scholarly journals Oncogenic Potential of the Dual-Function Protein MEX3A

Biology ◽  
2021 ◽  
Vol 10 (5) ◽  
pp. 415
Author(s):  
Marcell Lederer ◽  
Simon Müller ◽  
Markus Glaß ◽  
Nadine Bley ◽  
Christian Ihling ◽  
...  

MEX3A belongs to the MEX3 (Muscle EXcess) protein family consisting of four members (MEX3A-D) in humans. Characteristic for MEX3 proteins is their domain structure with 2 HNRNPK homology (KH) domains mediating RNA binding and a C-terminal really interesting new gene (RING) domain that harbors E3 ligase function. In agreement with their domain composition, MEX3 proteins were reported to modulate both RNA fate and protein ubiquitination. MEX3 paralogs exhibit an oncofetal expression pattern, they are severely downregulated postnatally, and re-expression is observed in various malignancies. Enforced expression of MEX3 proteins in various cancers correlates with poor prognosis, emphasizing their oncogenic potential. The latter is supported by MEX3A’s impact on proliferation, self-renewal as well as migration of tumor cells in vitro and tumor growth in xenograft studies.

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 468-468
Author(s):  
Sun-Mi Park ◽  
Raquel P Deering ◽  
Yuheng Lu ◽  
Tivnan Patrick ◽  
Varun Shenoy ◽  
...  

Hematopoiesis is a tightly regulated process in which different cell lineages of the blood and immune system are generated from hematopoietic stem cells (HSC). HSCs can self renew and also give rise to more differentiated progenitor cells through symmetric and asymmetric cell division respectively. Progenitors can further differentiate into more committed cells that can generate the mature lymphoid and myeloid compartments. In order to support a normal hematopoietic system HSCs must maintain normal cell fate decisions between symmetric and asymmetric divisions. Recent studies from our group and others have implicated Msi2 as a regulator of HSCs. Nevertheless, the exact role for Msi2 in HSCs and critical pathways regulated by Msi2 in these stem cells remains unclear. In addition to its high expression in normal HSC, MSI2 is upregulated in patients with poor clinical prognosis in acute myeloid leukemia and in the blast crisis phase of chronic myelogenous leukemia. To understand the role of Msi2 in normal HSCs, we are characterizing mice with a conditional deletion of Msi2 (Mx1-Cre::Msi2flox/flox mice). We previously demonstrated that the conditional ablation of Msi2 results in the failure of HSC maintenance due to a loss of quiescence and increased commitment divisions. Although Msi2 is critical for HSC engraftment, we observed a preferential requirement for Msi2 in the myeloid biased HSCs (My-HSCs). Based on the surface markers LSK CD34- CD150 high we found a ∼3 fold reduction in the frequency of My-HSCs and no differences in the frequency of the unbiased HSCs or in common lymphoid progenitors. Consistent with these results, we observed a dramatic decrease in chimerism in the myeloid versus lymphoid compartment of the Msi2 deleted cells in mice that were transplanted. In order to understand why hematopoietic stem and progenitors cells require Msi2, we previously performed global transcriptome profiling and RNA target analysis using high throughput sequencing of RNA isolated by crosslinking immunoprecipitation (HITS-CLIP). This overlap analysis implicated a variety of pathways including RNA translation, HSC self-renewal and TGF-β. The TGF-β pathway has been linked to the maintenance of both normal and leukemia stem cell self-renewal, and the control of myeloid biased HSCs. We examined the output of the TGF-β pathway in HSCs in the presence and absence of Msi2. We found reduced phosphorylation of Smad2/Smad3 in Msi2D/D HSCs grown in vitro or directly isolated from bone marrow. Recently, it has been reported that the loss of p57 in HSCs results in reduced quiescence and compensatory up-regulation of p27. Consistent with reduced signaling output of TGF-β, we observed a significant decrease in p57 expression and up-regulation of p27 in Msi2D/D LSKs. The effect of TGF-β treatment is biphasic in hematopoietic stem cells, with high levels of TGF-β blocking proliferation and low levels of this pathway activation leading to increased proliferation. We therefore examined the functional response of Msi2D/D HSCs to biphasic TGF-β signaling in vitro. Msi2D/D HSCs responded normally to high dose of exogenous TGF-β resulting in potent growth suppression. In contrast, when exposed to lower levels of TGF-β that has been shown to expand normal HSCs, Msi2D/D HSCs failed to respond, consistent with diminished sensitivity to the proliferative effects of TGF-β. These results demonstrate that the proliferative signals downstream of TGF-β signaling are impaired in the Msi2 deleted HSCs. Most intriguingly, our data suggests that Msi2 is required to maintain the response to TGF-β signaling and this contributes to the maintenance and fate of the My-HSCs. It will be interesting to find out if Msi2 is involved in the prevalence of My-HSCs during aging. Taken together, these results have important implications for understanding how the activity of RNA binding proteins contribute to HSC biology that govern normal tissue homeostasis. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2324-2324
Author(s):  
Sun Mi Park ◽  
Raquel Deering ◽  
Yuheng Lu ◽  
Patrick Tivnan ◽  
Varun Shenoy ◽  
...  

