Abstract Background Lichen striatus ( LS ) and linear lichen planus ( LLP ) are separate uncommon disorders belonging to linear inflammatory dermatoses. The immunotyping of inflammatory cells has been investigated in LS and lichen planus (LP), but epidermal proliferation and differentiation have little been described in LS and LLP . Methods The clinical and pathological data of eight patients with LS and seven with LLP were retrospectively collected. Immunotyping of infiltrated cells and expression of Ki‐67, K16, involucrin, and filaggrin were stained by immunohistochemistry in skin lesions of these patients and normal skin of eight healthy controls. Results Dermal infiltrates contained primarily CD3+ and CD68+ cells in three groups. CD4+ cells were predominantly located in the perivascular area, while CD8+ cells were frequently close to the junctional zone. Compared with control skin, epidermal and dermal CD1a+ cells, and dermal CD3+, CD4+, CD8+, and CD68+ cells were increased in LS and LLP ( P < 0.05), while Ki‐67+ cells were significantly high in LLP ( P < 0.05) but not in LS . K16 and involucrin expression in LLP were more extensive than in LS , and filaggrin expression was similar between both entities. Conclusions Our results indicate that the predominance of CD8+ cells and increased epidermal proliferation and abnormal keratinization are present in both dermatoses, although the levels of the above indexes are mild in LS as compared to LLP . These two entities might be due to the interaction of infiltrated cells and keratinocytes, and CD8+ cells could play a pivotal role in their pathogenesis.
The low efficiency of clustered, regularly interspaced, palindromic repeats-associated Cas (CRISPR/Cas) system editing genes in vivo limits the application. A components of the extracellular matrix (ECM), the extra domain A positive fibronectin (EDA+FN), may be a target for CRISPR/Cas system for the pro-oncogenic effects. The exclusion of EDA exon would alter the microenvironment and inhibit tumor progression, even the frequency of gene editing is still limited.The pro-oncogenic effects were confirmed by the exclusion of EDA exon from the fibronectin gene, as illustrated by the down-regulated proliferation, migration and invasion of CNE-2Z or SW480 cells (P<0.05). Furthermore, although the efficacy of EDA exon knockout through CRISPR/Cas system was shown to be low in vivo, the EDA+FN protein levels decrease obviously, inhibiting the tumor growth rate significantly (P<0.05), which was accompanied by a decrease in Ki-67 expression and microvessel numbers, and increased E-cadherin or decreased Vimentin expression (P<0.05).Human nasopharyngeal carcinoma cell line CNE-2Z, and the colorectal carcinoma cell line SW480 were transfected with CRISPR/Cas9 plasmids targeting EDA exon. The effects of the exclusion of EDA on the cell proliferation, motility and epithelial-mesenchymal transition (EMT) were investigated, and the western blot and real-time PCR were performed to analyze the underlying mechanisms. Furthermore, CRISPR/Cas9 plasmids were injected into xenograft tumors to knockout EDA exon in vivo, and tumor growth, cell proliferation, EMT rate, or vascularization were investigated using western blot, PCR and immunohistochemistry.CRISPR/Cas system targeting ECM components was shown to be an effective method for the inhibition of tumor progression, as these paracrine or autocrine molecules are necessary for various tumor cells. This may represent a novel strategy for overcoming the drug evasion or resistance, in addition, circumventing the low efficiency of CRISPR/Cas system in vivo.
