scholarly journals PROLIFERATION AND APOPTOSIS OF B-CELL LYMPHOMA CELLS UNDER TARGETED REGULATION OF FOXO3 BY miR-155

2020 ◽  
Vol 12 (1) ◽  
pp. e2020073
Author(s):  
Xiaoqiang Zheng ◽  
Hongbing Rui ◽  
Ying Liu ◽  
Jinfeng Dong

This study aimed to explore the proliferation and apoptosis of B-cell lymphoma cells under targeted regulation of FOXO3 by miR-155. We analyzed the differences between B-cell lymphoma cells and B lymphocytes in expressions of miR-155 and FOXO3, explored the effects of miR-155 on proliferation and apoptosis of B-cell lymphoma cells, and relevant mechanisms, and also analyzed the relationship between expressions of miR-155 and FOXO3 in 42 patients with diffuse large B-cell lymphoma (DLBCL) and clinical characteristics of them. B-cell lymphoma cells showed a higher expression of miR-155 and a low expression of FOXO3 than B lymphocytes (both P<0.05). B-cell lymphoma cells transfected with miR-155-inhibitor showed significantly decreased expression of miR-155, significantly weakened cell proliferation ability and increased cell apoptosis rate (all P<0.05), and they also showed up-regulated expression of FOXO3 (P<0.05). Dual luciferase reporter assay revealed that there were targeted binding sites between miR-155 and FOXO3. Compared with B-cell lymphoma cells transfected with miR-155-inhibitor alone, those with co-transfection showed lower expression of FOXO3, higher proliferation and lower cell apoptosis rate (all P<0.05). The expression of miR-155 in DLBCL tissues was higher than that in tumor-adjacent tissues (P<0.05), and the expressions of miR-155 and FOXO3 were closely related to the international prognostic index (IPI) and the 5-year prognosis and survival of the patients (P<0.05). miR-155 can promote the proliferation of B-cell lymphoma cells and suppress apoptosis of them by targeted inhibiting FOXO3, and both over-expression of miR-155 and low expression of FOXO3 are related to poor prognosis of DLBCL patients.

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 3781-3781
Author(s):  
Hai-Jun Zhou ◽  
Archie Tamayo ◽  
Lan Pham ◽  
Yen-Chiu Lin-Lee ◽  
Richard J. Ford

Abstract CD40 plays important roles in the proliferation, survival and differentiation of lymphocytes. Constitutively active CD40 recruits TRAFs and IKKs within the lipid rafts to form a signalosome that mediates pivotal downstream proliferation and survival mechanisms involving NF-kB. Recently, we have reported that nuclear localization of CD40, through its interaction with c-Rel, promotes growth, cell cycle progression and survival in large B cell lymphoma. Our studies have opened a new paradigm in the functional role of CD40 in non-Hodgkin lymphomas of B cell origin (NHL-B). However, the mechanism about how CD40 enters nuclear still remains elusive. In this study, we show that CD40 ligation enhances its nuclear accumulation with activation of c-Rel in both normal B-lymphocytes and B cell lymphoma cells with cell fractionation assay and con-focal microscopy. Over-expression of c-Rel in B cell lymphoma cells drives CD40 into cell nucleus. We hypothesize that the route CD40 enters nucleus may involve endosome-endoplasmic reticulum-nuclear pore complex. Indeed, further studies show CD40 co-localizes with endosome marker-EEA1 and endoplasmic reticulum marker-Sec61. Furthermore, our co-immunoprecipitation assay has demonstrated CD40 interacts with Sec61. CD40 also co-localizes and immuno-precipitates with nuclear pore complex (NPC) proteins-NUP62 in normal B-lymphocytes and B lymphoma cells, which suggests NPC proteins may facilitate the nuclear translocation of CD40 protein. Overall, our study suggests that translocation of CD40 into cell nucleus involves multiple pathways. Blocking nuclear localization may modulate the function of CD40 in lymphoma cells; which could provide a new-targeted therapeutic approach for lymphoma therapy.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 464-464 ◽  
Author(s):  
Liat Nadav Dagan, ◽  
Xiaoyu Jiang ◽  
Izidore S Lossos