Abstract Abstract 2324 Hematopoietic stem cells (HSC) must maintain normal cell fate decisions between symmetric and asymmetric divisions as alterations can lead to hematopoietic malignancies. The MSI2 RNA binding protein is upregulated in patients with a poor clinical prognosis in acute myeloid leukemia and in the blast crisis phase of chronic myelogenous leukemia. The related RNA-binding protein Msi1 has been shown to block translation of specific target mRNAs by interacting with the 3'UTR. To understand the role of Msi2 in both normal and leukemic contexts, we are characterizing mice with a conditional deletion of Msi2 in the hematopoietic compartment. Msi2 conditional knockouts have reduced overall number of HSCs in the bone marrow and have decreased engraftment capacity in congenic transplants, however the molecular mechanisms through which Msi2 elicits these effects remains unclear. To understand how Msi2 alters HSC self renewal, we utilized Mx1-Cre::Msi2flox/flox conditional mice, enabling Msi2 inactivation via poly(I):poly(C) injection. We first tested if Msi2 deleted HSCs or MPPs had an intrinsic defect in cell fate decision and proliferation. We performed proliferation and colony assays on sorted HSCs and MPPs and found reduced cellular numbers in both the HSCs and MPPs with an increase in the frequency of more differentiated cells based on Mac/Gr1 staining after seven days. These reduced overall cell numbers in vitro may have resulted from a defect in maintaining the stem cell population or a decrease in proliferative capacity. To assess if there was a defect in the initial HSC cell division, we examined Numb protein levels and distribution as a surrogate readout for asymmetric cell division. Numb mRNA is a known target of Msi binding and translational repression. Surprisingly, in the Msi2 null HSC or MPPs we observed no difference in the overall staining of Numb indicating that Msi2 deficiency does not globally alter Numb levels. However, we did observe a decrease in the percentage of cells that underwent asymmetric Numb segregation in the MPPs and an increase in the percentage of cells that showed increased Numb staining in some of the daughter pairs in both HSCs and MPPs, indicating increased commitment away from the hematopoietic stem and progenitors. Although controversial, Notch signaling has been implicated in self renewal of HSCs and as a critical downstream target of the Msi family through Numb inactivation. However, the Notch signaling pathway was not noted to be significantly altered in gene set analysis from microarrays performed on Msi2 deleted HSC enriched populations. Moreover using quantitative PCR for Notch target genes in sorted Linlow, c-kit+ and Sca+(LSK) cells we found no statistical difference in the expression of Notch targets (Notch1, Notch2, Hes1 or Myc). These interesting negative results prompted us take a more global approach in characterizing the direct targets of Msi2 in hematopoietic cells. Due to the requirement for a large number of cells, we utilized K562 cells overexpressing a FLAG-tagged version of MSI2 to identify its direct RNA-binding targets. Using recently developed technologies, we performed experiments with UV-cross-linked and immunoprecipitated MSI2 which was then followed by RNA-sequencing (HITS-CLIP) to identify the global direct binding. Our analysis characterized the distribution of binding across the genome. Additionally, gene set enrichment analysis (GSEA) indicated a positive correlation of genes that were upregulated in the Msi2 deleted LSKs and the top 3-fold bound RNA targets (2,713 genes). Using the entire RNA target list we identified gene set signatures including “Cell Cycle”, “Self-renewal” and “HSC to CMP” that matched our results in the microarray from Msi2 deleted LSKs. In summary, direct RNA target analysis for MSI2 in human leukemia cells overlapped with self renewal and differentiation gene sets in mouse HSC enriched populations and correlated with the phenotypes we observed in isolated HSCs grown in vitro lacking Msi2. These results uncover the complexity of MSI2 RNA binding targets and have important implications for both normal and leukemic stem cell biology. Disclosures: Ebert: Celgene: Consultancy; Genoptix: Consultancy.


2018 ◽  
Author(s):  
Nan Tian ◽  
Wenbing Shangguan ◽  
Zuolin Zhou ◽  
Yao yao ◽  
Chunlei Fan ◽  
...  

