scholarly journals ZIP10 drives osteosarcoma proliferation and chemoresistance through ITGA10-mediated activation of the PI3K/AKT pathway

Author(s):  
Hongyu Li ◽  
Xin Shen ◽  
Mengjun Ma ◽  
Wenzhou Liu ◽  
Wen Yang ◽  
...  

Abstract Background The zinc transporters Zrt- and Irt-related protein (ZIP/SLC39) are overexpressed in human tumors and correlate with poor prognosis; however, their contributions to carcinogenesis and chemoresistance in osteosarcoma (OS) remain unclear. Methods We collected 64 OS patient tissues with (n = 12) or without (n = 52) chemotherapy. The expression levels of ZIP10 were measured by immunohistochemistry and applied to prognostic analysis. ZIP10 was knocked down or overexpressed in OS cell lines to explore its effect on proliferation and chemoresistance. RNA sequencing, quantitative real-time PCR, and western blotting analysis were performed to explore ZIP10-regulated downstream target genes. A xenograft mouse model was established to evaluate the mechanisms by which ZIP10 modulates chemoresistance in OS cells. Results The expression of ZIP10 was significantly induced by chemotherapy and highly associated with the clinical outcomes of OS. Knockdown of ZIP10 suppressed OS cell proliferation and chemoresistance. In addition, ZIP10 promoted Zn content-induced cAMP-response element binding protein (CREB) phosphorylation and activation, which are required for integrin α10 (ITGA10) transcription and ITGA10-mediated PI3K/AKT pathway activation. Importantly, ITGA10 stimulated PI3K/AKT signaling but not the classical FAK or SRC pathway. Moreover, overexpression of ZIP10 promoted ITGA10 expression and conferred chemoresistance. Treatment with the CREB inhibitor 666–15 or the PI3K/AKT inhibitor GSK690693 impaired tumor chemoresistance in ZIP10-overexpressing cells. Finally, a xenograft mouse model established by subcutaneous injection of 143B cells confirmed that ZIP10 mediates chemotherapy resistance in OS cells via the ZIP10-ITGA10-PI3K/AKT axis. Conclusions We demonstrate that ZIP10 drives OS proliferation and chemoresistance through ITGA10-mediated activation of the PI3K/AKT pathway, which might serve as a target for OS treatment.

2017 ◽  
Vol 313 (2) ◽  
pp. E148-E166 ◽  
Author(s):  
Abdul Bari Muhammad ◽  
Bowen Xing ◽  
Chengyang Liu ◽  
Ali Naji ◽  
Xiaosong Ma ◽  
...  

Menin is a scaffold protein that interacts with several epigenetic mediators to regulate gene transcription, and suppresses pancreatic β-cell proliferation. Tamoxifen-inducible deletion of multiple endocrine neoplasia type 1 ( MEN1) gene, which encodes the protein menin, increases β-cell mass in multiple murine models of diabetes and ameliorates diabetes. Glucagon-like-peptide-1 (GLP1) is another key physiological modulator of β-cell mass and glucose homeostasis. However, it is not clearly understood whether menin crosstalks with GLP1 signaling. Here, we show that menin and protein arginine methyltransferase 5 (PRMT5) suppress GLP1 receptor (GLP1R) transcript levels. Notably, a GLP1R agonist induces phosphorylation of forkhead box protein O1 (FOXO1) at S253, and the phosphorylation is mediated by PKA. Interestingly, menin suppresses GLP1-induced and PKA-mediated phosphorylation of both FOXO1 and cAMP response element binding protein (CREB), likely through a protein arginine methyltransferase. Menin-mediated suppression of FOXO1 and CREB phosphorylation increases FOXO1 levels and suppresses CREB target genes, respectively. A small-molecule menin inhibitor reverses menin-mediated suppression of both FOXO1 and CREB phosphorylation. In addition, ex vivo treatment of both mouse and human pancreatic islets with a menin inhibitor increases levels of proliferation marker Ki67. In conclusion, our results suggest that menin and PRMT5 suppress GLP1R transcript levels and PKA-mediated phosphorylation of FOXO1 and CREB, and a menin inhibitor may reverse this suppression to induce β-cell proliferation.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3923-3923
Author(s):  
Fenghuang Zhan ◽  
Fang Xiao ◽  
Maurizio Zangari ◽  
Hongwei Xu ◽  
Guido J. Tricot

