Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 13(1): 7414, 2022 12 02.
Article in English | MEDLINE | ID: mdl-36460681

ABSTRACT

Pluripotent stem cells hold great promise in regenerative medicine and developmental biology studies. Mitochondrial metabolites, including tricarboxylic acid (TCA) cycle intermediates, have been reported to play critical roles in pluripotency. Here we show that TCA cycle enzymes including Pdha1, Pcb, Aco2, Cs, Idh3a, Ogdh, Sdha and Mdh2 are translocated to the nucleus during somatic cell reprogramming, primed-to-naive transition and totipotency acquisition. The nuclear-localized TCA cycle enzymes Pdha1, Pcb, Aco2, Cs, Idh3a promote somatic cell reprogramming and primed-to-naive transition. In addition, nuclear-localized TCA cycle enzymes, particularly nuclear-targeted Pdha1, facilitate the 2-cell program in pluripotent stem cells. Mechanistically, nuclear Pdha1 increases the acetyl-CoA and metabolite pool in the nucleus, leading to chromatin remodeling at pluripotency genes by enhancing histone H3 acetylation. Our results reveal an important role of mitochondrial TCA cycle enzymes in the epigenetic regulation of pluripotency that constitutes a mitochondria-to-nucleus retrograde signaling mode in different states of pluripotent acquisition.


Subject(s)
Epigenesis, Genetic , Histones , Acetylation , Cell Nucleus , Mitochondria
2.
STAR Protoc ; 2(2): 100568, 2021 06 18.
Article in English | MEDLINE | ID: mdl-34151295

ABSTRACT

Somatic cells can be reprogrammed into induced pluripotent stem cells (iPSCs) by defined factors. Here, we describe a protocol for imaging mitochondrial permeability transition pore (mPTP) opening in reprogramming of somatic cells using a confocal microscope. We also describe a method to sort high and low mPTP opening somatic cells by calcein fluorescence and reprogram these sorted cells to iPSCs. These protocols are also suitable for imaging mPTP opening and uncovering the mechanisms of mPTP function in other cell fate conversions. For complete details on the use and execution of this protocol, please refer to Ying et al. (2018).


Subject(s)
Cellular Reprogramming , Mitochondrial Permeability Transition Pore/metabolism , Animals , Cells, Cultured , Culture Media , Mice , Mice, Transgenic
4.
Nat Metab ; 2(9): 882-892, 2020 09.
Article in English | MEDLINE | ID: mdl-32839595

ABSTRACT

Somatic cell reprogramming provides insight into basic principles of cell fate determination, which remain poorly understood. Here we show that the transcription factor Glis1 induces multi-level epigenetic and metabolic remodelling in stem cells that facilitates the induction of pluripotency. We find that Glis1 enables reprogramming of senescent cells into pluripotent cells and improves genome stability. During early phases of reprogramming, Glis1 directly binds to and opens chromatin at glycolytic genes, whereas it closes chromatin at somatic genes to upregulate glycolysis. Subsequently, higher glycolytic flux enhances cellular acetyl-CoA and lactate levels, thereby enhancing acetylation (H3K27Ac) and lactylation (H3K18la) at so-called 'second-wave' and pluripotency gene loci, opening them up to facilitate cellular reprogramming. Our work highlights Glis1 as a powerful reprogramming factor, and reveals an epigenome-metabolome-epigenome signalling cascade that involves the glycolysis-driven coordination of histone acetylation and lactylation in the context of cell fate determination.


Subject(s)
DNA-Binding Proteins/metabolism , Epigenome , Induced Pluripotent Stem Cells , Metabolome , Signal Transduction/genetics , Signal Transduction/physiology , Transcription Factors/metabolism , Acetyl Coenzyme A/metabolism , Animals , Cellular Reprogramming , Cellular Senescence , Chromatin Immunoprecipitation , Glucose/metabolism , Lactic Acid/metabolism , Male , Mice , Plasmids/genetics
5.
Autophagy ; 13(9): 1543-1555, 2017 Sep 02.
Article in English | MEDLINE | ID: mdl-28722510

ABSTRACT

Induced pluripotent stem cells (iPSCs) have fewer and immature mitochondria than somatic cells and mainly rely on glycolysis for energy source. During somatic cell reprogramming, somatic mitochondria and other organelles get remodeled. However, events of organelle remodeling and interaction during somatic cell reprogramming have not been extensively explored. We show that both SKP/SKO (Sox2, Klf4, Pou5f1/Oct4) and SKPM/SKOM (SKP/SKO plus Myc/c-Myc) reprogramming lead to decreased mitochondrial mass but with different kinetics and by divergent pathways. Rapid, MYC/c-MYC-induced cell proliferation may function as the main driver of mitochondrial decrease in SKPM/SKOM reprogramming. In SKP/SKO reprogramming, however, mitochondrial mass initially increases and subsequently decreases via mitophagy. This mitophagy is dependent on the mitochondrial outer membrane receptor BNIP3L/NIX but not on mitochondrial membrane potential (ΔΨm) dissipation, and this SKP/SKO-induced mitophagy functions in an important role during the reprogramming process. Furthermore, endosome-related RAB5 is involved in mitophagosome formation in SKP/SKO reprogramming. These results reveal a novel role of mitophagy in reprogramming that entails the interaction between mitochondria, macroautophagy/autophagy and endosomes.


Subject(s)
Cellular Reprogramming , Membrane Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Mitophagy , Animals , Embryo, Mammalian/cytology , Endosomes/metabolism , Endosomes/ultrastructure , Fibroblasts/metabolism , Kruppel-Like Factor 4 , Membrane Potential, Mitochondrial , Mice , Mitochondria/ultrastructure , Models, Biological , Transcription Factors/metabolism , rab5 GTP-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL