scholarly journals MIR-140 TARGETS LNCRNA DNAJC3-AS1 TO SUPPRESS CELL PROLIFERATION IN ACUTE MYELOID LEUKEMIA

2022 ◽  
Vol 14 (1) ◽  
pp. e2022005
Author(s):  
Hong Li ◽  
Kehong Bi ◽  
Saran Feng ◽  
Yan Yan Wang ◽  
Chuansheng Zhu

Objectives:MiR-140 and DNAJC3-AS1 have been proven to play critical roles in cancer biology, while their participations in acute myeloid leukemia (AML) are unclear. This study aimed to explore the role of miR-140 and DNAJC3-AS1 in AML. Methods:Analysis of the expression of DNAJC3-AS1 and miR-140 was done with RT-qPCR. The role of DNAJC3-AS1 and miR-140 in the expression of each other was explored with overexpression assay. The direct interaction between DNAJC3-AS1 and miR-140 was analyzed with RNA pull-down assay. The subcellular location of DNAJC3-AS1 was explored with cellular subcellular fractionation assay. Cell proliferation analysis was done with BrdU assay. Results:AML patients showed increased expression of DNAJC3-AS1 and decreased expression of miR-140. DNAJC3-AS1 was detected in both nuclear and cytoplasm samples, and a direct interaction between DNAJC3-AS1 and miR-140 was observed.   Discussion:Reduced expression of DNAJC3-AS1 was observed after miR-140 overexpression in AML cells. DNAJC3-AS1 increased cell proliferation and inhibited the role of miR-140 in suppressing cell proliferation. Conclusion:In conclusion, miR-140 may target DNAJC3-AS1 to suppress cell proliferation in AML.

Blood ◽  
2021 ◽  
Author(s):  
Huan Cai ◽  
Makoto Kondo ◽  
Lakshmi Sandhow ◽  
Pingnan Xiao ◽  
Anne-Sofie Johansson ◽  
...  

Impairement of normal hmatopoiesis and leukemia progression are two well-linked processes during leukemia development and controlled by the bone marrow (BM) niche. Extracellular matrix proteins including laminin are important BM niche components. However, their role in hematopoiesis regeneration and leukemia is unknown. Laminin α4 (Lama4), a major receptor-binding chain of several laminins, is altered in BM niches in mice with acute myeloid leukemia (AML). So far, the impact of Lama4 on leukemia progression remains unknown. We here report that Lama4 deletion in mice resulted in impaired hematopoiesis regeneration following irradiation-induced stress, which is accompanied with altered BM niche composition and inflammation. Importantly, in a transplantation-induced MLL-AF9 AML mouse model, we demonstrate accelerated AML progression and relapse in Lama4-/-mice. Upon AML exposure, Lama4-/- mesenchymal stem cells (MSCs) exhibited dramatic molecular alterations including upregulation of inflammatory cytokines that favor AML growth. Lama4-/- MSCs displayed increased anti-oxidant activities and promoted AML stem cell proliferation and chemoresistance to cytarabine, which was accompanied by increased mitochondrial transfer from the MSCs to AML cells and reduced reactive oxygen species in AML cells in vitro. Similarly, we detected lower levels of reactive oxygen species in AML cells from Lama4-/- mice post-cytarabine treatment. Notably, LAMA4 inhibition or knockdown in human MSCs promoted human AML cell proliferation and chemoprotection. Together, our study for the first time demonstrates a critical role of Lama4 in impeding AML progression and chemoresistance. Targeting Lama4 signaling pathways may offer potential new therapeutic options for AML.


Author(s):  
Banban Li ◽  
Ruinan Jia ◽  
Wei Li ◽  
Ying Zhou ◽  
Dongmei Guo ◽  
...  

