi.org/10.3892/ijmm.2020.4612