Abstract Abstract 464 HGAL is a germinal center (GC)-specific gene that inhibits lymphocyte and lymphoma cell motility and whose expression predicts improved survival of patients with diffuse large B-cell lymphoma (DLBCL) and classical Hodgkin lymphoma (cHL). Its specific expression only in the GC B-cells and GC-derived lymphomas suggest the existence of a tight regulatory process. We have previously reported that IL-4 and IL-13 can induce HGAL RNA and protein expression (Lossos et al, Blood. 2003; Natkunam et al, Blood. 2007) and at this meeting we also report that HGAL transcription is regulated by BLIMP1 protein (Cubedo et al). However, recent observations suggest that the fine-tuned control of gene expression, necessary for the immune cell differentiation-specific gene expression, is frequently accomplished by miRNAs. Therefore we have searched three prediction algorithms, PicTar (http://pictar.mdc-berlin.de/), miRanda (http://cbio.mskcc.org/ mirnaviewer/), and TargetScan (http://www.targetscan.org/) to identify miRNAs potentially regulating HGAL expression. We have focused on miR155, predicted to target HGAL in the TargetScan algorithm, since miR155 is overexpressed in aggressive activated B-cell like (ABC) type of DLBCL, characterized by low expression of HGAL. Furthermore, miR155 plays an important role in the regulation of GC reaction (Thai et al, Science. 2007) and its overexpression in mice B-lymphocytes leads to preleukemic B cell proliferation followed by full blown B-cell malignancy (Costinean et al, Proc Natl Acad Sci USA. 2006). To test HGAL regulation by the miR155, precursor of hsa-miR-155 was transfected into VAL and Raji lymphoma cells that express endogenous HGAL protein. Western blotting of whole-cell lysates showed a decrease of native HGAL in both cell lines compared with control miRNA transfection. Transfection of hsa-miR155 precursor into 3 lymphoma cell lines did not result in uniform decreases in HGAL mRNA expression levels, suggesting main regulation at the protein translation level. Transfection efficacy in each experiment was confirmed by measurements of miR155 by TaqMan MicroRNA Assay. To confirm direct effect on HGAL expression, we fused 2614 bp 3'-UTR sequences of HGAL containing two putative binding sites (M1 positions 2285–2291 and M2 positions 1844–1849) to a luciferase reporter gene. Cotransfection of the hsa-miR155 precursor with the luciferase reporter significantly repressed luciferase activity compared to a nontargeting control. To identify the binding site and to further demonstrate the specificity of the interaction, we generated luciferase reporter constructs with mutations in the M1 and M2 binding sites. Mutagenesis of M2 but not M1 site reversed the inhibitory effect of miR155, thus establishing M2 as the binding site. Since HGAL inhibits motility and chemotaxis of B lymphocytes and B cell lymphoma cells, we next examined effect of mir155 on SDF1 and IL-6-induced chemotaxis and spontaneous motility of VAL and Raji cells. Decrease in HGAL protein levels by hsa-miR155 precursor led to increased cell chemotaxis, cell motility, and cell velocity and was associated with decreased actin polymerization (F-actin content). The-miR155 effect on chemotaxis was rescued by transfection of HGAL-encoding vector. Since HGAL exerts its motility inhibitory effects by activation of RhoA, we next evaluated effect of miR155 on levels of activated GTP-bound RhoA. Overexpression of miR155 resulted in decreased levels of GTP-bound RhoA in both Raji and Val cell lines. Our findings demonstrate that miR155 may not only lead to oncogenic transformation of B lymphocytes, but also increases lymphoma cell motility by down regulation of HGAL expression. This effect may contribute to lymphoma cell dissemination and aggressiveness, characteristic of ABC-like DLBCL typically expressing high levels of miR155 and lacking HGAL expression. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1391-1391 ◽  
Author(s):  
Cristina Bertolo ◽  
Raquel Malumbres ◽  
Ainara Sagardoy ◽  
Eloy F Robles ◽  
Jose I Martinez-Ferrandis ◽  
...  

