scholarly journals Targeting Upstream Kinases of STAT3 in Human Medulloblastoma Cells

2019 ◽  
Vol 19 (7) ◽  
pp. 571-582 ◽  
Author(s):  
Jia Wei ◽  
Ling Ma ◽  
Chenglong Li ◽  
Christopher R. Pierson ◽  
Jonathan L. Finlay ◽  
...  

Background:Medulloblastoma is the most common malignant brain tumor in children. Despite improvement in overall survival rate, it still lacks an effective targeted treatment strategy. The Janus family of cytoplasmic tyrosine kinases (JAKs) and Src kinases, upstream protein kinases of signal transducer and activator of transcription 3 (STAT3), play important roles in medulloblastoma pathogenesis and therefore represent potential therapeutic targets.Methods:In this report, we examined the inhibitory efficacy of the JAK1/2 inhibitor, ruxolitinib, the JAK3 inhibitor, tofacitinib and two Src inhibitors, KX2-391 and dasatinib.Results:These small molecule drugs significantly reduce cell viability and inhibit cell migration and colony formation in human medulloblastoma cells in vitro. Src inhibitors have more potent efficacy than JAK inhibitors in inhibiting medulloblastoma cell migration ability. The Src inhibitors can inhibit both phosphorylation of STAT3 and Src while JAK inhibitors reduce JAK/STAT3 phosphorylation. We also investigated the combined effect of the Src inhibitor, dasatinib with cisplatin. The results show that dasatinib exerts synergistic effects with cisplatin in human medulloblastoma cells through the inhibition of STAT3 and Src.Conclusion:Our results suggest that the small molecule inhibitors of STAT3 upstream kinases, ruxolitinib, tofacitinib, KX2-391, and dasatinib could be novel and attractive candidate drugs for the treatment of human medulloblastoma.

Molecules ◽  
2021 ◽  
Vol 26 (8) ◽  
pp. 2204
Author(s):  
Meng-Die Yang ◽  
Yang Sun ◽  
Wen-Jun Zhou ◽  
Xiao-Zheng Xie ◽  
Qian-Mei Zhou ◽  
...  

Triple-negative breast cancer (TNBC) is a refractory type of breast cancer that does not yet have clinically effective drugs. The aim of this study is to investigate the synergistic effects and mechanisms of resveratrol combined with cisplatin on human breast cancer MDA-MB-231 (MDA231) cell viability, migration, and invasion in vivo and in vitro. In vitro, MTS assays showed that resveratrol combined with cisplatin inhibits cell viability as a concentration-dependent manner, and produced synergistic effects (CI < 1). Transwell assay showed that the combined treatment inhibits TGF-β1-induced cell migration and invasion. Immunofluorescence assays confirmed that resveratrol upregulated E-cadherin expression and downregulated vimentin expression. Western blot assay demonstrated that resveratrol combined with cisplatin significantly reduced the expression of fibronectin, vimentin, P-AKT, P-PI3K, P-JNK, P-ERK, Sma2, and Smad3 induced by TGF-β1 (p < 0.05), and increased the expression of E-cadherin (p < 0.05), respectively. In vivo, resveratrol enhanced tumor growth inhibition and reduced body weight loss and kidney function impairment by cisplatin in MDA231 xenografts, and significantly reduced the expressions of P-AKT, P-PI3K, Smad2, Smad3, P-JNK, P-ERK, and NF-κB in tumor tissues (p < 0.05). These results indicated that resveratrol combined with cisplatin inhibits the viability of breast cancer MDA231 cells synergistically, and inhibits MDA231 cells invasion and migration through Epithelial-mesenchymal transition (EMT) approach, and resveratrol enhanced anti-tumor effect and reduced side of cisplatin in MDA231 xenografts. The mechanism may be involved in the regulations of PI3K/AKT, JNK, ERK and NF-κB expressions.


2011 ◽  
Vol 493-494 ◽  
pp. 836-839
Author(s):  
Masanori Kikuchi

International standard for test method on cell migration into a scaffold is one of the important things to evaluate the scaffold. The "cell migration" ability can divide into two parts. One is infiltration of cell suspension before in vitro cell culture on the scaffold. Another is migration of adherent cells from the edge of scaffold. The latter one could be closely related to cell/tissue migration into the scaffold when it is implanted into bone. Thus, in the present study, the cell migration ability was evaluated toward standardization of in vitro evaluation method for in vivo cell/tissue migration ability using several bioactive ceramics and composites including commercially available materials. The specimen 5 mm in diameter was placed on confluent MG63 cell layer. After 3 days incubation, the specimen was harvested, fixed and divided into two parts. Inside and outside of the scaffold were stained by Giemsa and observed by optical microscopy. In addition, the same specimen was critical point dried and observed with scanning electron microscope (SEM). From microscopic observation, MG63 cells migrated to pore walls of the specimen as well as a sidewall. Maximum migration distances were different among specimens and seemed to depend on pore structure and size as well as porosity. Similar behaviors were observed with SEM.Even relations between this test method and in vivo cell/tissue migration have not been evaluated, this test method is potentially a good method for testing cell migration ability of porous bioactive ceramics as well as other porous scaffold materials.


