4.7 Article

Neuromyelitis optica (NMO)-IgG-driven organelle reorganization in human iPSC-derived astrocytes

期刊

FASEB JOURNAL
卷 35, 期 10, 页码 -

出版社

WILEY
DOI: 10.1096/fj.202100637R

关键词

astrocytes; autophagy; endoplasmic reticulum; human iPSC; lysosome; metabolic flux; mitochondria; neuromyelitis optica (NMO)

资金

  1. Ministry of Education [2020R1A6A3A01099337]
  2. Ministry of Science and ICT of Korea [20-CoE-BT-01]
  3. NRF [2019R1A2C1010634]
  4. KBRI basic research program [21-BR-01-11]
  5. National Research Foundation of Korea [20-COE-BT-01, 2019R1A2C1010634] Funding Source: Korea Institute of Science & Technology Information (KISTI), National Science & Technology Information Service (NTIS)

向作者/读者索取更多资源

NMO is an autoimmune disease that targets astrocytes through the action of NMO-IgG against AQP4. Research shows that NMO-IgG induces structural alterations in astrocyte mitochondria, leading to impaired mitochondrial functions and dynamics, as well as changes in endoplasmic reticulum and lysosomes. The study using an in vitro human astrocyte model system with iPSCs technology provides insights into how NMO-IgG rearranges cellular organelles and affects their functions, offering opportunities for studying the pathophysiological mechanisms of NMO for potential therapeutic compounds targeting astrocytic abnormalities in patients with NMO.
Neuromyelitis optica (NMO) is an autoimmune disease that primarily targets astrocytes. Autoantibodies (NMO-IgG) against the water channel protein, aquaporin 4 (AQP4), are a serologic marker in NMO patients, and they are known to be responsible for the pathophysiology of the disease. In the brain, AQP4 is mainly expressed in astrocytes, especially at the end-feet, where they form the blood-brain barrier. Following the interaction between NMO-IgG and AQP4 in astrocytes, rapid AQP4 endocytosis initiates pathogenesis. However, the cellular and molecular mechanisms of astrocyte destruction by autoantibodies remain largely elusive. We established an in vitro human astrocyte model system using induced pluripotent stem cells (iPSCs) technology in combination with NMO patient-derived serum and IgG to elucidate the cellular and functional changes caused by NMO-IgG. Herein, we observed that NMO-IgG induces structural alterations in mitochondria and their association with the endoplasmic reticulum (ER) and lysosomes at the ultrastructural level, which potentially leads to impaired mitochondrial functions and dynamics. Indeed, human astrocytes display impaired mitochondrial bioenergetics and autophagy activity in the presence of NMO-IgG. We further demonstrated NMO-IgG-driven ER membrane deformation into a multilamellar structure in human astrocytes. Together, we show that NMO-IgG rearranges cellular organelles and alter their functions and that our in vitro system using human iPSCs offers previously unavailable experimental opportunities to study the pathophysiological mechanisms of NMO in human astrocytes or conduct large-scale screening for potential therapeutic compounds targeting astrocytic abnormalities in patients with NMO.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.7
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据