scholarly journals AF10 (MLLT10) prevents somatic cell reprogramming through regulation of DOT1L-mediated H3K79 methylation

2021 ◽  
Vol 14 (1) ◽  
Author(s):  
Deniz Uğurlu-Çimen ◽  
Deniz Odluyurt ◽  
Kenan Sevinç ◽  
Nazlı Ezgi Özkan-Küçük ◽  
Burcu Özçimen ◽  
...  

Abstract Background The histone H3 lysine 79 (H3K79) methyltransferase DOT1L is a key chromatin-based barrier to somatic cell reprogramming. However, the mechanisms by which DOT1L safeguards cell identity and somatic-specific transcriptional programs remain unknown. Results We employed a proteomic approach using proximity-based labeling to identify DOT1L-interacting proteins and investigated their effects on reprogramming. Among DOT1L interactors, suppression of AF10 (MLLT10) via RNA interference or CRISPR/Cas9, significantly increases reprogramming efficiency. In somatic cells and induced pluripotent stem cells (iPSCs) higher order H3K79 methylation is dependent on AF10 expression. In AF10 knock-out cells, re-expression wild-type AF10, but not a DOT1L binding-impaired mutant, rescues overall H3K79 methylation and reduces reprogramming efficiency. Transcriptomic analyses during reprogramming show that AF10 suppression results in downregulation of fibroblast-specific genes and accelerates the activation of pluripotency-associated genes. Conclusions Our findings establish AF10 as a novel barrier to reprogramming by regulating H3K79 methylation and thereby sheds light on the mechanism by which cell identity is maintained in somatic cells.

2020 ◽  
Author(s):  
Deniz Uğurlu-Çimen ◽  
Deniz Odluyurt ◽  
Kenan Sevinç ◽  
Nazlı Ezgi Özkan-Küçük ◽  
Burcu Özçimen ◽  
...  

SummaryThe histone H3 lysine 79 (H3K79) methyltransferase DOT1L is a key chromatin-based barrier to somatic cell reprogramming. However, the mechanisms by which DOT1L safeguards cell identity and somatic-specific transcriptional programs remain unknown. Here, we employed a proteomic approach using proximity-based labeling to identify DOT1L-interacting proteins and investigated their effects on reprogramming. Among DOT1L interactors, suppression of AF10 (MLLT10) via RNA interference or CRISPR/Cas9, significantly increases reprogramming efficiency. In somatic cells and induced pluripotent stem cells (iPSCs) H3K79 di-methylation is dependent on AF10 expression. In AF10 knockout cells, re-expression wildtype AF10, but not a mutant defective in DOT1L binding, rescues overall H3K79 methylation and reduces reprogramming efficiency. Transcriptomic analyses during reprogramming show that AF10 suppression results in downregulation of fibroblast-specific genes and accelerates the activation of pluripotency-associated genes. Our findings establish AF10 as a novel barrier to reprogramming by regulating H3K79 methylation and thereby sheds light on the mechanism by which cell identity is maintained in somatic cells.


2021 ◽  
Author(s):  
Sherif Mahrous Khodeer ◽  
Arne Klungland ◽  
John Arne Dahl

Establishment of the pluripotency regulatory network in somatic cells by introducing four transcriptional factors, (Octamer binding transcription factor 4 (OCT4), SRY (sex determining region Y)-box 2 (SOX2), Kruppel - like factor 4 (KLF4), and cellular-Myelocytomatosis (c-MYC) provides a promising tool for cell-based therapies in regenerative medicine. Still, the mechanisms at play when generating induced pluripotent stem cells from somatic cells is only partly understood. Here we show that the RNA specific N6-methyladenosine (m6A) demethylase ALKBH5 regulates somatic cell reprogramming in a stage specific manner. Knockdown or knockout of Alkbh5 in the early reprogramming phase impairs the reprogramming efficiency by reducing the proliferation rate through arresting the cells at G2/M phase and decreasing the mesenchymal-to-epithelial transition (MET) rate. However, there is no significant change in reprogramming efficiency when Alkbh5 is depleted at the late phase of reprogramming. On the other hand, ALKBH5 overexpression at the earlyreprogramming phase has no significant impact on reprogramming efficiency, while overexpression at the late phase enhances the reprogramming by stabilizing Nanog transcripts resulting in upregulated Nanog expression. Our study provides mechanistic insight into the crucial dynamic role of ALKBH5 in regulating somatic cell reprogramming at the posttranscriptional level.


2012 ◽  
Vol 2012 ◽  
pp. 1-4 ◽  
Author(s):  
Chih-Hao Kuo ◽  
Shao-Yao Ying

The use of somatic cells to generate induced-pluripotent stem cells (iPSCs), which have gene characteristic resembling those of human embryonic stem cells (hESCs), has opened up a new avenue to produce patient-specific stem cells for regenerative medicine. MicroRNAs (miRNAs) have gained much attention over the past few years due to their pivotal role in many biological activites, including metabolism, host immunity, and cancer. Soon after the discovery of embryonic-stem-cell- (ESC-) specific miRNAs, researchers began to investigate their functions in embryonic development and differentiation, as well as their potential roles in somatic cell reprogramming (SCR). Several approaches for ESC-specific miRNA-mediated reprogramming have been developed using cancer and somatic cells to generate ESC-like cells with similarity to iPSCs and/or hESCs. However, the use of virus-integration to introduce reprogramming factors limits future clinical applications. This paper discusses the possible underlying mechanism for miRNA-mediated somatic cell reprogramming and the approaches used by different groups to induce iPSCs with miRNAs.