2020 ◽  
Author(s):  
Gurpreet Arora ◽  
Arun Gupta ◽  
Tong Guo ◽  
Aakash Gandhi ◽  
Aaron Laine ◽  
...  

ABSTRACTBackgroundCachexia (CX), a syndrome of muscle atrophy, adipose loss, and anorexia, is associated with reduced survival in cancer patients. The colon adenocarcinoma C26c20 cell line secretes the cytokine leukemia inhibitor factor (LIF) which induces CX. We characterized how LIF promotes CX-associated weight loss and anorexia in mice through JAK-dependent changes in adipose and hypothalamic tissues.MethodsCX was induced in vivo with C26c20 colon adenocarcinoma cells or recombinant LIF administration in the absence or presence of JAK inhibitors. Blood, adipose, and hypothalamic tissues were collected and processed for cyto/adipokine ELISAs, immunoblot analysis, and quantitative RT-PCR. CX was induced in vitro by stimulating differentiated adipocytes with recombinant LIF or IL-6 in the absence or presence of lipase or JAK inhibitors. These activated adipocytes were processed for lipolysis, immunoblot analysis, and RT-PCR.ResultsTumor-secreted LIF induced changes in adipose tissue expression and serum levels of IL-6 and leptin in a JAK-dependent manner influencing CX-associated adipose wasting and anorexia. We identified two JAK inhibitors that block cytokine-mediated adipocyte lipolysis and IL-6 induction using an in vitro CX lipolysis assay. JAK inhibitors administered to in vivo colon cancer CX mouse models led to 1) a decrease in STAT3 phosphorylation in hypothalamic and adipose tissues, 2) a reverse in the CX serum cyto/adipokine signature, 3) a delay in colon cancer CX-associated anorexia and adipose loss, and 4) an improvement in overall survival.ConclusionsJAK inhibitors suppress cytokine-associated adipose loss and anorexia in multiple in vitro and in vivo models of cancer CX.


2020 ◽  
Author(s):  
Rakesh K. Singh ◽  
KyuKwang Kim ◽  
Rachael B. Rowswell-Turner ◽  
Jeanne N. Hansen ◽  
Negar Khazan ◽  
...  

AbstractVitamin-D receptor (VDR) mRNA is enriched in malignant lung, ovarian and pancreatic tissues and showed poor prognoses. Calcitriol and stable or CRISPR-directed VDR upregulation increased PD-L1mRNA and protein expression in cancer cells in-vitro. A ChIP assay showed the binding of VDR with VDREPD-L1. Stattic, a STAT3 phosphorylation inhibitor blocked calcitriol or VDR overexpression induced PD-L1 upregulation. MeTC7, a VDR antagonist developed by us, reduced PD-L1 expression on macrophages, ovarian, lung, breast, and pancreatic cancer cells in-vitro. In radiotherapy inducible PD-L1 model of orthotopic MC38 murine colon cancer, MeTC7 decreased PD-L1 surface expression, suppressed inflammatory monocytes (IMs) population and increased intra-tumoral CD69+PD1+CD8+T-cells. Intriguingly, MeTC7 reduced TH-MYCN transgenic neuroblastoma tumor growth without affecting PD-L1 and tumor immune milieu. In summary, Vitamin-D/VDR drives PD-L1 expression on cancer cells via STAT-3. Inhibiting VDR exhibited anti-checkpoint effects in orthotopic colon tumors, whereas PDL1-independent and anti-VDR/MYCN effects controlled growth of transgenic neuroblastoma and xenografted tumors.SummaryVitamin-D/VDR induces PD-L1 expression on cancer cells via STAT-3; and targeting VDR by a novel small molecule antagonist MeTC7 exhibits both anti-PD-L1 and anti-VDR/MYCN effects in tumor models.


2020 ◽  
Author(s):  
Haipeng Zhou ◽  
Ying He ◽  
Yinxiang Yang ◽  
Zhaoyan Wang ◽  
Qian Wang ◽  
...  

