LncIHAT is induced by hypoxia-inducible factor 1 and promotes breast cancer progression

2020 ◽  
pp. molcanres.0383.2020
Author(s):  
Lin Chen ◽  
Lei Bao ◽  
Yanling Niu ◽  
Jennifer E. Wang ◽  
Ashwani Kumar ◽  
...  
2020 ◽  
Author(s):  
Jianlong Wang ◽  
Xuyi Chen ◽  
Ning Zhang ◽  
Jianzhao Gao ◽  
Bin Liu

Abstract The authors have requested that this preprint be withdrawn due to author disagreement.


2020 ◽  
Author(s):  
Jianlong Wang ◽  
Xuyi Chen ◽  
Ning Zhang ◽  
Jianzhao Gao ◽  
Bin Liu

Abstract Background: Hypoxia induces a series of cellular adaptive responses that enable to promote inflammation and cancer development. However, only few have been fully characterized about the roles of long noncoding RNAs (lncRNAs) in hypoxia-associated cancer progression. Methods: The involvement of lncRNAs in hypoxia-related cancer progression was screened by qRT-PCR. Based on the public databases and integrating bioinformatics analysis, the alteration of prostate cancer associated transcript-1 (PCAT-1) in breast cancer tissues was detected and validated in a cohort of breast cancer tissues. Overexpression and knocking down experiments were performed to uncover the biological roles of PCAT-1 on cell hypoxia-associated phenotypes and biological behaviors. RNA immunoprecipitation (RIP) and RNA pull-down were carried out to reveal the physical interaction between PCAT-1 and receptor of activated protein-C kinase-1 (RACK1). Moreover, xenograft mouse models were used to evaluate the influence of PCAT-1 on cancer progression and metastasis in vivo.Results: We identified PCAT-1 as a hypoxia-inducible lncRNA that regulated the hypoxia-inducible factor-1α (HIF-1α) stability, crucial for cancer progression. Extensive analyses of clinical data indicated that PCAT-1 was elevated in breast cancer patients and was associated with pathological grade, tumor size and poor clinical outcomes. Through gain and loss of function experiments, we found that PCAT-1 promoted hypoxia-associated breast cancer progression including growth, migration, invasion, colony formation, and metabolic regulation. Mechanistically, PCAT-1 directly interacted with RACK1 protein and prevented RACK1 from binding to HIF-1α, thus protecting HIF-1α from RACK1-induced oxygen-independent degradation.Conclusions: These findings provide a new insight into lncRNA-mediated mechanisms for HIF-1α stability and suggest that a novel role of PCAT-1 as a potential therapeutic target for breast cancer.


2014 ◽  
Author(s):  
Mattia Capulli ◽  
Adriano Angelucci ◽  
Anna Teti ◽  
Patrizia Sanita ◽  
Luca Ventura ◽  
...  

2020 ◽  
Author(s):  
Lungwani Muungo

Tumor hypoxia and hypoxia-inducible factor 1 (HIF-1) activationare associated with cancer progression. Here, we demonstrate thatthe transcription factor TAp73 opposes HIF-1 activity through anontranscriptional mechanism, thus affecting tumor angiogenesis.TAp73-deficient mice have an increased incidence of spontaneousand chemically induced tumors that also display enhanced vascularization.Mechanistically, TAp73 interacts with the regulatory subunit(α) of HIF-1 and recruits mouse double minute 2 homolog intothe protein complex, thus promoting HIF-1α polyubiquitination andconsequent proteasomal degradation in an oxygen-independentmanner. In human lung cancer datasets, TAp73 strongly predictsgood patient prognosis, and its expression is associated with lowHIF-1 activation and angiogenesis. Our findings, supported by invivo and clinical evidence, demonstrate a mechanism for oxygenindependentHIF-1 regulation, which has important implicationsfor individualizing therapies in patients with cancer.


Sign in / Sign up

Export Citation Format

Share Document