Abstract Abstract 3923 Background: Autologous transplantation in combination with the newer drugs has significantly prolonged survival of patients with myeloma (MM). However, the majority of patients still relapses. Our previous study has shown that NEK2, a chromosomal instability (CIN) gene, is up-regulated in many types of cancer cells and is highly related to drug resistance. Recently, premature senescence has been determined to be an indicator of the efficacy of anti-cancer drugs. In this study, we investigate the correlation between the inhibition of premature senescence and NEK2 induced drug resistance. Materials and Methods: Four MM cell lines (Δ47, KMS28PE, ARP1 and U266) and 2 other cancer cell lines (MCF7, H1299) over- and under-expressing NEK2 using a lentiviral delivery system were studied. NEK2 induced drug resistance was evaluated by colonogenic and standard apoptotic assays. The phenotypic changes of premature senescence were determined by using SA β-galactosidase (SA β-gal) staining. The mechanisms of NEK2-induced anti-senescence activity were studied in vitro and in the 5TGM1 murine MM mouse model. Results: Over-expression of NEK2 stimulated cancer cells into increased S and G2/M phase, resulting in polyploidy. All NEK2 over-expressing cancer cells were able to override doxorubicin-induced premature senescence and to maintain their proliferative potential. Mechanistic studies revealed that NEK2 activated the PI3K/Akt pathway. This was confirmed by the use of a specific PI3K/Akt inhibitor, KP372-1, which abrogated NEK2-induced drug resistance by decreasing clonogenicity of NEK2 over-expressing cells and by decreasing tumor burden and extending mouse survival in the 5TGM1 myeloma mouse model even better than bortezomib. In contrast, knockdown NEK2 by shRNA also induced premature senescence. We determined that NEK2 activates PI3K/Akt pathway by directly interacting with pp1. And the Akt down-stream targets GSK-3, MDM2, NF-kB and the tumor suppressor genes p53, p21, p27, the pro-apoptotic and pro-survival genes of the Bcl-2 family as well as the ABC transporters that related to drug resistance were identified to contribute to the inhibition of premature senescence resulting in drug resistance. Conclusion: NEK2 inhibits doxorubicin-induced premature senescence, resulting in increased drug resistance by activating the PI3K/Akt pathway. Alternatively, down regulation of NEK2 by shRNA inhibits cell proliferation and decreases drug resistance. Thus, targeting PI3K/Akt pathway should eliminate NEK2 induced drug resistance by re-establishing drug-induced premature senescence. Disclosures: No relevant conflicts of interest to declare.


2011 ◽  
Vol 209 (1) ◽  
pp. 55-64 ◽  
Author(s):  
Mahsa Hamzeh ◽  
Bernard Robaire

Androgens are the primary regulators of epididymal structure and functions. In the classical view of androgen action, binding of androgen to the intracellular androgen receptor (AR) produces the receptor-steroid complex that has high affinity for DNA response elements and regulates the transcription of target genes. In this study, we demonstrate that in epididymal cells, 5α-dihydrotestosterone (DHT) can cause an alternative and rapid response that is independent of AR–DNA interactions and is mediated by activation of signaling pathways through the AR. We examined changes in AKT and extracellular signal-regulated protein kinases (ERK1/2) activation at early time points after DHT supplementation in the mouse proximal caput epididymis-1 cell line. DHT had no significant effect on AKT activation at any time point. However, DHT activated the ERK pathway as early as at 1 min, the pathway remained activated at 10 min, but activation was not sustained at later time points. Interestingly, ERK activation was blocked by hydroxyflutamide (HF), indicating that early ERK activation was an AR-mediated response. DHT phosphorylates steroid receptor co-activator (SRC) kinase, and this activation was required for the ERK response. EGFR and IGF1R were downstream of SRC, and these two receptors together contributed to enhance ERK and cAMP response element-binding protein (CREB) phosphorylation. We postulate that this rapid action of androgen may ultimately act to modulate the transcription of genes regulated by AR in the nucleus. These results support the hypothesis that DHT can activate a pathway involving the sequential activation of MEK, ERK1/2, and CREB through the EGFR/IGF1R in an epididymal cell line.