Abstract Results from several studies demonstrate an inverse relationship between circulating levels of Vitamin D and risk of colorectal cancer. Eldecalcitol (1α,25-dihydroxy-2β-(3-hydroxypropyloxy) Vitamin D3; ED-71) is a novel analog of calcitriol, the most active form of Vitamin D3. ED-71 is more potent in stimulating bone remodeling and has been approved in Japan for the treatment of osteoporosis. The goal of this study was to assess the ability of ED-71 to inhibit spontaneous colorectal adenomas in a unique strain of multiple intestinal neoplasia (Apc+/Min-FCCC) mice. Male mice (6 wks of age) were randomized to treatment groups based on colon tumor status (endoscopic confirmation as tumor-free or -bearing) and body weight (bw) and administered: vehicle (MCT), calcitriol (0.25 μg/kg bw), or ED-71 (0.05 or 0.1 μg/kg bw) by gavage. Mice were treated every other day and bws were recorded weekly. After 14 wks of treatment, the small intestines and colons were excised and examined for gross tumors. Colorectal tumors >3 mm in diameter were frozen for gene expression analyses. The remaining tissue was fixed in formalin and processed for histological review. The multiplicity of gross small intestinal tumors in animals treated with ED-71 (both doses) was comparable to that of vehicle-treated controls. In contrast, the multiplicity of small intestinal tumors was elevated 34.6% over that of controls in animals treated with calcitriol (Mean ± SEM: 28.4 ± 2.6 vs. 21.1 ± 2.8, respectively; P = 0.034). The ability of ED-71 to inhibit colon tumorigenesis was evaluated independently in mice with vs. without tumors at treatment initiation. In tumor-bearing mice, neither calcitriol nor ED-71 (both doses) had any significant effect on the multiplicity of colorectal tumors as compared to control mice. However in tumor-free mice, ED-71 (0.1 μg/kg bw) reduced the mean incidence of adenomas (> 4 crypts) by 47% (ED-71 - 45.5%, controls 92.3%; P = 0.02) and the multiplicity by 47.6% (Mean ± SEM: ED-71 - 1.1 ± 0.39, controls - 2.1 ± 0.72; P > 0.05). In contrast, the multiplicity of microadenomas (≤ 4 crypts) was increased in mice treated with ED-71 (0.1 μg/kg bw) as compared to controls (0.9 ± 0.22 vs. 0.5 ± 0.25 P = 0.06, respectively). These findings suggest that ED-71 is effective in preventing the transition of microadenomas to mature adenomas. The lack of an effect of ED-71 on colon tumor development in mice bearing tumors at baseline could be due in part to loss of the Vitamin D receptor in colon adenomas, as confirmed by real-time PCR. Insight into the mechanism by which ED-71 inhibits adenoma development is being gained from analyses of Vitamin D signaling and microRNA expression in treated colonic epithelial cells. These promising data provide support for future studies to determine the potential utility of ED-71 in preventing colorectal cancer in high-risk patients found to be tumor-free during surveillance endoscopy. (Supported by NCI HHSN261201200015I) Citation Format: Wen-Chi L. Chang, Harry S. Cooper, Esther Kaunga, Lisa Vanderveer, Jing Peng, Suen S. Chen, Margie L. Clapper. ED-71, an analogue of Vitamin D3, blocks the promotion but not the initiation of colorectal tumors in the Apc+/Min-FCCC mouse model. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 5248.
Abstract LukS‐PV is one of the two components of Panton‐Valentine leucocidin (PVL). Our previous study showed that LukS‐PV can induce apoptosis in human acute myeloid leukemia (AML) THP‐1 and HL‐60 cells. C5aR (C5a receptor) is the receptor for PVL, but whether C5aR plays a key role in LukS‐PV induced apoptosis is unclear. The aim of this study was to establish whether C5aR plays a physiological role in apoptosis of leukemia cells induced by LukS‐PV. We investigated the role of C5aR in leukemia cell apoptosis induced by LukS‐PV by pretreatment of THP‐1 and HL‐60 cells with C5aR antagonist and transfection to knockdown C5aR in THP‐1 cells or overexpress C5aR in Jurkat cells before treatment with LukS‐PV. Cell apoptosis was analyzed by staining with Annexin V/propidium iodide or Annexin V‐PE/7‐AAD. Mitochondrial membrane potential (MMP) was determined using JC‐1 dye. The expression of apoptosis‐associated genes and proteins was identified by qRT‐polymerase chain reaction and Western blotting analysis, respectively. As the C5aR antagonist concentration increased, the rate of apoptosis induced by LukS‐PV decreased, the MMP increased, and expression of pro‐apoptotic Bax and Bak genes and proteins was downregulated while that of anti‐apoptotic Bcl‐2 and Bcl‐x genes and proteins was upregulated. Knockdown of C5aR also decreased LukS‐PV–induced THP‐1 cell apoptosis. LukS‐PV did not induce apoptosis of Jurkat cells, which have no endogenous C5aR expression; however, LukS‐PV did induce apoptosis in Jurkat cells after overexpression of C5aR. Correspondingly, the MMP decreased and Bax and Bak were upregulated while Bcl‐2 and Bcl‐x were downregulated. LukS‐PV can induce apoptosis in AML cells by targeting C5aR. C5aR may be a potential therapeutic target for AML and LukS‐PV is a candidate targeted drug for the treatment of AML.