BackgroundChemoresistance is emerging as a major barrier to successful treatment in acute myeloid leukemia (AML), and bone marrow stromal cells (BMSCs) protect leukemia cells from chemotherapy eventually leading to recurrence. This study was designed to investigate the role of p21-activated kinase 1 (PAK1) in AML progression and chemosensitivity, highlighting the mechanism of stroma-mediated chemoresistance.MethodsThe GEPIA and TCGA datasets were used to analyze the relationship between PAK1 mRNA expression and various clinical parameters of AML patients. Cell proliferation and apoptosis were examined to evaluate the role of PAK1 on chemosensitivity in AML by silencing PAK1 with shRNA or small molecular inhibitor. Human BMSC (HS-5) was utilized to mimic the leukemia bone marrow microenvironment (BMM) in vitro, and co-culture model was established to investigate the role of PAK1 in BMSC-mediated drug resistance.Resultsp21-activated kinase 1 high expression was shown to be associated with shorter overall survival in AML patients. The silence of PAK1 could repress cell proliferation, promote apoptosis, and enhance the sensitivity of AML cells to chemotherapeutic agents. More importantly, BMSCs induced PAK1 up-regulation in AML cells, subsequently activating the ERK1/2 signaling pathway. The effect of BMSC-mediated apoptotic-resistance could be partly reversed by knock down of PAK1.Conclusionp21-activated kinase 1 is a potential prognostic predictor for AML patients. PAK1 may play a pivotal role in mediating BMM-induced drug resistance, representing a novel therapeutic target in AML.


2020 ◽  
Author(s):  
Ping Qiang ◽  
Chao Fang ◽  
Kaidi Song ◽  
Lan Shi ◽  
Yuanyuan Dai ◽  
...  

Abstract Background: Our previous studies have demonstrated that Luks-PV have good anti-leukemia ability effects and could possibly be a promising therapy for adult acute myeloid leukemia (AML). Aberrant over-expression of HOXA9 is a prominent feature of AML driven by multiple oncogenes, and therapeutic degrading of HOXA9 may be an effective treatment strategy in AML. This paper focused on the Luks-PV-regulating autophagy pathway, aiming to investigate the role of Luks-PV in mixed-lineage leukemia (MLL)-rearranged AML. Methods: The data of leukemia patients were downloaded from the gene expression profiling of TCGA datasets. Taking primary AML and THP-1 cells as the model system in vitro, Luks-PV-inhibited cell proliferation was determined by CCK‐8 and flow cytometry assays. The role of Luks-PV in autophagy regulation was analyzed using immunoblotting, transfection and immunofluorescence.Results: HOXA9 was over-expressed and associated with a poor prognosis in AML patients bearing MLL rearrangement. After the application of pharmacologic inhibitors of autophagy, Luks-PV induced cytotoxic autophagy in AML cells, as suggested by biochemical and microscopy results. HOXA9 molecules were detectable within autophagosomes after Luks-PV treatment, indicating that autophagy induction accounted for the degradation of HOXA9. Moreover, Luks-PV-induced HOXA9 downregulation inhibited AML cell proliferation, suggesting that HOXA9 could be degraded through Luks-PV-induced autophagy.Conclusion: Luks-PV suppresses AML cell proliferation by inducing HOXA9 degradation.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1616-1616
Author(s):  
Martin Carroll ◽  
Tae Kon Kim ◽  
Kenichi Higashino ◽  
Alan M. Gewirtz

