scholarly journals IM-8 Significance of IL-1 pathways in Glioblastoma

2021 ◽  
Vol 3 (Supplement_6) ◽  
pp. vi8-vi8
Author(s):  
Keitaro Kai ◽  
Yoshihiro Komohara ◽  
Takahiro Yamamoto ◽  
Ken Uekawa ◽  
Tatsuya Takezaki ◽  
...  

Abstract Purpose: Previous studies have revealed that macrophages affect the prognosis of glioblastoma. However, there are still many unknown parts about the mechanism. In this study, we conducted an experiment with the aim of elucidating the mechanism by which tumor associated macrophages (TAM) work on tumors in the tumor microenvironment (TME). Method: Experiments were carried out using two glioblastoma cell strains, T98G, and U251. For clinical data, we analyzed it based on databases such as Protein Atlas, Ivy Glioblastoma Atlas, brain TIME database. Results: In 3D culture, we confirmed that IL-1β stimulation promoted glioblastoma cell proliferation and sphere formation. The addition of IL-1β increased mRNA expression of various cytokines such as IL-6 and CXCL8, and increased phosphorylation of STAT3 in arrays. When we administered IL-6 and CXCL8, the growth was significantly increased in cells administered with IL-6 and CXCL8. As a result, we speculated that STAT3 pathway and NFκB pathway via IL-6 and CXCL8 are involved in cell proliferation by IL-1β. In order to confirm these things, western blot was performed, and it was confirmed that phosphorylation of STAT3 and NFκB were increased. In addition, STAT3 inhibitors and NFκB inhibitors suppressed tumor growth. Clinically analysis was carried out based on the database, and it was found that IL-1β and macrophages were related. Furthermore, IL-1β was found in many cases around tumor necrosis. Discussion: This study clarifies some of the effects of IL-1β on glioblastoma. However, there are still many unknown points, and it is necessary to continue to consider them in the future.

2018 ◽  
Vol 47 (5) ◽  
pp. 1871-1882 ◽  
Author(s):  
Wei-Qiang Jia ◽  
Zhao-Tao Wang ◽  
Ming-Ming Zou ◽  
Jian-Hao Lin ◽  
Ye-Hai Li ◽  
...  

Background/Aims: As a natural antioxidant, verbascoside (VB) is proved to be a promising method for the treatment of oxidative-stress-related neurodegenerative diseases. Thus, this study aimed to investigate the effects of VB on glioblastoma cell proliferation, apoptosis, migration, and invasion as well as the mechanism involving signal transducer and activator of transcription 3 (STAT3) and Src homology 2 domain-containing protein tyrosine phosphatase 1 (SHP-1). Methods: U87 cells were assigned to different treatments. The MTT assay was used to test cell proliferation, flow cytometry was used to detect cell apoptosis, and a Transwell assay was used for cell migration and invasion. We analyzed the glioblastoma tumor growth in a xenograft mouse model. Western blot analysis was employed to determine the protein expression of related genes. Results: Glioblastoma cells exhibited decreased cell proliferation, migration, invasion, and increased apoptosis when treated with VB or TMZ. Western blot analysis revealed elevated SHP-1 expression and reduced phosphorylated (p)-STAT3 expression in glioblastoma cells treated with VB compared with controls. Correspondingly, in a xenograft mouse model treated with VB, glioblastoma tumor volume and growth were decreased. Glioblastoma xenograft tumors treated with VB showed elevated SHP-1, Bax, cleaved caspase-3, and cleaved PARP expression and reduced p-STAT3, Bcl-2, survivin, MMP-2, and MMP-9 expression. siRNA-SHP-1 inhibited the VB effects on glioblastoma. Conclusion: This study demonstrates that VB inhibits glioblastoma cell proliferation, migration, and invasion while promoting apoptosis via SHP-1 activation and inhibition of STAT3 phosphorylation.


2014 ◽  
Vol 289 (25) ◽  
pp. 17689-17698 ◽  
Author(s):  
Jacqueline Sayyah ◽  
Alena Bartakova ◽  
Nekeisha Nogal ◽  
Lawrence A. Quilliam ◽  
Dwayne G. Stupack ◽  
...  

