TET3 promotes AML growth and epigenetically regulates glucose metabolism and leukemic stem cell associated pathways

Leukemia ◽  
2021 ◽  
Author(s):  
Alex Jose Pulikkottil ◽  
Shiva Bamezai ◽  
Tobias Ammer ◽  
Fabian Mohr ◽  
Kristin Feder ◽  
...  
2007 ◽  
Vol 20 (1) ◽  
pp. 13-18 ◽  
Author(s):  
Craig T. Jordan
Keyword(s):  

2021 ◽  
Author(s):  
John F. Woolley ◽  
Keyue Chen ◽  
Gizem E. Genc ◽  
Daniel K.C. Lee ◽  
Irakli Dzneladze ◽  
...  

Despite an increased understanding of leukemogenesis, specific mechanisms that underlie stemness in leukemia remain largely undefined. Here, we report a novel pathway which regulates leukemic differentiation through control of lysosomal biology. We show that disruption of INPP4B results in dysregulated lysosomal gene networks, reduced lysosomal numbers and proteolytic capacity in leukemia. Inpp4b-deficient HSCs and LSCs are functionally compromised. Inpp4b-deficient leukemia models develop more differentiated leukemias with reduced disease initiating potential, and improved overall survival compared to Inpp4b-expressing leukemias. Together, our data is consistent with a model where INPP4B restricts differentiation of LSCs through regulation of lysosomal function. These data provide a mechanism to explain the association of INPP4B with aggressive AML and highlight avenues for LSC-specific leukemia therapies.


Blood ◽  
2015 ◽  
Vol 125 (1) ◽  
pp. 90-101 ◽  
Author(s):  
Ruth Scheicher ◽  
Andrea Hoelbl-Kovacic ◽  
Florian Bellutti ◽  
Anca-Sarmiza Tigan ◽  
Michaela Prchal-Murphy ◽  
...  

Key Points CDK6 acts as a transcriptional regulator to suppress Egr1 in HSCs and LSCs, allowing their activation. Cdk6 −/− HSCs fail to contribute to repopulation in competitive transplants, and BCR-ABLp210+Cdk6−/− LSCs fail to inflict disease.


2021 ◽  
Author(s):  
Xi Zhou ◽  
Junbo Li ◽  
Jin Wang ◽  
Huifang Yang ◽  
Jingzeng Wang ◽  
...  

Abstract Mesenchymal stem cells (MSCs) are widely used to treat and prevent liver ischemia–reperfusion injury (LIRI), which commonly occurs after liver surgery. Lin28 is an RNA-binding protein crucial for early embryonic development, stem cell differentiation/reprogramming, tumorigenesis, and metabolism. However, whether Lin28 can enhance metabolism in human placental MSCs (PMSCs) during hypoxia to improve the protective effect against LIRI remains unclear. First, a Lin28 overexpression construct was introduced into PMSCs; glucose metabolism, the expression of glucose metabolism - and PI3K-AKT pathway-related proteins, and the levels of microRNA Let-7 family members were examined using a glucose metabolism kit, western blots, and real-time quantitative PCR, respectively. Next, treatment with an AKT inhibitor was performed to understand the association of Lin28 with the PI3K-Akt pathway. Subsequently, AML12 cells were co-cultured with PMSCs to construct an in vitro model of PMSC protecting liver cells from hypoxia injury. Finally, C57BL/6J mice were used to establish a partial warm hepatic ischemia–reperfusion model in vivo. Lin28 increased the glycolysis capacity of PMSCs, allowing these cells to produce more energy under hypoxic conditions. Lin28 also activated PI3K-Akt signaling under hypoxic conditions, and AKT inhibition attenuated the effects of Lin28. In addition, Lin28 overexpression was found to protect cells against LIRI-induced liver damage, inflammation, and apoptosis and attenuate hypoxia-induced hepatocyte injury. Inconclusion, Lin28 enhances glucose metabolism under hypoxic conditions in PMSCs, thereby providing protective effects against LIRI via the activation of the PI3K-Akt signaling pathway. Our study first reported the application of gene-modified mesenchymal stem cell-based therapy in LIRI.


Cell Reports ◽  
2019 ◽  
Vol 27 (13) ◽  
pp. 3939-3955.e6 ◽  
Author(s):  
Nora Yucel ◽  
Yu Xin Wang ◽  
Thach Mai ◽  
Ermelinda Porpiglia ◽  
Peder J. Lund ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document