Abstract Signal transducer and activator of transcription (STAT) family proteins play crucial roles in the cytokine signaling pathways which regulate survival and proliferation of normal hematopoietic cells. However, the role of STAT proteins in regulating survival in leukemia remains poorly defined. STAT3 and STAT5, for example, have been reported to be constitutively activated in acute myeloid leukemia (AML) cells, however the physiologic significance of this activation is unknown. In order to better understand the role of STAT3 and STAT5 in AML biology, we studied their expression, activation, and requirement for cell growth in several AML cell lines and primary AML cells collected from patients at the University of Pennsylvania Cancer Center. We first confirmed the activation of STAT3 and STAT5 in primary AML cells by western blotting. An analysis of AML patient samples revealed elevated levels of constitutive STAT3 phosphorylation in 6 of 7 patient samples and constitutive STAT5 phosphorylation in 8 of 9 patient samples. In addition, 6 AML cell lines (K562, HL-60, MOLM-14, U937, KG-1, NB4) displayed constitutive STAT3 and STAT5 activation. In order to evaluate the functional significance of constitutive activation of STAT3 and STAT5 in AML cells, we designed and synthesized short interfering RNAs (siRNAs) to silence the expression of these proteins. For initial characterization, the siRNAs were delivered to MOLM-14 cells using an AMAXA nucleofector device (AMAXA, Inc. Gaithersburg, MD)(Program O-17/Solution V). Nucleofected siRNA diminished STAT3 expression by 85% at 24 hour but had little effect on cell proliferation (13%±3% decrease at 24 hour, 8%±4% at 48 hour, 5%±1% at 72 hour) compared to control siRNA treated cells. In contrast, STAT5 siRNA decreased STAT5 expression 80% at 24 hour compared to control treated cells but inhibited cell proliferation by 19%±1% at 24 hour, 22%±1% at 48 hour, 16%±3% at 72 hour in comparison to control siRNA treated cells suggesting a more important role for STAT5 in regulating cell proliferation. To study the effect of these siRNA molecules in primary AML cells, we first determined our ability to nucleofect primary cells by examining delivery efficiency of fluorescein labeled siRNA. Four different patient samples were evaluated and the mean ± SD of cells successfully transfected was 52%±12. Transfection of multiple AML patient samples with STAT3 siRNA decreased STAT3 expression but led to only modest decrease (6–18% at 48 hour, 7–36% at 72 hour) in AML cell survival. However, transfection of cells with STAT5 siRNA, but not control siRNA, led to a consistent decrease (25–54% at 48 hour, 21–60% at 72 hour) in AML cell survival. The decrease in survival was proportional to the transfection efficiency in the different samples. These results provide the first evidence that STAT5 expression and activation is necessary for the survival of primary AML cells. In fact, the data suggests a greater role of STAT5 in survival of primary cells than in survival of AML cell lines. Accordingly, STAT5 appears to be a legitimate target for the treatment of AML.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3674-3674
Author(s):  
Michihiro Kobayashi ◽  
Yuanshu Dong ◽  
Hao Yu ◽  
Yunpeng Bai ◽  
Sisi Chen ◽  
...  

Abstract The phosphatase of regenerating liver family of phosphatases, consisting of PRL1, PRL2 and PRL3, represents an intriguing group of proteins implicated in cell proliferation and tumorigenesis. However, the role of PRLs in normal and malignant hematopoiesis is largely unknown. While SCF/KIT signaling plays an important role in hematopoietic stem and progenitor cell (HSPC) maintenance, how SCF/KIT signaling is regulated in HSPCs remains poorly understood. We here report that PRL2 regulates HSPC maintenance through regulating SCF/KIT signaling. To define the role of PRL2 in hematopoiesis, we analyzed the hematopoietic stem cell (HSC) behavior in Prl2 deficient mice generated by our group. Prl2 deficiency results in ineffective hematopoiesis and impairs the long-term repopulating ability of HSCs. In addition, Prl2 null HSPCs are less proliferative and show decreased colony formation in response to SCF stimulation. Furthermore, Prl2 null HSPCs show reduced activation of the PI3K/AKT and ERK signaling in steady state and following SCF stimulation. Importantly, we found that PRL2 associates with KIT and the ability of PRL2 to enhance SCF signaling depends on its enzymatic activity, demonstrating that PRL2 mediates SCF/KIT signaling in HSPCs. Thus, PRL2 plays a critical role in hematopoietic stem and progenitor cell maintenance through regulating SCF/KIT signaling. Furthermore, loss of Prl2 decreased the ability of oncogenic KITD814V mutant in promoting hematopoietic progenitor cell proliferation and in activation of signaling pathways. We also checked the expression of PRL2 proteins in human AML cell lines and found increased level of PRL2 proteins in some acute myeloid leukemia (AML) cells compared with normal human bone marrow cells, indicating that PRL2 may play a pathological role in AML. Our results suggest that the PRL2 phosphatase may be a druggable target in myeloproliferative disease (MPD) and acute myeloid leukemia (AML) with oncogenic KIT mutations. Disclosures: No relevant conflicts of interest to declare.