Abstract Abstract 1391 LITAF was discovered as a p53-induced transcript that promoted TNFa secretion in monocytes in response to LPS. We previously reported that LITAF is inactivated by deletion or promoter hypermethylation in germinal center-derived B-cell lymphomas. However, the function of LITAF in B lymphocytes is unknown. Using gene expression analysis of isolated B-cell subpopulation and immunohistochemical studies of tonsil lymphoid follicles we found that LITAF is expressed in naïve B lymphocytes and is repressed within the germinal centers (GCs). Thus, LITAF showed an opposite expression to BCL6, an essential regulator of GC development and function. Likewise, expression of LITAF and BCL6 were inversely correlated in cell lines and biopsies from patients with B-cell lymphoma, further suggesting a link between LITAF and BCL6. ChIP-on-chip and ChIP-sequencing analyses of B cells coupled with luciferase reporter assays revealed that BCL6 repressed LITAF expression by binding to its promoter. Accordingly, BCL6 silencing with siRNAs or after exposure to a BCL6-inhibitor peptide increased LITAF expression, indicating that LITAF is transcriptionally repressed by BCL6 in GC B lymphocytes and in B-cell lymphoma cells. To initially elucidate the function of LITAF in B cells, gain-and-loss of function experiments were performed in different cellular models. LITAF expression was not related to TNFa secretion after LPS exposure, nor modulated cell proliferation or apoptosis in B cells. However, sustained expression of LITAF in B-cell lymphoma cells increased cell size, lysosome content and mitochondrial mass. Gene expression microarray studies defined a LITAF-related transcriptional signature containing genes involved in the regulation of endomembranes, vesicle trafficking and protein transport. Accordingly, immunofluorescence analysis co-localized LITAF with lysosomes and with autophagosomes expressing LC3, the mammalian homolog of yeast autophagy-related protein (Atg8), as well as with the lysosomal sorting-associated proteins NEDD4 and TSG101, both in normal CD19+ B lymphocytes and in B-cell lymphoma cells. In addition, LITAF expression induced autophagic activity in B cells, shown by an increase in the FL1/FL3 ratio after acridine orange staining and by converting LC3-I to LC3-II, which were more evident upon cell starvation. Together, these data suggest that LITAF may play a role in the processing of proteins in autophagosomes through regulating autophagy. To investigate LITAF function in vivo, we generated mice with targeted deletion of the Litaf gene in B lymphocytes by using the Cre-loxP system. Litaf -mb1-Cre (Litaf−/− ) mice developed healthy and showed normal distribution of hematopoietic cell subpopulations. However, Litaf−/− mice were unable to develop full T-cell dependent immune responses, presenting PNA-stained, Litaf-negative GCs that were absent or had marked reduction in size and number. Accordingly, reduced amounts of IgM, IgG1 and IgG3 antibodies as a consequence of abnormal class switch recombination (CSR) were detected in immunized mice. However, in experiments testing CSR in vitro, in which B cells are artificially activated in the absence of T cells, the amounts of IgM/IgG1/IgG3 did not differ between knock-out and control groups. Similarly, mouse immunization with a T-cell independent antigen did not induce differences in immunoglobulin production. Further studies of GCs in T-cell immunized Litaf−/− mice using an antibody for the Class II-associated invariant chain peptide (CLIP) revealed that the atrophic GCs in Litaf−/− mice showed strong CLIP expression in comparison to wild-type littermates. In normal immune responses, CLIP peptides bind to MHC class II molecules in endolysosomes, until they are displaced by the antigen, then releasing CLIP and allowing MHC II-antigen complexes to be transported to the cell membrane for T-cell presentation. The failure to develop appropriate immune responses together with the accumulation of CLIP peptides in Litaf -deficient mice indicate that Litaf is essential for adequate T-cell dependent immune responses in GC B lymphocytes, possibly through facilitating the presentation of the antigens to MHC II molecules in the endolysosomes. Once this process is assembled and the T-cell activated B lymphocytes enter the GCs, BCL6 represses LITAF to prevent additional interactions between B and T cells during BCR editing. Disclosures: No relevant conflicts of interest to declare.


