IL-6 induces tumor suppressor protein tyrosine phosphatase receptor type D by inhibiting miR-34a to prevent IL-6 signaling overactivation

2020 ◽  
Vol 473 (1-2) ◽  
pp. 1-13
Author(s):  
Fan Zhang ◽  
Bo Wang ◽  
Tao Qin ◽  
Lu Wang ◽  
Qingqing Zhang ◽  
...  
Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 2914-2914
Author(s):  
Sarmila Majumder ◽  
Tasneem Motiwala ◽  
Kalpana Ghoshal ◽  
Huban Kutay ◽  
Jharna Datta ◽  
...  

Abstract Regulation of protein phosphorylation by concerted action of protein kinases and phosphatases is important for normal physiological processes. Altered function or expression of one or more components of these regulatory molecules leads to many pathological conditions including cancer. We have previously shown that the truncated form of the receptor-type protein tyrosine phosphatase PTPROt predominantly expressed in haematopoietic cells is suppressed in Chronic Lymphocytic Leukemia (B-CLL). A direct correlation of CpG island methylation and reduced expression of the gene was observed in primary CLL samples and in several leukemia cell lines. To assess the functional significance of loss of PTPROt function in leukemia, we selected K562 cells as a model system, as PTPROt expression is silenced in these cells and is reactivated upon treatment with DNA hypomethylating agents. Ectopic expression of the catalytically active PTPROt inhibited growth of K562 cells and their clonogenic survival in soft agar (a characteristic of cancer cells). Further, cells expressing PTPROt exhibited delayed entry into S-phase from G0/G1 phase. Induction of apoptosis increased significantly in K562 cells expressing functional phosphatase upon serum withdrawal or exposure to the apoptogenic agent camptothecin. Tumorigenic potential of K562 cells in athymic nude mice was also significantly reduced upon ectopic expression of PTPROt. Finally, we demonstrate that the Bcr-Abl fusion protein, product of abnormal chromosomal translocation [t(9;22)] in chronic myelogenous leukemia, is a substrate of PTPROt. Tyrosine phosphorylation of this potent kinase was markedly reduced in K562 cells expressing the catalytically active PTPROt. Enhanced dephosphorylation of Bcr-Abl by PTPROt both in vivo and in vitro explains the observed phenotypes of the PTPROt expressing K562 cells. These data taken together delineate the molecular mechanism of tumor suppressor function of PTPROt in leukemic cells characterized by Philadelphia chromosome. (This work was supported by a grant CA101956 from the National Institutes of Health).


2011 ◽  
Vol 131 (3) ◽  
pp. E216-E226 ◽  
Author(s):  
Sally R. Lambert ◽  
Catherine A. Harwood ◽  
Karin J. Purdie ◽  
Abha Gulati ◽  
Rubeta N. Matin ◽  
...  

2010 ◽  
Vol 70 (21) ◽  
pp. 8896-8906 ◽  
Author(s):  
Marco Della Peruta ◽  
Giovanni Martinelli ◽  
Elisabetta Moratti ◽  
Davide Pintani ◽  
Marzia Vezzalini ◽  
...  

Oncogene ◽  
2021 ◽  
Author(s):  
Yanling Lin ◽  
Xiaohan Zhou ◽  
Kaifan Yang ◽  
Yuting Chen ◽  
Lingzhi Wang ◽  
...  

AbstractRadiotherapy is essential to the treatment of nasopharyngeal carcinoma (NPC) and acquired or innate resistance to this therapeutic modality is a major clinical problem. However, the underlying molecular mechanisms in the radiation resistance in NPC are not fully understood. Here, we reanalyzed the microarray data from public databases and identified the protein tyrosine phosphatase receptor type D (PTPRD) as a candidate gene. We found that PTPRD was downregulated in clinical NPC tissues and NPC cell lines with its promoter hypermethylated. Functional assays revealed that PTPRD overexpression sensitized NPC to radiation in vitro and in vivo. Importantly, miR-454-3p directly targets PTPRD to inhibit its expression and biological effect. Interestingly, mechanistic analyses indicate that PTPRD directly dephosphorylates STAT3 to enhance Autophagy-Related 5 (ATG5) transcription, resulting in triggering radiation-induced autophagy. The immunohistochemical staining of 107 NPC revealed that low PTPRD and high p-STAT3 levels predicted poor clinical outcome. Overall, we showed that PTPRD promotes radiosensitivity by triggering radiation-induced autophagy via the dephosphorylation of STAT3, thus providing a potentially useful predictive biomarker for NPC radiosensitivity and drug target for NPC radiosensitization.


2021 ◽  
Vol 5 (2) ◽  
Author(s):  
Ye Qin ◽  
Xinke Xie

Pathologist Virchow has proposed a hypothesis that the origin of tumors comes from chronic inflammation. Clinically, liver tumors can be divided into three types Hepatocellular carcinoma (HCC) is the most common type, which is closely related to various kinds of inflammation. Studies have shown that protein tyrosine phosphatase receptor type O (PTPRO) is a new type of protein tyrosine phosphatase, which is negatively correlated with tumorigenesis. As a new tumor suppressor protein, PTPRO is of great significance for the diagnosis and treatment of HCC in the future. This paper aims to discuss the mechanism of PTPRO in HCC.


2018 ◽  
Vol 126 (1) ◽  
pp. 27-33 ◽  
Author(s):  
Malgorzata Maciukiewicz ◽  
Ilona Gorbovskaya ◽  
Arun K. Tiwari ◽  
Clement C. Zai ◽  
Natalie Freeman ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document