21 research outputs found

    Malignant B Cells Induce the Conversion of CD4+CD25− T Cells to Regulatory T Cells in B-Cell Non-Hodgkin Lymphoma

    Get PDF
    Recent evidence has demonstrated that regulatory T cells (Treg) were enriched in the tumor sites of patients with B-cell non-Hodgkin lymphoma (NHL). However, the causes of enrichment and suppressive mechanisms need to be further elucidated. Here we demonstrated that CD4+CD25+FoxP3+CD127lo Treg were markedly increased and their phenotypes were different in peripheral blood (PB) as well as bone marrow (BM) from newly diagnosed patients with B-cell NHL compared with those from healthy volunteers (HVs). Involved lymphatic tissues also showed higher frequencies of Treg than benign lymph nodes. Moreover, the frequencies of Treg were significantly higher in involved lymphatic tissues than those from PB as well as BM in the same patients. Suppression mediated by CD4+CD25+ Treg co-cultured with allogeneic CFSE-labeled CD4+CD25− responder cells was also higher in involved lymphatic tissues from B-cell NHL than that mediated by Treg from HVs. In addition, we found that malignant B cells significantly induced FoxP3 expression and regulatory function in CD4+CD25− T cells in vitro. In contrast, normal B cells could not induce the conversion of CD4+CD25− T cells to Treg. We also showed that the PD-1/B7-H1 pathway might play an important role in Treg induction. Taken together, our results suggest that malignant B cells induce the conversion of CD4+CD25− T cells to Treg, which may play a role in the pathogenesis of B-cell NHL and represent a promising therapeutic target

    Pretreatment platelet count predicts survival outcome of patients with de novo non-M3 acute myeloid leukemia

    No full text
    Background Pretreatment platelet count has been reported as a potential tool to predict survival outcome in several solid tumors. However, the predictive value of pretreatment platelet count remains obscure in de novo acute myeloid leukemia (AML) excluding acute promyelocytic leukemia (M3). Methods We conducted a retrospective review of 209 patients with de novo non-M3 AML in our institute over a period of 8 years (2007–2015). Receiver operating characteristic (ROC) curve analysis was used to determine the optimal platelet (PLT) cutoff in patients. We analyzed the overall survival (OS) and disease free survival (DFS) using the log-rank test and Cox regression analysis. Results By defining the platelet count 50 × 109/L and 120 × 109/L as two cut-off points, we categorized the patients into three groups: low (120 × 109/L). On univariate analysis, patients with medium platelet count had longer OS and DFS than those with low or high platelet count. However, the multivariate analysis showed that only longer DFS was observed in patients with medium platelet count than those with low or high platelet count. Conclusion Our findings indicate that pretreatment platelet count has a predictive value for the prognosis of patients with non-M3 AML

    A novel miR-375-HOXB3-CDCA3/DNMT3B regulatory circuitry contributes to leukemogenesis in acute myeloid leukemia

    No full text
    Abstract Background Acute myeloid leukemia (AML) is a heterogeneous group of hematopoietic malignancies due to sophisticated genetic mutations and epigenetic dysregulation. MicroRNAs (miRNAs), a class of small non-coding RNAs, are important regulators of gene expression in all biological processes, including leukemogenesis. Recently, miR-375 has been reported to be a suppressive miRNA in multiple types of cancers, but its underlying anti-leukemia activity in AML is largely unknown. Methods Quantitative reverse transcriptase PCR (qRT-PCR) was used to measure the expression of miR-375 and HOXB3 in leukemic cells and normal controls. Targets of miR-375 were confirmed by western blot and luciferase assay. Phenotypic effects of miR-375 overexpression and HOXB3 knockdown were assessed using viability (trypan blue exclusion assay), colony formation/replating, as well as tumor xenograft assays in vivo. Results The expression of miR-375 was substantially decreased in leukemic cell lines and primary AML blasts compared with normal controls, because DNA hypermethylation of precursor-miR-375 (pre-miR-375) promoter was discovered in leukemic cells but not in normal controls. Lower expression of miR-375 predicted poor outcome in AML patients. Furthermore, forced expression of miR-375 not only decreased proliferation and colony formation in leukemic cells but also reduced xenograft tumor size and prolonged the survival time in a leukemia xenograft mouse model. Mechanistically, overexpression of miR-375 reduced HOXB3 expression and repressed the activity of a luciferase reporter through binding 3′-untranslated regions (3’-UTR) of HOXB3 mRNA. Overexpression of HOXB3 partially blocked miR-375-induced arrest of proliferation and reduction of colony number, suggesting that HOXB3 plays an important role in miR-375-induced anti-leukemia activity. Knockdown of HOXB3 by short hairpin RNAs reduced the expression of cell division cycle associated 3 (CDCA3), which decreased cell proliferation. Furthermore, HOXB3 induced DNA methyltransferase 3B (DNMT3B) expression to bind in the pre-miR-375 promoter and enhanced DNA hypermethylation of pre-miR-375, leading to the lower expression of miR-375. Conclusions Collectively, we have identified a miR-375-HOXB3-CDCA3/DNMT3B regulatory circuitry which contributes to leukemogenesis and suggests a therapeutic strategy of restoring miR-375 expression in AML