AbstractNG2 and A2B5 are important biological markers of human oligodendrocyte progenitor cells. To study their functional differences during the development of human oligodendrocyte progenitor cells to oligodendrocytes, we used cell sorting technology and obtained a large number of sterile, high-purity NG2+/- and A2B5+/- cells with high viability. Further research was then conducted via in vitro cell proliferation and migration assays, single-cell sequencing, mRNA sequencing, and cell transplantation into shiverer mice. The results showed that the migration ability of the cells was inversely proportional to the myelination ability. NG2 may be a marker of early oligodendrocyte progenitor cells and is conducive to cell migration and proliferation, while A2B5 may be a marker of slightly mature oligodendrocyte progenitor cells and is conducive to cell differentiation. Further, cell migration, proliferation, and myelination capacity of the negative cell population were stronger than those of the positive cell population. In summary, these results suggest that oligodendrocyte progenitor cells in the mid-stage may be more suitable for clinical cell transplantation to treat demyelinating diseases.Summary statementThis research found that oligodendrocyte progenitor cells in the middle developmental stages may be more suitable for cell transplantation to treat demyelinating diseases.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 1339-1339
Author(s):  
Aurélie Cabannes ◽  
Aline Schmidt ◽  
Eolia Brissot ◽  
Marie Balsat ◽  
Sébastien Maury ◽  
...  

Background. We previously reported that IL7R-pathway genes are mutated in 29% of T-ALL and that surface IL7R is expressed in 53% of T-ALL (Rathana K et al. abstract S131, EHA 2019). In these samples, in vitro and in vivo (IL7R+PDX T-ALL) sensitivity to JAK inhibitors is determined by the expression of IL7R regardless of the IL7Rp genomic status. BCL2 inhibition has been shown to be synergistic with the IL7R pathway inhibition (Degrise S et al., Leukemia 2018;32(3):788-800). We therefore explored whether the combination of tofacitinib, a potent JAK3 inhibitor, with venetoclax may improve the hematological status of patients with relapse/refractory T-ALL. Methods. Patients with relapse/refractory T-ALL, with surface IL7R expression or IL7R-pathway mutations and BCL2 expression were eligible if they had failed all available therapeutic options (including clofarabine and/or allogenic HSCT if eligible). Patients were offered to be treated with venetoclax, 100 mg/d day1; 200 mg/d day2; 300 mg/d day3 and 400 mg/day thereafter and for subsequent 28 days cycles combined with tofacitinib, 10mg twice a day started from day 5 cycle 1 (off-label use). Responses were assessed after cycle 1 and cycle 3. Responding patients may continue therapy until relapse, death or allogenic HSCT if eligible. Results. Eight patients were treated including 7 ETP-ALL and 1 T-cell lymphoblastic lymphoma. Median age was 56 years (27-69) and sex ratio (M/F) was 4/4. Four patients were refractory to at least 2 lines of therapy and 4 patients relapsed, all on therapy. Relapsing patients were in failure after at least one salvage therapy. All patients expressed BCL2. A mutation of JAK3 L857P was found in 5 cases including one patient with both JAK3 and JAK1 mutations. except one with a mutation of IL7-R. Other patients expressed IL7R. A response to therapy was observed in 5 out of seven evaluable cases (71%) including 2 CR associated with a negative MRD (sensitivity 10-4) and 3 partial responses (medullar blasts less than 15%). The remaining patient has just started the first cycle of therapy. Duration of responses was 10 months and +6 months (allo HSCT planned) for the 2 patients in CR and 3 months or less for the partial responders. No serious adverse event related to therapy was observed. Conclusion. IL7-pathway is a possible target for therapy with JAK inhibitors in patients with T-ALL in relapse or refractory to conventional therapy. A combination of venetoclax and tofacitinib may offer a potential savage for these patients with limited therapeutic options. Disclosures Chevallier: Daiichi Sankyo: Honoraria; Incyte: Consultancy, Honoraria; Jazz Pharmaceuticals: Honoraria. Dombret:Institut de Recherches Internationales Servier (IRIS): Research Funding; AGIOS: Honoraria; CELGENE: Consultancy, Honoraria. Boissel:NOVARTIS: Consultancy. Rousselot:Pfizer: Research Funding; Incyte: Research Funding. OffLabel Disclosure: Venetoclax in T-ALL Tofacitinib in T-ALL


2020 ◽  
Vol 22 (Supplement_2) ◽  
pp. ii29-ii29
Author(s):  
Paula Schiapparelli ◽  
Paola Suarez Meade ◽  
Pierre Miranda-Herrera ◽  
Alexandra R Bechtle ◽  
Farren Issacs ◽  
...  