2021 ◽  
Vol 21 (1) ◽  
Author(s):  
Li Wei ◽  
Ya Peng ◽  
Naiyuan Shao ◽  
Peng Zhou

Abstract Background Glioblastoma remains one of the most lethal brain cancers. T-cell immunoglobulin and mucin domain 1 (Tim-1) is associated with various immune diseases. The molecular mechanism of Tim-1 in regulating glioblastoma cell proliferation, invasion, and migration is still unknown. Moreover, it has shown that miR-133a plays an important role in glioblastoma. However, little is known about the interaction between Tim-1 and miR-133a in glioblastoma. Methods Tim-1 expression in glioblastoma and normal brain tissues was detected by qPCR, Western Blot and IHC. After Tim-1 knockdown in U251 and U87 cells, genes showing significantly differential expression, along with the significant differential miRNAs were analyzed using RNA-seq analysis. The binding sites were verified using dual-luciferase reporter gene assay. U251 and U87 cells were allocated into the small harpin-negative control (sh-NC), sh-Tim-1, sh-Tim-1 + inhibitor NC, and sh-Tim-1 + miR-133a inhibitor group. Cell proliferation, migration, and invasion were determined by CCK-8, flow cytometry, wound-healing and Transwell assays, respectively. Next, U251 and U87 cells were allocated into the mimic NC, miR-133a mimic, miR-133a mimic + pcDNA3.1, and miR-133a mimic + pcDNA3.1-TGFBR1 groups, followed by the detection of cell proliferation, migration, and invasion. Western blot was used to identify the expression of vital kinases in the Wnt/β-catenin pathway. Results Tim-1 was highly expressed in glioblastoma tissues compared with that in normal brain tissues. RNA-seq analysis showed that Tim-1 knockdown could lead to the downregulation of TGFBR1 and the upregulation of miR-133a. The binding sites between TGFBR1 and miR-133a were confirmed. Tim-1 knockdown impaired the invasion, migration, proliferation of U251 and U87 cells, which could be reversed by miR-133a downregulation. miR-133a upregulation inhibited the proliferation, invasion, and migration of U251 and U87 cells, which could be reversed by TGFBR1 upregulation. Tim-1 knockdown and miR-133a upregulation could inhibit the activation of the Wnt/β-catenin pathway, while the elevation of TGFBR1 showed opposite effects. Conclusion Tim-1 knockdown inhibited glioblastoma cell proliferation, invasion, and migration through the miR-133a/TGFBR1 axis and restrained the activation of the Wnt/β-catenin pathway.


2020 ◽  
Vol 4 (Supplement_1) ◽  
Author(s):  
Lauren Hillers-Ziemer ◽  
Rachel McMahon ◽  
Margaret Hieptas ◽  
Gretchen Paderta ◽  
Jessica McCready ◽  
...  

Abstract Obesity is a growing health concern worldwide and increases the incidence of multiple types of cancer, including breast cancer. Obese breast cancer patients often develop more aggressive tumors than lean patients and have increased risk for metastasis, tumor recurrence and mortality. Here, we sought to address how obesity alters the biology of breast cancer to promote aggressive tumors. To induce obesity, we fed mice either a control diet (CD) or high fat diet (HFD) for 16 weeks, then transplanted Met-1 tumor cells into mammary fat pads and monitored tumor growth. At end stage, tumors from HFD-fed mice were significantly larger than tumors from CD-fed mice, suggesting obesity promotes tumor growth. To investigate how obesity promotes tumor aggression, we dissociated the tumors from CD- and HFD-fed mice and plated isolated tumor cells in tumorsphere and invasion assays to test for cells with cancer stem-like cell (CSC) properties. Tumor cells from HFD-fed mice demonstrated increased tumorsphere formation and increased capacity for invasion compared to tumor cells from CD-fed mice, suggesting that obesity selects for tumor cells with CSC properties. Next, to address how obesity impacts the tumor microenvironment, we evaluated tumor necrosis and blood vessel formation through CD31 staining. Tumors from HFD-fed mice had significantly less necrosis and greater CD31 staining than those from CD-fed mice, suggesting that obesity promotes tumor angiogenesis. Since obesity promotes chronic, macrophage-driven inflammation within adipose tissue of the mammary gland, we stained tumors for the macrophage marker, F4/80. As with obese mammary glands, tumors from HFD-fed mice had significantly greater macrophage recruitment than tumors from CD-fed mice, together suggesting that obesity alters the tumor microenvironment. To determine how obesity stimulates tumor angiogenesis, we performed an in vitro assay by culturing dissociated tumor cells from HFD or CD-fed mice alone or with macrophages. Conditioned media (CM) isolated from tumor cells from HFD-fed mice cultured with macrophages enhanced the ability of endothelial cells to form networks in vitro. In contrast, CM from HFD tumor cells alone, macrophages alone, or those from CD-fed mice did not promote network formation. Together, these results suggest that cooperation between macrophages and tumor cells from HFD-fed mice promotes angiogenesis. Next, to investigate how macrophages and tumor cells interacting in obesity, we depleted macrophages using anti-F4/80 antibodies in CD-fed and HFD-fed tumor-bearing mice. In HFD-fed mice, macrophage depletion significantly reduced tumor volume and CD31 staining while increasing tumor necrosis compared to controls. Obesity promotes interactions between tumor cells and macrophages to enhance tumor angiogenesis and progression.


