Advanced Search

Submit Manuscript

ADVANCE ONLINE PUBLICATION

ORIGINAL ARTICLES

Nuclear RIPK1 promotes chromatin remodeling to mediate inflammatory response

Wanjin Li1,2,* , Bing Shan2 , Chengyu Zou2 , Huibing Wang1 , Meng-Meng Zhang2 , Hong Zhu1 , Masanori Gomi Naito1 , Daichao Xu2 , Vica Jean Manuel1 , Lauren Mifflin1 , Zhaodong Hou2 , John Ravits3 , Junying Yuan1,2,*

1Department of Cell Biology, Harvard Medical School, Boston, MA, USA
2Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
3ALS Translational Research Program, Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
* Correspondence: Wanjin Li(wanjinli@sioc.ac.cn)Junying Yuan(junying_yuan@sioc.ac.cn)

RIPK1 is a master regulator of multiple cell death pathways, including apoptosis and necroptosis, and inflammation. Importantly, activation of RIPK1 has also been shown to promote the transcriptional induction of proinflammatory cytokines in cells undergoing necroptosis, in animal models of amyotrophic lateral sclerosis (ALS) and Alzheimer’s disease (AD), and in human ALS and AD. Rare human genetic carriers of non-cleavable RIPK1 variants (D324V and D324H) exhibit distinct symptoms of recurrent fevers and increased transcription of proinflammatory cytokines. Multiple RIPK1 inhibitors have been advanced into human clinical trials as new therapeutics for human inflammatory and neurodegenerative diseases, such as ALS and AD. However, it is unclear whether and how RIPK1 kinase activity directly mediates inflammation independent of cell death as the nuclear function of RIPK1 has not yet been explored. Here we show that nuclear RIPK1 is physically associated with the BAF complex. Upon RIPK1 activation, the RIPK1/BAF complex is recruited by specific transcription factors to active enhancers and promoters marked by H3K4me1 and H3K27ac. Activated nuclear RIPK1 mediates the phosphorylation of SMARCC2, a key component of the BAF complex, to promote chromatin remodeling and the transcription of specific proinflammatory genes. Increased nuclear RIPK1 activation and RIPK1/BAF-mediated chromatin-remodeling activity were found in cells expressing non-cleavable RIPK1, and increased enrichment of activated RIPK1 on active enhancers and promoters was found in an animal model and human pathological samples of ALS. Our results suggest that RIPK1 kinase serves as a transcriptional coregulator in nucleus that can transmit extracellular stimuli to the BAF complex to modulate chromatin accessibility and directly regulate the transcription of specific genes involved in mediating inflammatory responses.

https://doi.org/10.1038/s41422-022-00673-3

FULL TEXT | PDF

Browse 401