Animals ◽  
2020 ◽  
Vol 10 (10) ◽  
pp. 1848
Author(s):  
Mirae Kim ◽  
Seon-Ung Hwang ◽  
Junchul David Yoon ◽  
Yeon Woo Jeong ◽  
Eunhye Kim ◽  
...  

Canine induced pluripotent stem cells (ciPSCs) can provide great potential for regenerative veterinary medicine. Several reports have described the generation of canine somatic cell-derived iPSCs; however, none have described the canine somatic cell reprogramming using a non-integrating and self-replicating RNA transfection method. The purpose of this study was to investigate the optimal strategy using this approach and characterize the transition stage of ciPSCs. In this study, fibroblasts obtained from a 13-year-old dog were reprogrammed using a non-integrating Venezuelan equine encephalitis (VEE) RNA virus replicon, which has four reprogramming factors (collectively referred to as T7-VEE-OKS-iG and comprised of hOct4, hKlf4, hSox2, and hGlis1) and co-transfected with the T7-VEE-OKS-iG RNA and B18R mRNA for 4 h. One day after the final transfection, the cells were selected with puromycin (0.5 µg/mL) until day 10. After about 25 days, putative ciPSC colonies were identified showing TRA-1-60 expression and alkaline phosphatase activity. To determine the optimal culture conditions, the basic fibroblast growth factor in the culture medium was replaced with a modified medium supplemented with murine leukemia inhibitory factor (mLIF) and two kinase inhibitors (2i), PD0325901(MEK1/2 inhibitor) and CHIR99021 (GSK3β inhibitor). The derived colonies showed resemblance to naïve iPSCs in their morphology (dome-shaped) and are dependent on mLIF and 2i condition to maintain an undifferentiated phenotype. The expression of endogenous pluripotency markers such as Oct4, Nanog, and Rex1 transcripts were confirmed, suggesting that induced ciPSCs were in the late intermediate stage of reprogramming. In conclusion, the non-integrating and self-replicating VEE RNA replicon system can potentially make a great contribution to the generation of clinically applicable ciPSCs, and the findings of this study suggest a new method to utilize the VEE RNA approach for canine somatic cell reprogramming.


Cell Research ◽  
2011 ◽  
Vol 22 (1) ◽  
pp. 168-177 ◽  
Author(s):  
Athanasia D Panopoulos ◽  
Oscar Yanes ◽  
Sergio Ruiz ◽  
Yasuyuki S Kida ◽  
Dinh Diep ◽  
...  

2017 ◽  
Vol 25 ◽  
pp. 88-97 ◽  
Author(s):  
Ling Wang ◽  
Delun Huang ◽  
Chang Huang ◽  
Yexuan Yin ◽  
Kaneha Vali ◽  
...  

Zygote ◽  
2011 ◽  
Vol 20 (3) ◽  
pp. 277-280
Author(s):  
J. Cardona-Costa ◽  
M. Pérez-Camps ◽  
F. García-Ximénez

SummarySomatic cells derived from zebrafish haploid larval (both androgenetic and gynogenetic) cultures were used as donors for nuclear transplant into non-enucleated oocytes. Nuclei were transplanted either before or simultaneously with oocyte activation in the central region and in the incipient animal pole, respectively. Against expected results, 20% of transplanted embryos during oocyte activation using cells of gynogenetic origin reached the 100% epiboly stage, even two survived for up to 5 days, whereas no development was observed when cells from androgenetic origin were used. Results derived from this work open a novel possibility of studying somatic cell reprogramming and imprinting phenomena in zebrafish.


2021 ◽  
Author(s):  
Xu Jinhong ◽  
Fang Shi ◽  
Wang Naweng ◽  
Li Bo ◽  
Huang Yongheng ◽  
...  

Abstract Background: Human induced pluripotent stem cells (hiPSCs) hold great potential in disease modeling, drug screening and cell therapy. However, efficiency and costs of hiPSCs preparation still need to be improved.Methods: We screened the compounds that target signaling pathways, epigenetic modifications or metabolic-process regulation to replace the growth factors. After small molecules treatment, TRA-1-60 staining was performed to quantify the efficiency of somatic cell reprogramming. Next, small molecule cocktail induced ESCs or iPSCs were examined with pluripotent markers expression. Finally, Genome-wide gene expression profile was then analyzed by RNA-seq to illustrate the mechanism of human somatic cell reprogramming. Result: Here, we found that a dual-specificity tyrosine phosphorylation-regulated kinase inhibitor ID-8 robustly enhanced human somatic cell reprogramming by upregulation of PDK4 and activation of glycolysis. Furthermore, we identified a novel growth-factor-free hiPSC generation system using small molecules ID-8/Kartogenin (IK). Finally, we developed IK medium combined with Low-dose bFGF to support the long-term expansion of human pluripotent stem cells. IK-iPSCs showed pluripotency and normal karyotype. Conclusions: Our studies may provide a novel growth-factor-free culture system to facilitate the generation of hiPSCs for multiple application in regenerative medicine.


Sign in / Sign up

Export Citation Format

Share Document