AbstractChemoresistance remains a big challenge in hepatocellular carcinoma (HCC) treatment. Several studies indicated that RNA-binding protein Lin28B serves an oncogenic role in HCC, but its activity in HCC chemotherapy has never been assessed. In this study, we found that overexpression of Lin28B significantly increased the paclitaxel chemoresistance in two different HCC cells lines while silencing Lin28B reduced the chemoresistance in paclitaxel-resistance HCC cells. Curcumin, a natural anti-cancer agent, increased the sensitivity of HCC cells to paclitaxel through inhibiting NF-κB stimulated Lin28B expression both in vitro and in vivo. Furthermore, by analyzing TCGA (The Cancer Genome Atlas) LIHC (liver hepatocellular carcinoma) and GSE14520 databases, we found that Lin28B was highly up-regulated in HCC tissue compared with that in normal tissue and associated with α-fetoprotein levels, and that patients with Lin28B higher expression had a significant shorter overall survival time than those with Lin28B lower expression. Our data reveal that Lin28B may serve as a predictive biomarker and a treatment target to reverse HCC chemotherapy resistance in future clinical practice.Summary statementupregulation of Lin28B not only confers poor prognosis in HCC patients but also increases chemoresistance in HCC cells. Thus, Lin28B may serve as a predictive biomarker for use to reverse chemoresistance in clinical practice.


2021 ◽  
Vol 6 (1) ◽  
Author(s):  
Jun Liu ◽  
Jipeng Li ◽  
Ke Wang ◽  
Haiming Liu ◽  
Jianyong Sun ◽  
...  

AbstractFork-head box protein M1 (FoxM1) is a transcriptional factor which plays critical roles in cancer development and progression. However, the general regulatory mechanism of FoxM1 is still limited. STMN1 is a microtubule-binding protein which can inhibit the assembly of microtubule dimer or promote depolymerization of microtubules. It was reported as a major responsive factor of paclitaxel resistance for clinical chemotherapy of tumor patients. But the function of abnormally high level of STMN1 and its regulation mechanism in cancer cells remain unclear. In this study, we used public database and tissue microarrays to analyze the expression pattern of FoxM1 and STMN1 and found a strong positive correlation between FoxM1 and STMN1 in multiple types of cancer. Lentivirus-mediated FoxM1/STMN1-knockdown cell lines were established to study the function of FoxM1/STMN1 by performing cell viability assay, plate clone formation assay, soft agar assay in vitro and xenograft mouse model in vivo. Our results showed that FoxM1 promotes cell proliferation by upregulating STMN1. Further ChIP assay showed that FoxM1 upregulates STMN1 in a transcriptional level. Prognostic analysis showed that a high level of FoxM1 and STMN1 is related to poor prognosis in solid tumors. Moreover, a high co-expression of FoxM1 and STMN1 has a more significant correlation with poor prognosis. Our findings suggest that a general FoxM1-STMN1 axis contributes to cell proliferation and tumorigenesis in hepatocellular carcinoma, gastric cancer and colorectal cancer. The combination of FoxM1 and STMN1 can be a more precise biomarker for prognostic prediction.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Saikat Bhattacharya ◽  
Michaella J. Levy ◽  
Ning Zhang ◽  
Hua Li ◽  
Laurence Florens ◽  
...  

AbstractHeterogeneous ribonucleoproteins (hnRNPs) are RNA binding molecules that are involved in key processes such as RNA splicing and transcription. One such hnRNP protein, hnRNP L, regulates alternative splicing (AS) by binding to pre-mRNA transcripts. However, it is unclear what factors contribute to hnRNP L-regulated AS events. Using proteomic approaches, we identified several key factors that co-purify with hnRNP L. We demonstrate that one such factor, the histone methyltransferase SETD2, specifically interacts with hnRNP L in vitro and in vivo. This interaction occurs through a previously uncharacterized domain in SETD2, the SETD2-hnRNP Interaction (SHI) domain, the deletion of which, leads to a reduced H3K36me3 deposition. Functionally, SETD2 regulates a subset of hnRNP L-targeted AS events. Our findings demonstrate that SETD2, by interacting with Pol II as well as hnRNP L, can mediate the crosstalk between the transcription and the splicing machinery.


2021 ◽  
pp. 1-10
Author(s):  
Yu Wang ◽  
Han Zhao ◽  
Ping Zhao ◽  
Xingang Wang