2020 ◽  
Vol 21 (23) ◽  
pp. 8994
Author(s):  
Matteo Spinelli ◽  
Francesca Natale ◽  
Marco Rinaudo ◽  
Lucia Leone ◽  
Daniele Mezzogori ◽  
...  

Overnutrition and metabolic disorders impair cognitive functions through molecular mechanisms still poorly understood. In mice fed with a high fat diet (HFD) we analysed the expression of synaptic plasticity-related genes and the activation of cAMP response element-binding protein (CREB)-brain-derived neurotrophic factor (BDNF)-tropomyosin receptor kinase B (TrkB) signalling. We found that a HFD inhibited both CREB phosphorylation and the expression of a set of CREB target genes in the hippocampus. The intranasal administration of neural stem cell (NSC)-derived exosomes (exo-NSC) epigenetically restored the transcription of Bdnf, nNOS, Sirt1, Egr3, and RelA genes by inducing the recruitment of CREB on their regulatory sequences. Finally, exo-NSC administration rescued both BDNF signalling and memory in HFD mice. Collectively, our findings highlight novel mechanisms underlying HFD-related memory impairment and provide evidence of the potential therapeutic effect of exo-NSC against metabolic disease-related cognitive decline.


2002 ◽  
Vol 88 (3) ◽  
pp. 1270-1278 ◽  
Author(s):  
Yun-Fei Lu ◽  
Robert D. Hawkins

We previously found that the nitric oxide (NO)-cGMP-cGMP-dependent protein kinase (PKG) signaling pathway acts in parallel with the cAMP-cAMP-dependent protein kinase (PKA) pathway to produce protein and RNA synthesis-dependent late-phase long-term potentiation (L-LTP) and cAMP response element-binding protein (CREB) phosphorylation in the CA1 region of mouse hippocampus. We have now investigated the possible involvement of a downstream target of PKG, ryanodine receptors. L-LTP can be induced by either multiple-train tetanization, NO or 8-Br-cGMP paired with one-train tetanization, or the cAMP activator forskolin, and all three types of potentiation are accompanied by an increase in phospho-CREB immunofluorescence in the CA1 cell body area. Both the potentiation and the increase in phospho-CREB immunofluorescence induced by multiple-train tetanization or 8-Br-cGMP paired with one-train tetanization are reduced by prolonged perfusion with ryanodine, which blocks Ca2+ release from ryanodine-sensitive Ca2+ stores. By contrast, neither the potentiation nor the increase in immunofluorescence induced by forskolin are reduced by depletion of ryanodine and inositol-1,4,5-triphosphate (IP3)-sensitive Ca2+ stores. These results suggest that NO, cGMP, and PKG cause release of Ca2+ from ryanodine-sensitive stores, which in turn causes phosphorylation of CREB in parallel with PKA during the induction of L-LTP.


