Proteomics

Dataset Information

0

Inability to switch from ARID1A-BAF to ARID1B-BAF impairs exit from pluripotency and commitment towards neural crest formation in ARID1B-related neurodevelopmental disorders


ABSTRACT: The BAF complex modulates chromatin accessibility. Specific BAF configurations have functional consequences, and subunit switches are essential for cell differentiation. ARID1B and its paralog ARID1A encode for mutually exclusive BAF subunits. De novo ARID1B haploinsufficient mutations cause a neurodevelopmental disorder spectrum, including Coffin-Siris syndrome, which is characterized by neurological and craniofacial features. Here, we reprogrammed ARID1B+/- Coffin-Siris patient-derived skin fibroblasts into iPSCs, and modeled cranial neural crest cell (CNCC) formation. We discovered that ARID1B is active only during the first stage of this process, coinciding with neuroectoderm specification, where it is part of a lineage-specific BAF configuration (ARID1B-BAF), including SMARCA4, and nine additional subunits. ARID1B-BAF acts as a gate-keeper, ensuring exit from pluripotency and lineage commitment, by attenuating NANOG, SOX2 and the thousands of enhancers directly regulated by these two pluripotency factors at the iPSC stage. In iPSCs, these enhancers are maintained active by an ARID1A-containing BAF. At the onset of differentiation, cells transition from ARID1A-BAF to ARID1B-BAF, eliciting attenuation of the NANOG/SOX2 networks, and triggering pluripotency exit. Coffin-Siris patient cells fail to perform the ARID1A/ARID1B switch, and maintain ARID1A-BAF at pluripotency enhancers throughout all stages of CNCC formation. This leads to a persistent and aberrant SOX2 and NANOG activity, which impairs CNCC formation. In fact, despite showing the typical neural crest signature (TFAP2A+, SOX9+), the ARID1B-haploinsufficient CNCCs are also NANOG-positive, in stark contrast with the ARID1B-wt CNCCs, which are NANOG-negative. These findings suggest a connection between ARID1B mutations, neuroectoderm formation, and a pathogenic mechanism for Coffin-Siris syndrome.

INSTRUMENT(S): Q Exactive HF

ORGANISM(S): Homo Sapiens (human)

TISSUE(S): Cell Culture

SUBMITTER: Sarah Offley  

LAB HEAD: Marco Trizzino

PROVIDER: PXD028557 | Pride | 2021-10-12

REPOSITORIES: Pride

altmetric image

Publications

Inability to switch from ARID1A-BAF to ARID1B-BAF impairs exit from pluripotency and commitment towards neural crest formation in ARID1B-related neurodevelopmental disorders.

Pagliaroli Luca L   Porazzi Patrizia P   Curtis Alyxandra T AT   Scopa Chiara C   Mikkers Harald M M HMM   Freund Christian C   Daxinger Lucia L   Deliard Sandra S   Welsh Sarah A SA   Offley Sarah S   Ott Connor A CA   Calabretta Bruno B   Brugmann Samantha A SA   Santen Gijs W E GWE   Trizzino Marco M  

Nature communications 20211109 1


Subunit switches in the BAF chromatin remodeler are essential during development. ARID1B and its paralog ARID1A encode for mutually exclusive BAF subunits. De novo ARID1B haploinsufficient mutations cause neurodevelopmental disorders, including Coffin-Siris syndrome, which is characterized by neurological and craniofacial features. Here, we leveraged ARID1B<sup>+/-</sup> Coffin-Siris patient-derived iPSCs and modeled cranial neural crest cell (CNCC) formation. We discovered that ARID1B is active  ...[more]

Similar Datasets

2021-09-22 | GSE169654 | GEO
2018-10-18 | PXD010670 | Pride
2016-01-11 | E-GEOD-71932 | biostudies-arrayexpress
2016-01-11 | E-GEOD-71933 | biostudies-arrayexpress
2020-01-21 | PXD015756 | Pride
2019-05-06 | PXD013546 | Pride
2020-01-24 | GSE136870 | GEO
2005-09-23 | E-WMIT-5 | biostudies-arrayexpress
2014-12-09 | E-GEOD-60763 | biostudies-arrayexpress
2008-02-28 | E-TABM-409 | biostudies-arrayexpress