2020 ◽  
Vol 40 (4) ◽  
Author(s):  
Yi Xiao ◽  
Taoran Deng ◽  
Xi Ming ◽  
Jinhuang Xu

Abstract Tripartite motif (TRIM) 31 is a member of TRIM family and exerts oncogenic role in the progression and drug resistance of several cancers. However, little is known about the relevance of TRIM31 in acute myeloid leukemia (AML). Herein, we investigated the role of TRIM31 in AML. We examined the expression levels of TRIM31 in the blood samples from 34 patients with AML and 34 healthy volunteers using qRT-PCR. The mRNA levels of TRIM31 in human bone marrow stromal cells (HS-5) and five AML cell lines were also detected. Loss/gain-of-function assays were performed to assess the role of TRIM31 in AML cells proliferation, apoptosis and sensitivity to daunorubicin. The expression levels of pro-caspase 3, cleaved caspase 3, Wnt3a, β-catenin, cyclin D1 and c-Myc were measured using Western blot. TRIM31 expression levels were significantly up-regulated in AML patients and cell lines. Knockdown of TRIM31 suppressed cell proliferation and promoted apoptosis in AML-5 and U937 cells. The IC50 of daunorubicin was significantly decreased in TRIM31 siRNA (si-TRIM31) transfected cells. Oppositely, induced cell proliferation and decreased cell apoptosis were observed in pcDNA-3.1-TRIM31 transfected cells. Furthermore, knockdown of TRIM31 suppressed the activation of Wnt/β-catenin pathway in AML cells. Activation of Wnt/β-catenin pathway by LiCl abolished the effects of si-TRIM31 on cell proliferation, apoptosis and sensitivity to daunorubicin in AML cells. In conclusion, the results indicated that TRIM31 promoted leukemogenesis and chemoresistance to daunorubicin in AML. The oncogenic role of TRIM31 in AML was mediated by the Wnt/β-catenin pathway. Thus, TRIM31 might serve as a therapeutic target for the AML treatment.


2019 ◽  
Vol XIV (1) ◽  
Author(s):  
A.M. Radzhabova ◽  
S.V. Voloshin ◽  
I.S. Martynkevich ◽  
A.A. Kuzyaeva ◽  
V.A. Shuvaev ◽  
...  

2021 ◽  
Vol 10 (1) ◽  
Author(s):  
Yiyi Yao ◽  
Fenglin Li ◽  
Jiansong Huang ◽  
Jie Jin ◽  
Huafeng Wang

AbstractDespite the advances in intensive chemotherapy regimens and targeted therapies, overall survival (OS) of acute myeloid leukemia (AML) remains unfavorable due to inevitable chemotherapy resistance and high relapse rate, which mainly caused by the persistence existence of leukemia stem cells (LSCs). Bone marrow microenvironment (BMM), the home of hematopoiesis, has been considered to play a crucial role in both hematopoiesis and leukemogenesis. When interrupted by the AML cells, a malignant BMM formed and thus provided a refuge for LSCs and protecting them from the cytotoxic effects of chemotherapy. In this review, we summarized the alterations in the bidirectional interplay between hematopoietic cells and BMM in the normal/AML hematopoietic environment, and pointed out the key role of these alterations in pathogenesis and chemotherapy resistance of AML. Finally, we focused on the current potential BMM-targeted strategies together with future prospects and challenges. Accordingly, while further research is necessary to elucidate the underlying mechanisms behind LSC–BMM interaction, targeting the interaction is perceived as a potential therapeutic strategy to eradicate LSCs and ultimately improve the outcome of AML.


Sign in / Sign up

Export Citation Format

Share Document