2020 ◽  
Vol 10 (6) ◽  
pp. 804-811
Author(s):  
Guangyao Hu ◽  
Dianxiu Wu

Oral squamous cell carcinoma (OSCC), a frequently happened cancer, is still an important threaten to human with unsatisfactory prognosis. Increasing evidence indicated that abnormal miRNA expressions were related to the development of OSCC. microRNA (miR)-4262 has been considered to be a cancer suppressor in various tumors, however its exact role in OSCC remains to be clarified. During the current research, we proposed to probe the biological activity and fundamental mechanism of miR-4262 in OSCC. The expression levels of miR-4262 in SCC9 and HOK cells were analyzed using quantitative real time polymerase chain reaction (qRT-PCR). TargetScan and luciferase reporter assay were carried out to quest the possible target gene of miR-4262. qRT-PCR and Western blotting analysis were employed to measure the expressions of B-cell lymphoma-2 (Bcl-2) in OSCC tissues, adjacent non-cancerous tissues, SCC9 and HOK cells. Cell proliferation and apoptosis of SCC9 cells were detected by Thiazolyl Blue Tetrazolium Bromide (MTT) and flow cytometry (FCM) analysis. The expressions of apoptosis-related proteins Bcl-2 and Bcl2-associated X protein (Bax) were checked by Western blotting analysis. Firstly, we found that miR-4262 expressed lower level in OSCC cells than that in the control. Results from TargetScan and luciferase reporter analysis showed that miR-4262 directly targeted Bcl-2. Then, up-regulated Bcl-2 was detected in OSCC tissues and cells compared with controls. Subsequently, Bcl-2-siRNA was found to be able to decrease cell viability while promote cell apoptosis in SCC9 cells, accompanied with the reduction of Bcl-2 and promotion of Bax. The Bcl-2/Bax ratio was reduced in Bcl-2-siRNA transfected group. In addition, miR-4262 mimic significantly suppressed the Bcl-2 expression, whereas the suppression was reversed by Bcl-2-plasmid. Furthermore, our results presented that miR-4262 mimic notably reduced cell viability and promoted cell apoptosis in SCC9 cell lines. Up-regulated miR-4262 could also downregulate Bcl-2 expression, upregulate the expression of Bax and decrease the Bcl-2/Bax ratio in SCC9 cells. However, Bcl-2-plasmid attenuated all these effects of miR-4262 mimic. Taken together, our findings indicated that miR-4262 exerted tumor-suppressive effects through targeting Bcl-2, resulting in promotion of cell apoptosis and inhibition of cell viability in OSCC cell lines. Therefore, miR-4262 might be a promising prognostic biomarker and novel therapeutic target during the therapy of OSCC.


2021 ◽  
Vol 39 (S2) ◽  
Author(s):  
J. Devin ◽  
T. Cañeque ◽  
Y.‐L. Lin ◽  
L. Mondoulet ◽  
J.‐L. Veyrune ◽  
...  

2012 ◽  
Vol 287 (27) ◽  
pp. 23184-23195 ◽  
Author(s):  
Gang Chen ◽  
Paritosh Ghosh ◽  
Thomas O'Farrell ◽  
Rachel Munk ◽  
Louis J. Rezanka ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document