BACKGROUND: Pyruvate kinase M2 (PKM2) was overexpressed in many cancers, and high PKM2 expression was related with poor prognosis and chemoresistance. OBJECTIVE: We investigated the expression of PKM2 in breast cancer and analyzed the relation of PKM2 expression with chemotherapy resistance to the neoadjuvant chemotherapy (NAC). We also investigated whether PKM2 could reverse chemoresistance in breast cancer cells in vitro and in vivo. METHODS: Immunohistochemistry (IHC) was performed in 130 surgical resected breast cancer tissues. 78 core needle biopsies were collected from breast cancer patients before neoadjuvant chemotherapy. The relation of PKM2 expression and multi-drug resistance to NAC was compared. The effect of PKM2 silencing or overexpression on Doxorubicin (DOX) sensitivity in the MCF-7 cells in vitro and in vivo was compared. RESULTS: PKM2 was intensively expressed in breast cancer tissues compared to adjacent normal tissues. In addition, high expression of PKM2 was associated with poor prognosis in breast cancer patients. The NAC patients with high PKM2 expression had short survival. PKM2 was an independent prognostic predictor for surgical resected breast cancer and NAC patients. High PKM2 expression was correlated with neoadjuvant treatment resistance. High PKM2 expression significantly distinguished chemoresistant patients from chemosensitive patients. In vitro and in vivo knockdown of PKM2 expression decreases the resistance to DOX in breast cancer cells in vitro and tumors in vivo. CONCLUSION: PKM2 expression was associated with chemoresistance of breast cancers, and could be used to predict the chemosensitivity. Furthermore, targeting PKM2 could reverse chemoresistance, which provides an effective treatment methods for patients with breast cancer.


Processes ◽  
2020 ◽  
Vol 9 (1) ◽  
pp. 45
Author(s):  
Ghmkin Hassan ◽  
Said M. Afify ◽  
Shiro Kitano ◽  
Akimasa Seno ◽  
Hiroko Ishii ◽  
...  

Defined by its potential for self-renewal, differentiation and tumorigenicity, cancer stem cells (CSCs) are considered responsible for drug resistance and relapse. To understand the behavior of CSC, the effects of the microenvironment in each tissue are a matter of great concerns for scientists in cancer biology. However, there are many complicated obstacles in the mimicking the microenvironment of CSCs even with current advanced technology. In this context, novel biomaterials have widely been assessed as in vitro platforms for their ability to mimic cancer microenvironment. These efforts should be successful to identify and characterize various CSCs specific in each type of cancer. Therefore, extracellular matrix scaffolds made of biomaterial will modulate the interactions and facilitate the investigation of CSC associated with biological phenomena simplifying the complexity of the microenvironment. In this review, we summarize latest advances in biomaterial scaffolds, which are exploited to mimic CSC microenvironment, and their chemical and biological requirements with discussion. The discussion includes the possible effects on both cells in tumors and microenvironment to propose what the critical factors are in controlling the CSC microenvironment focusing the future investigation. Our insights on their availability in drug screening will also follow the discussion.


Author(s):  
Zizhen Si ◽  
Lei Yu ◽  
Haoyu Jing ◽  
Lun Wu ◽  
Xidi Wang

Abstract Background Long non-coding RNAs (lncRNA) are reported to influence colorectal cancer (CRC) progression. Currently, the functions of the lncRNA ZNF561 antisense RNA 1 (ZNF561-AS1) in CRC are unknown. Methods ZNF561-AS1 and SRSF6 expression in CRC patient samples and CRC cell lines was evaluated through TCGA database analysis, western blot along with real-time PCR. SRSF6 expression in CRC cells was also examined upon ZNF561-AS1 depletion or overexpression. Interaction between miR-26a-3p, miR-128-5p, ZNF561-AS1, and SRSF6 was examined by dual luciferase reporter assay, as well as RNA binding protein immunoprecipitation (RIP) assay. Small interfering RNA (siRNA) mediated knockdown experiments were performed to assess the role of ZNF561-AS1 and SRSF6 in the proliferative actives and apoptosis rate of CRC cells. A mouse xenograft model was employed to assess tumor growth upon ZNF561-AS1 knockdown and SRSF6 rescue. Results We find that ZNF561-AS1 and SRSF6 were upregulated in CRC patient tissues. ZNF561-AS1 expression was reduced in tissues from treated CRC patients but upregulated in CRC tissues from relapsed patients. SRSF6 expression was suppressed and enhanced by ZNF561-AS1 depletion and overexpression, respectively. Mechanistically, ZNF561-AS1 regulated SRSF6 expression by sponging miR-26a-3p and miR-128-5p. ZNF561-AS1-miR-26a-3p/miR-128-5p-SRSF6 axis was required for CRC proliferation and survival. ZNF561-AS1 knockdown suppressed CRC cell proliferation and triggered apoptosis. ZNF561-AS1 depletion suppressed the growth of tumors in a model of a nude mouse xenograft. Similar observations were made upon SRSF6 depletion. SRSF6 overexpression reversed the inhibitory activities of ZNF561-AS1 in vivo, as well as in vitro. Conclusion In summary, we find that ZNF561-AS1 promotes CRC progression via the miR-26a-3p/miR-128-5p-SRSF6 axis. This study reveals new perspectives into the role of ZNF561-AS1 in CRC.


Sign in / Sign up

Export Citation Format

Share Document