Blood ◽  
2002 ◽  
Vol 100 (2) ◽  
pp. 467-473 ◽  
Author(s):  
Arjen-Kars Boer ◽  
A. Lyndsay Drayer ◽  
Hallgeir Rui ◽  
Edo Vellenga

Abstract Erythroid colony formation in response to erythropoietin (EPO) stimulation is enhanced by costimulating the cells with prostaglandin-E2 (PGE2). The present study further analyzed the underlying mechanisms and demonstrated that EPO-mediated STAT5 transactivation in the erythroid AS-E2 cell line was enhanced 6-fold by PGE2 (10 μM), without affecting the STAT5 tyrosine phosphorylation or STAT5-DNA binding. Moreover, the PGE2-enhancing effect was independent of STAT5 serine phosphorylation. In AS-E2 cells STAT5 is constitutively phosphorylated on Ser780 (STAT5A) and EPO-dependently phosphorylated on Ser726/731 (STAT5A/STAT5B), but overexpression of STAT5 serine mutants did not affect STAT5 transactivation. In addition, PGE2 did not affect STAT5 serine phosphorylation. Instead, the stimulatory effect of PGE2 on STAT5 signaling could be mimicked by dibutyryl-cyclic adenosine monophosphate (cAMP) and the phosphodiesterase inhibitor IBMX, suggesting that the effect was mediated by cAMP. Activation of the cAMP pathway resulted in cAMP-response element binding protein (CREB) phosphorylation, which was sustained in the presence of EPO plus PGE2 and transient on EPO stimulation alone. The costimulatory effect of PGE2 on EPO-mediated STAT5 transactivation was inhibited by overexpression of serine-dead CREB or protein kinase A (PKA) inhibitor (PKI), in contrast to EPO-mediated transactivation, which was PKA independent. Furthermore, CREB-binding protein (CBP)/p300 was shown to be involved in EPO-mediated STAT5 transactivation, and a CBP mutant with increased affinity for CREB resulted in an additional enhancement of the PGE2 effect. Finally, we demonstrated that the STAT5 target genes Bcl-X, SOCS2, andSOCS3 were up-regulated by costimulation with PGE2. In summary, these studies demonstrate that PGE2 enhancement of EPO-induced STAT5 transactivation is mediated by the cAMP/PKA/CREB pathway.


2017 ◽  
Vol 8 (5) ◽  
pp. e2830-e2830 ◽  
Author(s):  
Lifen Zhao ◽  
Yujia Shan ◽  
Bing Liu ◽  
Yang Li ◽  
Li Jia

Abstract Chronic myeloid leukemia (CML) is caused by the constitutively active BCR-ABL tyrosine kinase. Although great progress has been made for improvement in clinical treatment during the past decades, it is common for patients to develop chemotherapy resistance. Therefore, further exploring novel therapeutic strategies are still crucial for improving disease outcome. MicroRNAs (miRNAs) represent a novel class of genes that function as negative regulators of gene expression. Recently, miRNAs have been implicated in several cancers. Previously, we identified 41 miRNAs that were dysregulated in resistant compared with adriamycin (ADR)-sensitive parental cells in CML. In the present study, we reported that miR-3142 are overexpressed in ADR-resistant K562/ADR cells and CML/multiple drug resistance patients, as compared with K562 cells and CML patients. Upregulation of miR-3142 in K562 cells accelerated colony formation ability and enhanced resisitance to ADR in vitro. Conversely, inhibition of miR-3142 expression in K562/ADR cells decreased colony-formation ability and enhanced sensitivity to ADR in vitro and in vivo. Significantly, our results showed miR-3142-induced ADR resistance through targeting phosphatase and tensin homologue deleted on chromosome 10 (PTEN), which led to downregulation of PTEN protein and activation of PI3 kinase (PI3K)/Akt pathway. Inhibition of Akt using Akt inhibitor or introduction of PTEN largely abrogated miR-3142-induced resistance. These findings indicated that miR-3142 induces cell proliferation and ADR resistance primarily through targeting the PTEN/PI3K/Akt pathway and implicate the potential application of miR-3142 in cancer therapy.


2021 ◽  
Vol 11 ◽  
Author(s):  
Jiancheng Lv ◽  
Zijian Zhou ◽  
Jingzi Wang ◽  
Xiao Yang ◽  
Hao Yu ◽  
...  

