Back to Search Start Over

Hmga2 protein loss alters nuclear envelope and 3D chromatin structure

Authors :
Giuseppina Divisato
Andrea M. Chiariello
Andrea Esposito
Pietro Zoppoli
Federico Zambelli
Maria Antonietta Elia
Graziano Pesole
Danny Incarnato
Fabiana Passaro
Silvia Piscitelli
Salvatore Oliviero
Mario Nicodemi
Silvia Parisi
Tommaso Russo
Molecular Genetics
Divisato, Giuseppina
Chiariello, Andrea M
Esposito, Andrea
Zoppoli, Pietro
Zambelli, Federico
Elia, Maria Antonietta
Pesole, Graziano
Incarnato, Danny
Passaro, Fabiana
Piscitelli, Silvia
Oliviero, Salvatore
Nicodemi, Mario
Parisi, Silvia
Russo, Tommaso
Source :
BMC Biology, 20(1):171. BioMed Central Ltd.
Publication Year :
2022

Abstract

Background The high-mobility group Hmga family of proteins are non-histone chromatin-interacting proteins which have been associated with a number of nuclear functions, including heterochromatin formation, replication, recombination, DNA repair, transcription, and formation of enhanceosomes. Due to its role based on dynamic interaction with chromatin, Hmga2 has a pathogenic role in diverse tumors and has been mainly studied in a cancer context; however, whether Hmga2 has similar physiological functions in normal cells remains less explored. Hmga2 was additionally shown to be required during the exit of embryonic stem cells (ESCs) from the ground state of pluripotency, to allow their transition into epiblast-like cells (EpiLCs), and here, we use that system to gain further understanding of normal Hmga2 function. Results We demonstrated that Hmga2 KO pluripotent stem cells fail to develop into EpiLCs. By using this experimental system, we studied the chromatin changes that take place upon the induction of EpiLCs and we observed that the loss of Hmga2 affects the histone mark H3K27me3, whose levels are higher in Hmga2 KO cells. Accordingly, a sustained expression of polycomb repressive complex 2 (PRC2), responsible for H3K27me3 deposition, was observed in KO cells. However, gene expression differences between differentiating wt vs Hmga2 KO cells did not show any significant enrichments of PRC2 targets. Similarly, endogenous Hmga2 association to chromatin in epiblast stem cells did not show any clear relationships with gene expression modification observed in Hmga2 KO. Hmga2 ChIP-seq confirmed that this protein preferentially binds to the chromatin regions associated with nuclear lamina. Starting from this observation, we demonstrated that nuclear lamina underwent severe alterations when Hmga2 KO or KD cells were induced to exit from the naïve state and this phenomenon is accompanied by a mislocalization of the heterochromatin mark H3K9me3 within the nucleus. As nuclear lamina (NL) is involved in the organization of 3D chromatin structure, we explored the possible effects of Hmga2 loss on this phenomenon. The analysis of Hi-C data in wt and Hmga2 KO cells allowed us to observe that inter-TAD (topologically associated domains) interactions in Hmga2 KO cells are different from those observed in wt cells. These differences clearly show a peculiar compartmentalization of inter-TAD interactions in chromatin regions associated or not to nuclear lamina. Conclusions Overall, our results indicate that Hmga2 interacts with heterochromatic lamin-associated domains, and highlight a role for Hmga2 in the crosstalk between chromatin and nuclear lamina, affecting the establishment of inter-TAD interactions.

Details

Language :
English
ISSN :
17417007
Database :
OpenAIRE
Journal :
BMC Biology, 20(1):171. BioMed Central Ltd.
Accession number :
edsair.doi.dedup.....c298ccc5ff2be73b34d7f0662548e7c7