    Expression of CD56 is a risk factor for acute lymphocytic leukemia with central nervous system involvement in adults

    No full text
    <p><b>Objective:</b> To gain further insights into the predisposing risk factors for central nervous system (CNS) involvement in patients with acute lymphocytic leukemia (ALL), the impact of CD56 expression in these patients was investigated.</p> <p><b>Methods:</b> We reviewed the clinical features of CD56 expression in 588 consecutive ALL patients treated with systemic chemotherapy regimens between 2000 and 2014. The categorical data from CD56<sup>+</sup> ALL patients were compared with those from CD56<sup>−</sup> ALL patients.</p> <p><b>Results:</b> Among the 588 patients studied, 18.9% showed CD56 expression. The expression was significantly associated with CD33<sup>+</sup>, CD10<sup>−</sup>, CD15<sup>+</sup>, TdT<sup>−</sup>, and CD5<sup>+</sup> immunophenotypes. After systemic chemotherapy, 8.8% patients showed CNS involvement, of which 3.2% exhibited combined recurrences and 5.6% exhibited isolated CNS involvement. The 5-year event-free survival was significantly lower for patients with CD56<sup>+</sup> immunophenotype compared with patients with CD56<sup>−</sup> immunophenotype (22.5% vs. 32.7%, <i>P</i> = 0.04). Cumulative incidences of CNS involvement were significantly greater in the CD56<sup>+</sup> cohort compared with the CD56<sup>−</sup> cohort (14.4% vs. 7.5%, <i>P</i> = 0.02). Multivariate analysis revealed CD56 expression to be statistically significant risk factors for CNS involvement.</p> <p><b>Conclusion:</b> CD56 expression should be regarded as an independent risk factor for ALL with CNS involvement in adults.</p

    Additional file 8: of A novel miR-375-HOXB3-CDCA3/DNMT3B regulatory circuitry contributes to leukemogenesis in acute myeloid leukemia

    No full text
    Figure S5. The anti-leukemia effects of miR-375 in vivo. (A) Volumes of all tumors were detected every 3 days after 2 weeks. *P < 0.05 versus MSCV-NC. (B) HE staining was taken to indicate the infiltration of THP1 leukemic cells in murine spleens. (TIFF 367 kb

    Additional file 3: of A novel miR-375-HOXB3-CDCA3/DNMT3B regulatory circuitry contributes to leukemogenesis in acute myeloid leukemia

    No full text
    Figure S1. The expression of miR-126 and miR-375 in normal controls, CpG islands around the region encoding miR-375, and expression of pre-miR-375. (A) The expressions of miR-126 and miR-375 were detected in all CD34+ cells from 20 normal controls (NC). Housekeeping gene U6 is used as a reference. The lowest expression of miR-375 in one NC was set to 1.0 and then the expressions of miR-375 and miR-126 in all other specimens were normalized by this lowest specimen. The fold change of miR-375 and miR-126 were calculated by Student’s t-test. (B) CpG islands around the region encoding pre-miR-375 were analyzed by MethPrimer software. (C) The expression of pre-miR-375 was detected by qRT-PCR in HL-60 and THP1 cells, which were transduced with MSCV-miR-375 or MSCV-NC. *P < 0.01 versus MSCV-NC. (TIFF 221 kb

    Additional file 7: of A novel miR-375-HOXB3-CDCA3/DNMT3B regulatory circuitry contributes to leukemogenesis in acute myeloid leukemia

    No full text
    Figure S4. HOXB3 enhances the expression of DNMT3B to bind in pre-miR-375 promoter. (A) A schematic representation of the promoter regions amplified by ChIP-PCR assay. (B) HL-60 and THP1 cells were transduced with sh-NC or sh-HOXB3. Soluble chromatin from these cells was immunoprecipitated with anti-DNMT3B antibody. Immunoprecipitated DNA was analyzed by qRT-PCR. *P < 0.01 versus sh-NC. (C) The expression of miR-375 was detected in HL-60 and THP1 cells transduced with sh-NC or sh-HOXB3. *P < 0.01 versus sh-NC. (TIFF 310 kb
    corecore