2020 ◽  
Vol 20 (1) ◽  
Author(s):  
Mingjun Li ◽  
Qianqian Wang ◽  
Xiaofei Zhang ◽  
Ningning Yan ◽  
Xingya Li

Abstract Background Exosomes, emerging mediators of intercellular communication, are reported to transfer certain non-coding RNAs, such as microRNAs (miRNAs), which play a crucial role in cancer progression. The objective of this study was to determine the function of exosomal miR-126 and provide a novel mechanism of miR-126 action in NSCLC. Methods The morphology of exosomes was identified by transmission electron microscope (TEM), and the exosomal surface markers were quantified by western blot. The expression of miR-126 and integrin alpha-6 (ITGA6) mRNA was measured by quantitative real-time polymerase chain reaction (qRT-PCR), and ITGA6 protein expression was determined by western blot. For functional analyses, cell proliferation was assessed by colony formation assay and MTT assay. Cell cycle and cell apoptosis were monitored using flow cytometry assay. Cell migration and invasion were determined by transwell assay. ITGA6 was predicted as a target of miR-126 by bioinformatics analysis, which was verified by dual-luciferase reporter assay. The role of exosomal miR-126 in vivo was determined by Xenograft tumor models. Results NSCLC serum-derived exosomes harbored low expression of miR-126 and promoted NSCLC cell proliferation, cell cycle progression, cell migration and invasion. NSCLC serum-derived exosomes loaded with miR-126 mimic inhibits NSCLC cell proliferation, colony formation, migration and invasion but induced cell cycle arrest and apoptosis. Besides, exosomal miR-126 also blocked tumor growth in vivo. In mechanism, ITGA6 was a target of miR-126, and exosomal miR-126 weakened these NSCLC cell malignant behaviors and inhibited tumor growth by degrading the expression of ITGA6. Conclusion Exosomal miR-126 blocked the progression of NSCLC through the mediation of its target gene ITGA6, and exosomal miR-126 might be used as a promising biomarker for NSCLC therapy.


2020 ◽  
Vol 2020 ◽  
pp. 1-10 ◽  
Author(s):  
Ping Tian ◽  
Ling Tao ◽  
Yujun Wang ◽  
Xiaobing Han

Objective. Melanoma is the most common form of skin cancer with low survival rate and poor prognosis. MicroRNAs (miRNAs) have been reported to play essential roles in progression of melanoma. However, the role and mechanism of miR-127 in the process of melanoma remain poorly understood. Methods. The expressions of miR-127 and delta-like homologue 1 (DLK1) were measured in melanoma tissues and cells by quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot. Cell proliferation and apoptosis were measured by MTT assay, flow cytometry, and Western blot. The interaction between miR-127 and DLK1 was investigated by bioinformatics analysis, luciferase activity assay, and RNA immunoprecipitation (RIP). Murine xenograft model was conducted to investigate the effect of miR-127 on tumor growth in vivo. Results. miR-127 was inhibited and DLK1 mRNA was enhanced in melanoma tissues and cells. Low abundance of miR-127 in melanoma tissues predicted a poor prognosis and was associated with the malignant clinicopathological features. Overexpression of miR-127 inhibited cell proliferation and induced apoptosis in melanoma cells. Moreover, DLK1 was targeted by miR-127 and its restoration reversed the regulatory effect of miR-127 on the process of melanoma. Besides, the addition of miR-127 suppressed xenograft tumor growth via suppressing DLK1 protein level in nude mice. Conclusion. miR-127 blocked the development of melanoma by targeting DLK1, providing a novel biomarker for the treatment of melanoma.


