4.6 Article

PINK1 Inhibits Multimeric Aggregation and Signaling of MAVS and MAVS-Dependent Lung Pathology

出版社

AMER THORACIC SOC
DOI: 10.1165/rcmb.2020-0490OC

关键词

PTEN-induced kinase 1; mitochondrial antiviral signaling protein; MAVS aggregation; mitochondrial signaling; inflammasomes

资金

  1. Basic Science Research Program through the National Research Foundation of Korea [NRF-2013R1A6A3A03026988]
  2. Veterans Affairs [I01-BX004661]
  3. Department of Defense [PR181442]
  4. National Heart, Lung, and Blood Institute [1R01HL130283]
  5. National Institute on Aging [1R01AG053495]

向作者/读者索取更多资源

The study identified a novel role of PINK1 in regulating MAVS signaling, where PINK1 interacts with and stabilizes MAVS, inhibiting its multimeric aggregation. PINK1 deficiency enhances MAVS-mediated antiviral innate immune and NLRP3 inflammasome signaling, affecting pulmonary pathobiology in response to viral infection and fibrotic injury.
Mitochondria have emerged as important signaling organelles where intracellular perturbations are integrated and, consequently, intracellular signaling pathways are modulated to execute appropriate cellular functions. MAVS (mitochondrial antiviral signaling protein) represents such an example that functions as a platform molecule to mediate mitochondrial innate immune signaling. Recently, multimeric aggregation of MAVS has been identified as a key molecular process for its signaling. The underlying mechanisms to regulate this, however, are still incompletely understood. We hypothesized that PINK1 (PTEN-induced kinase 1) plays an important role in the regulation of multimeric MAVS aggregation and its consequent pathobiology. To test whether PINK1 interacts with MAVS, bimolecular fluorescence complementation analysis and IP were performed. RLH (RIG-I-like helicase) and NLRP3 inflammasome signaling were evaluated by in vitro assay. In vivo functional significance of PINK1 in the regulation of MAVS signaling was evaluated from both murine modeling of influenza viral infection and bleomycin-induced experimental pulmonary fibrosis, wherein MAVS plays important roles. Multimeric MAVS aggregation was induced by mitochondria dysfunction, and, during this event, the stabilized PINK1 interacted physically with MAVS and antagonized multimeric MAVS aggregation. Accordingly, the MAVS-mediated antiviral innate immune and NLRP3 inflammasome signaling were enhanced in PINK1 deficiency. In addition, in vivo studies revealed that MAVS-mediated pulmonary antiviral innate immune responses and fibrotic responses after bleomycin injury were enhanced in PINK1 deficiency. In conclusion, these results establish a new role of PINK1 in the regulation of MAVS signaling and the consequent pulmonary pathobiology.

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

4.6
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