Multiple myeloma (MM) is characterized by clonal proliferation of malignant plasma cells in the bone marrow. The anti-tumor activity of bortezomib (a proteosome inhibitor) in MM is challenged by emergence of drug resistance. MicroRNAs (miR) regulate and orchestrate multiple cellular pathways. We investigate the contribution miR-181a and miR-20a expressions' on cell proliferation and apoptosis in RPMI8226 cell line and their influence on bortezomib treatment. RNA isolation, quantitative real-time PCR (qRT-PCR), cell proliferation assay, cell cycle analysis, and cell apoptosis assay were done. Statistical analysis was performed using SPSS 17.0 software (SPSS, Chicago, IL, USA). P values of less than 0.05 were considered statistically significant. RPMI8226 cells seeded in 96-well plates and treated for 24 h with different concentrations of bortezomib showed dose-dependent growth inhibition; expression of both miR-181a and miR-20a were inhibited by bortezomib. We found decrease of miR-181a (60 %) and miR-20a (30 %) in cells transfected with 20-nM inhibitor. A relative increase of 14-fold in miR-181a and 11-fold in miR-20a was observed in cells transfected with mimics of the same concentration. Transient low expression of miR-181a/20a inhibited proliferation at day 4, and overexpression of miR-181a promoted proliferation. Cells transfected with miR-181a/20a inhibitor within day 4 showed lower survival rate, and low expression of miR-181a on the fourth day after transfection promoted apoptosis. Our findings suggest that miR-181a/20a has a higher expression in MM. miR-181-a expression is proportional to MM tumor burden and could be a biomaker for monitoring treatment. miR-20a shows the potential of a diagnostic biomarker.
Objective To determine the expressions of miR-155,miR-34a and miR-30a in diffuse large B-cell lymphoma and to explore their potential correlation with clinicopathological characteristics.Methods The expression level of miR 155,miR-34a and miR-30a in 46 DLBCL samples were determined with TaqMan real-time polymerase chain reaction.Interphase fluorescence in situ hybridization (I-FISH) was performed to detect MYC and p53 gene status and immunohistochemistry (Envision method) was used to evaluate the expression of CD3,CD10,CD20,BCL-6 and MUM-1 in DLBCL.The DLBCLs were classified into germinal center B cell-like (GCB) and non-germinal center B cell-like (non-GCB) subtypes according to Hans' criteria.Results Compared with normal controls,miR-155 expression level was significantly higher in DLBCL.The expression level of miR-155 in non-GCB type was higher than that in GCB type.It was shown that the patients with MYC rearrangement had lower expression level of miR-155 than the negative controls.Compared with p53 normal group,the expression level of miR-34a was significantly lower in p53 deletion group.It was also shown that the patients with BCL-6 protein positive had lower expression level of miR-30a than the negative group.Conclusion miR-155 expression level is different in normal controls,DLBCL and patients of subtype DLBCL.It therefore has a diagnosis value for DLBCL.miR-34a is of great prognostic significance.miR-155,miR-34a and miR-30a may be potential therapy targets for DLBCL.
Key words:
Diffuse large-cell lymphoma; Immunophenotype; MicroRNA; Gene expression
Recombinant interleukin-2 (rIL-2) is a pleiotropic cytokine that activates select immune effector cell responses associated with antitumor activity, including antibody-dependent cellular cytotoxicity (ADCC). Rituximab is an anti-CD20 monoclonal antibody that activates ADCC in non-Hodgkin lymphoma (NHL). The ability of rIL-2 to augment rituximab-dependent tumor responses was investigated. The efficacy of rIL-2 in combination with rituximab was evaluated in 2 NHL tumor xenograft models: the CD20hi, rituximab-sensitive, low-grade Daudi model and the CD20lo, aggressive, rituximab-resistant Namalwa model. Combination of rIL-2 plus rituximab was synergistic in a rituximab-sensitive Daudi tumor model, as evidenced by significant tumor regressions and increased time to tumor progression, compared with rIL-2 and rituximab single agents. In contrast, rituximab-resistant Namalwa tumors were responsive to single-agent rIL-2 and showed an increased response when combined with rituximab. Using in vitro killing assays, rIL-2 was shown to enhance activity of rituximab by activating ADCC and lymphokine-activated killer activity. Additionally, the activity of rIL-2 plus rituximab F(ab')2 was similar to that of rIL-2 alone, indicating a critical role for immunoglobulin G1 Fc-FcgammaR-effector responses in mediating ADCC. Antiproliferative and apoptotic tumor responses, along with an influx of immune effector cells, were observed by immunohistochemistry. Collectively, the data suggest that rIL-2 mediates potent tumoricidal activity against NHL tumors, in part, through activation and trafficking of monocytes and natural killer cells to tumors. These data support the mechanistic and therapeutic rationale for combination of rIL-2 with rituximab in NHL clinical trials and for single-agent rIL-2 in rituximab-resistant NHL patients.