2020 ◽  
pp. 1-10
Author(s):  
Dongdi Wu ◽  
Jia Zhu ◽  
Ying Fu ◽  
Chenqin Li ◽  
Biao Wu

Breast cancer is the most common malignancies worldwide. LncRNA HOX transcript antisense intergenic RNA (HOTAIR) has been shown to promote progression and metastasis of various cancers, including breast cancer. This reasearch aimed to investigate the downstream regulatory pathways of HOTAIR in breast cancer. The levels of HOTAIR and miR-129-5p were examined in breast cancer tissues and SKBR3 and MCF7 cells by quantitative real-time PCR (qRT-PCR). Cell proliferation was examined by Cell Counting Kit-8 (CCK-8) assay. Cell migration and invasion were estimated by transwell assay. Epithelial-to-mesenchymal transition (EMT)-related markers (E-cadherin, N-cadherin and Vimentin) were measured by Western blot assay. The expression of Frizzled 7 (FZD7) was detected using qRT-PCR or Western blot assay. Bioinformatics analysis, luciferase reporter assay or RNA Immunoprecipitation (RIP) assay was performed to explore the molecular mechanism of HOTAIR in breast cancer. Xenograft analysis was utilized to evaluate the tumor growth in vivo. HOTAIR and FZD7 were upregulated, while miR-129-5p was down-regulated in breast cancer tissues and cells. Knockdown of miR-129-5p reversed the effect of HOTAIR knockdown on cell proliferation, migration, invasion and EMT. FZD7 restored the inhibition of miR-129-5p on breast cancer progression. Furthermore, HOTAIR was a sponge of miR-129-5p and FZD7 was a target of miR-129-5p. Knockdown of HOTAIR inhibited the tumor growth in vivo. HOTAIR facilitated breast cancer progression by regulating the miR-129-5p/FZD7 axis, indicating that HOTAIR may be a potential biomarker and therapeutic target for breast cancer.


2021 ◽  
Vol 19 (1) ◽  
Author(s):  
Linghui Qin ◽  
Xiaosong Sun ◽  
Fei Zhou ◽  
Cheng Liu

Abstract Background Circular RNA low-density lipoprotein receptor-related protein 6 (circLRP6) is considered as an oncogene in many types of cancers. However, the function and mechanisms of circLRP6 in prostate cancer (PCa) tumorigenesis remain largely undefined. Methods Quantitative real-time polymerase chain reaction (qRT-PCR) and western blot assays were conducted to assess the expression of circLRP6, microRNA (miR)-330-5p, and nuclear receptor binding protein 1 (NRBP1). Cell counting kit-8 (CCK-8), colony formation, 5-ethynyl-2’-deoxyuridine (EDU) incorporation, flow cytometry, transwell, wound healing, and western blot assays were performed to detect cell proliferation, apoptosis, and metastasis in vitro. Subcutaneous tumor growth was observed in nude mice to investigate the role of circLRP6 in vivo. The targeting relationship between miR-330-5p and NRBP1 or circLRP6 was verified using dual-luciferase reporter, pull-down, and RNA immunoprecipitation (RIP) assays. Immunohistochemistry was employed to test relative protein expression. Results CircLRP6 was highly expressed in PCa tissues and cells, knockdown of circLRP6 impaired PCa cell growth and metastasis in vitro by affecting cell proliferation, apoptosis, invasion, migration, and epithelial-mesenchymal transition (EMT). Mechanistic studies showed that circLRP6 could competitively bind with miR-330-5p to prevent the degradation of its target gene NRBP1. Rescue assay suggested that miR-330-5p inhibition reversed the inhibitory effects of circLRP6 knockdown on PCa cell growth and metastasis. Moreover, overexpression of miR-330-5p suppressed PCa progression via NRBP1. Notably, tumor formation assay indicated that circLRP6 silencing impeded tumor growth and EMT in vivo. Conclusion Our findings demonstrated that circLRP6 promoted PCa tumorigenesis and metastasis through miR-330-5p/NRBP1 axis, suggesting a potential therapeutic target for PCa.


Sign in / Sign up

Export Citation Format

Share Document