Abstract Glioblastoma (GBM) is the most common and aggressive primary brain tumor in adults. Currently, treatment is focused on achieving maximal safe resection, followed by chemo- and radiotherapy. Although surgery remains the most important therapeutic component, complete surgical resection is virtually impossible, as GBM cells readily infiltrate the adjacent brain parenchyma. One fundamental mechanism for GBM cell migration is the ability of these cells to use cell volume regulation as a driving force of cell infiltration and potentially interfering with apoptosis and cell cycle progression. In this work, we targeted the master regulators of cell volume, the kinases STE20/SPS1-Related Proline-Alanine-Rich Protein Kinase (SPAK) and Oxidative stress-responsive kinase-1) (OSR1) by using a novel proprietary small molecule inhibitor. In order to study the effects of inhibiting SPAK and OSR1, we analyzed cell proliferation and migration. First, we determined the IC50 using six different patient-derived GBM cell lines; we found IC50 values ranging from 0.2–2μM. Next, cell proliferation was determined by cell cycle analysis through Edu labeling. We found a significant decrease in S phase and cell cycle arrest in G2M (p= 0.001, n=6). In addition, cell migration was determined using boyden chamber assay. We found a dose-dependent reduction in cell migration (p=0.001, n=3). These results correlated with the phenomenon observed in an orthotopic murine model of GBM, in which the inhibitor showed a decrease in tumor growth (p=0.0026, n=4) and greater survival rates in vivo (p=0.0046, n=8). When used in combination with radiation therapy, this small molecule inhibitor was capable of radio-sensitizing GBM cells decreasing clonogenicity (p=0.01, n=3) were observed in vitro. In summary, by targeting the SPAK/OSR1 kinases with a small molecule inhibitor, GBM cells become less aggressive, mainly by interfering with cell migration and proliferation and becoming more sensitive to radiation.


2021 ◽  
Vol 11 ◽  
Author(s):  
Hongjian Zhao ◽  
Junjun Huang ◽  
Ming Chen ◽  
Baoru Li ◽  
Xinran Chen ◽  
...  

Colorectal cancer (CRC) is one of the leading causes of cancer death worldwide, with most mortalities being caused by metastases. However, the underlying molecular mechanism of CRC metastases remains largely unknown. Emerging evidence has shown the role of the tripartite motif family, especially tripartite motif protein 6 (TRIM6), in carcinogenesis. In this study, we used CRC cell lines with TRIM6 knockdown and overexpression to investigate the function of TRIM6 in CRC metastasis. We found that TRIM6 promotes CRC cell migration and invasion both in vitro and in vivo. TRIM6 knockdown slows down the migration and invasion processes, whereas TRIM6 overexpression accelerates CRC cell migration and invasion. TRIM6 is potentially the upstream regulatory factor for signal transducer and activator of transcription 3 (STAT3) via the suppressor of cytokine signaling 2 (SOCS2). A total of 70 samples from patients with CRC further confirmed that TRIM6 expression level is positively correlated with STAT3 phosphorylation and negatively correlated with SOCS2 expression. Therefore, TRIM6 could be a potential therapeutic target for CRC metastasis.


2015 ◽  
Vol 8 ◽  
pp. CGM.S32801 ◽  
Author(s):  
Arabinda Das ◽  
Ron Ron Cheng ◽  
Megan L.T. Hilbert ◽  
Yaenette N. Dixon-Moh ◽  
Michele Decandio ◽  
...  

Glioblastoma (GB) is the most common malignant brain tumor. Drug resistance frequently develops in these tumors during chemotherapy. Therefore, predicting drug response in these patients remains a major challenge in the clinic. Thus, to improve the clinical outcome, more effective and tolerable combination treatment strategies are needed. Robust experimental evidence has shown that the main reason for failure of treatments is signal redundancy due to coactivation of several functionally linked receptor tyrosine kinases (RTKs), including anaplastic lymphoma kinase (ALK), c-Met (hepatocyte growth factor receptor), and oncogenic c-ros oncogene1 (ROS1: RTK class orphan) fusion kinase FIG (fused in GB)-ROS1. As such, these could be attractive targets for GB therapy. The study subjects consisted of 19 patients who underwent neurosurgical resection of GB tissues. Our in vitro and ex vivo models promisingly demonstrated that treatments with crizotinib (PF-02341066: dual ALK/c-Met inhibitor) and temozolomide in combination induced synergistic antitumor activity on FIG-ROS1-positive GB cells. Our results also showed that ex vivo FIG-ROS1+ slices (obtained from GB patients) when cultured were able to preserve tissue architecture, cell viability, and global gene-expression profiles for up to 14 days. Both in vitro and ex vivo studies indicated that combination blockade of FIG, p-ROS1, p-ALK, and p-Met augmented apoptosis, which mechanistically involves activation of Bim and inhibition of survivin, p-Akt, and Mcl-1 expression. However, it is important to note that we did not see any significant synergistic effect of crizotinib and temozolomide on FIG-ROS1-negative GB cells. Thus, these ex vivo culture results will have a significant impact on patient selection for clinical trials and in predicting response to crizotinib and temozolomide therapy. Further studies in different animal models of FIG-ROS1-positive GB cells are warranted to determine useful therapies for the management of human GBs.


Sign in / Sign up

Export Citation Format

Share Document