Cell-specific activation of RIPK1 and MLKL after intracerebral hemorrhage in mice

Author:

Lule Sevda1,Wu Limin1,Sarro-Schwartz Aliyah2,Edmiston William J1,Izzy Saef2,Songtachalert Tanya1,Ahn So Hee1,Fernandes Neil D1ORCID,Jin Gina1,Chung Joon Yong1,Balachandran Siddharth3,Lo Eng H45,Kaplan David6,Degterev Alexei7,Whalen Michael J1

Affiliation:

1. Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA

2. Department of Neurology, Brigham and Woman’s Hospital, Harvard Medical School, Boston, MA, USA

3. Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA, USA

4. Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA

5. Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA

6. Department of Biomedical Engineering, Tufts University, Medford, MA, USA

7. Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA, USA

Abstract

Receptor-interacting protein kinase-1 (RIPK1) is a master regulator of cell death and inflammation, and mediates programmed necrosis (necroptosis) via mixed-lineage kinase like (MLKL) protein. Prior studies in experimental intracerebral hemorrhage (ICH) implicated RIPK1 in the pathogenesis of neuronal death and cognitive outcome, but the relevant cell types involved and potential role of necroptosis remain unexplored. In mice subjected to autologous blood ICH, early RIPK1 activation was observed in neurons, endothelium and pericytes, but not in astrocytes. MLKL activation was detected in astrocytes and neurons but not endothelium or pericytes. Compared with WT controls, RIPK1 kinase-dead ( RIPK1D138N/D138N) mice had reduced brain edema (24 h) and blood-brain barrier (BBB) permeability (24 h, 30 d), and improved postinjury rotarod performance. Mice deficient in MLKL ( Mlkl-/-) had reduced neuronal death (24 h) and BBB permeability at 24 h but not 30d, and improved post-injury rotarod performance vs. WT. The data support a central role for RIPK1 in the pathogenesis of ICH, including cell death, edema, BBB permeability, and motor deficits. These effects may be mediated in part through the activation of MLKL-dependent necroptosis in neurons. The data support development of RIPK1 kinase inhibitors as therapeutic agents for human ICH.

Publisher

SAGE Publications

Subject

Cardiology and Cardiovascular Medicine,Neurology (clinical),Neurology

同舟云学术

1.学者识别学者识别

2.学术分析学术分析

3.人才评估人才评估

"同舟云学术"是以全球学者为主线,采集、加工和组织学术论文而形成的新型学术文献查询和分析系统,可以对全球学者进行文献检索和人才价值评估。用户可以通过关注某些学科领域的顶尖人物而持续追踪该领域的学科进展和研究前沿。经过近期的数据扩容,当前同舟云学术共收录了国内外主流学术期刊6万余种,收集的期刊论文及会议论文总量共计约1.5亿篇,并以每天添加12000余篇中外论文的速度递增。我们也可以为用户提供个性化、定制化的学者数据。欢迎来电咨询!咨询电话:010-8811{复制后删除}0370

www.globalauthorid.com

TOP

Copyright © 2019-2024 北京同舟云网络信息技术有限公司
京公网安备11010802033243号  京ICP备18003416号-3