4.7 Review

Dysfunction of X-linked inhibitor of apoptosis protein (XIAP) triggers neuropathological processes via altered p53 activity in Huntington's disease

Journal

PROGRESS IN NEUROBIOLOGY
Volume 204, Issue -, Pages -

Publisher

PERGAMON-ELSEVIER SCIENCE LTD
DOI: 10.1016/j.pneurobio.2021.102110

Keywords

XIAP; p53; Mitochondria; Neurodegeneration; Huntington's disease

Categories

Funding

  1. NIH [R01AG054156, R01NS109537]
  2. National Research Foundation (NRF) [2016M3C7A1904233, 2018M3C7A1056894, 2020M3E5D9079744]
  3. National Research Council of Science & Technology (NST) Grant from Korea Ministry of Science, ICT (MSIT) [CRC-15-04-KIST]
  4. Korea Institute of Science and Technology of South Korea [2E30951, 2E30954, 2E30962]
  5. NRF [2020R1H1A2013386]
  6. IITP Grant from MSIT of South Korea [2020-0-01343]
  7. National Research Foundation of Korea [2018M3C7A1056894, 2020R1H1A2013386, 2020M3E5D9079744, 2016M3C7A1904233] Funding Source: Korea Institute of Science & Technology Information (KISTI), National Science & Technology Information Service (NTIS)

Ask authors/readers for more resources

Mitochondrial dysfunction in Huntington's disease is associated with a decrease in XIAP-p53 colocalization and a reduction in XIAP levels due to mutant Huntingtin overexpression, leading to mitochondrial localization of p53. XIAP interacts with p53 C-terminal domain and decreases its stability via autophagy, preventing mitochondrial oxidative stress and cell death. Dysregulation of XIAP triggers mitochondrial dysfunction and neuropathological processes, while XIAP overexpression improves neuropathology in mouse models of HD.
Mitochondrial dysfunction is associated with neuronal damage in Huntington's disease (HD), but the precise mechanism of mitochondria-dependent pathogenesis is not understood yet. Herein, we found that colocalization of XIAP and p53 was prominent in the cytosolic compartments of normal subjects but reduced in HD patients and HD transgenic animal models. Overexpression of mutant Huntingtin (mHTT) reduced XIAP levels and elevated mitochondrial localization of p53 in striatal cells in vitro and in vivo. Interestingly, XIAP interacted directly with the C-terminal domain of p53 and decreased its stability via autophagy. Overexpression of XIAP prevented mitochondrially targeted-p53 (Mito-p53)-induced mitochondrial oxidative stress and striatal cell death, whereas, knockdown of XIAP exacerbated Mito-p53-induced neuronal damage in vitro. In vivo transduction of AAV-shRNA XIAP in the dorsal striatum induced rapid onset of disease and reduced the lifespan of HD transgenic (N171-82Q) mice compared to WT littermate mice. XIAP dysfunction led to ultrastructural changes of the mitochondrial cristae and nucleus morphology in striatal cells. Knockdown of XIAP exacerbated neuropathology and motor dysfunctions in N171-82Q mice. In contrast, XIAP overexpression improved neuropathology and motor behaviors in both AAV-mHTT-transduced mice and N171-82Q mice. Our data provides a molecular and pathological mechanism that deregulation of XIAP triggers mitochondria dysfunction and other neuropathological processes via the neurotoxic effect of p53 in HD. Together, the XIAP-p53 pathway is a novel pathological marker and can be a therapeutic target for improving the symptoms in HD.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available