IntroductionCircular RNAs (circRNAs) are non-coding RNAs that have the structure of a covalently closed loop. Increasing data have proven that circRNAs can influence the progression and chemotherapy sensitivity of tumors. Therefore, the underlying function and mechanisms of more circRNAs in progression and chemotherapy resistance are important.MethodsWe conducted RNA sequencing on five pairs of urothelial carcinoma samples and screened for circRNAs. CircFAM114A2 was found to be low expressed in urothelial carcinoma. The functions of circFAM114A2 in urothelial carcinoma were explored by cell cycle assay, IC50 determination assay, cell proliferation assay, apoptosis assay, and tumorigenesis assay.ResultsWe discovered that the levels of circFAM114A2 were decreased in urothelial carcinoma cell lines and tissues. According to follow-up data, urothelial carcinoma patients with higher circFAM114A2 expression had better survival. Importantly, the levels of circFAM114A2 were associated with the histological grade of urothelial carcinoma. CircFAM114A2 could inhibit cell proliferation and block more urothelial carcinoma cells in the G1 phase and then increase the sensitivity of urothelial carcinoma to cisplatin chemotherapy. Mechanistically, circFAM114A2 directly sponged miR-222-3p/miR-146a-5p and subsequently influenced the expressions of the downstream target genes P27/P21, which, in turn, inhibited the progression of urothelial carcinoma and increased the sensitivity of cancer cells to cisplatin chemotherapy.ConclusionCircFAM114A2 could inhibit progression and promote cisplatin sensitivity in urothelial carcinoma through novel circFAM114A2/miR-222-3p/P27 and circFAM114A2/miR-146a-5p/P21 pathways. CircFAM1142 has therefore great potential as a prognostic biomarker and therapeutic target for urothelial carcinoma.


Author(s):  
Lun Gao ◽  
Junhui Liu ◽  
Pengfei Xu ◽  
Gang Deng ◽  
Baohui Liu ◽  
...  

Abstract Background : Glioblastoma multiforme (GBM) is the most common and deadliest type of primary malignant tumor in the adult central nervous system. Temozolomide (TMZ) has limited effectiveness on glioblastoma, so it is urgent to develop new drugs to improve the prognosis of patients. SC66, a novel AKT inhibitor, was reported to exert antiproliferative activity in many types of cancer cells. However, it remains unclear whether SC66 has antitumor effects in GBM. Methods : Cell count kits-8(CCK8) assay, EdU-DNA incorporation assay and colony formation assay were used to evaluate cell proliferation of U87 and U251 cells.Wound -healing assay and transwell assay were used to detect migration and invasion. The cell cycle and apoptosis were detected by flow cytometry. Finally, xenograft mouse model was established to demonstrate the antitumor effect of SC66 in vitro. Results :SC66 obviously suppressed U87 and U251 cells proliferation in a dose-dependent manner. Additionally, SC66 treatment was found to significantly inhibit the invasion and migration of GBM cells as detected by Transwell invasion and wound healing assays. Moreover, treatment of SC66 induced GBM cells apoptosis through up-regulating BAX, Cleaved -caspase3 and down-regulating Bcl-2. SC66 also could arrested cell cycle in G0/G1 phase by decreasing cyclin D1.Furthermore, the results of western blot showed that SC66 significantly reduced level of phosphorylation of AKT, p-GSK-3β and β-catenin, while no change was observed in level of AKT and GSK3β.Then a GSK3-inhibitor,IM-12 was used and IM-12 could significantly restored proliferation, migration and invasion of glioma cells treated with SC66.Meanwhile, SC66 showed significantly suppressed the tumorigenicity compared to the control group in the xenograft mouse model. Conclusion : AKT inhibitor SC66 exerted powerful antitumor activity via down-regulating AKT/β-catenin pathways in vitro and in vivo. It might be as a potential chemotherapy drug to improve the therapeutic efficacy of GBM treatments.


Sign in / Sign up

Export Citation Format

Share Document