Abstract

Oxidative stress (OS) represents a state of an imbalanced amount of reactive oxygen species (ROS) and/or a hampered efficacy of the antioxidative defense system. Cells of the central nervous system are particularly sensitive to OS, as they have a massive need of oxygen to maintain proper function. Consequently, OS represents a common pathophysiological hallmark of neurodegenerative diseases and is discussed to contribute to the neurodegeneration observed amongst others in Alzheimer’s disease and Parkinson’s disease. In this context, accumulating evidence suggests that OS is involved in the pathophysiology of Niemann-Pick type C1 disease (NPC1). NPC1, a rare hereditary neurodegenerative disease, belongs to the family of lysosomal storage disorders. A major hallmark of the disease is the accumulation of cholesterol and other glycosphingolipids in lysosomes. Several studies describe OS both in murine in vivo and in vitro NPC1 models. However, studies based on human cells are limited to NPC1 patient-derived fibroblasts. Thus, we analyzed OS in a human neuronal model based on NPC1 patient-specific induced pluripotent stem cells (iPSCs). Higher ROS levels, as determined by DCF (dichlorodihydrofluorescein) fluorescence, indicated oxidative stress in all NPC1-deficient cell lines. This finding was further supported by reduced superoxide dismutase (SOD) activity. The analysis of mRNA and protein levels of SOD1 and SOD2 did not reveal any difference between control cells and NPC1-deficient cells. Interestingly, we observed a striking decrease in catalase mRNA and protein levels in all NPC1-deficient cell lines. As catalase is a key enzyme of the cellular antioxidative defense system, we concluded that the lack of catalase contributes to the elevated ROS levels observed in NPC1-deficient cells. Thus, a restitution of a physiological catalase level may pose an intervention strategy to rescue NPC1-deficient cells from the repercussions of oxidative stress contributing to the neurodegeneration observed in NPC1.

Highlights

  • The disease Niemann-Pick type C1 (NPC1) is a rare neurovisceral lysosomal storage disorder that is caused by autosomal recessive mutations in the Niemann-Pick type C1 disease (NPC1) gene

  • As we have demonstrated that proteins of NPC1-deficient cells undergo nitration, and since SOD2 is a main target for the nitration of tyrosyl residues in the active center of the protein [35,38,39], we determined the ratio of nitrated SOD1 and total amount of SOD1, as well as the nSOD2/SOD2 ratio, in NPC1-deficient cells

  • We demonstrated for the first time that neuronal cells derived from NPC1 patient-specific induced pluripotent stem cells suffer from oxidative stress

Read more

Summary

Introduction

The disease Niemann-Pick type C1 (NPC1) is a rare neurovisceral lysosomal storage disorder that is caused by autosomal recessive mutations in the NPC1 gene. While the transport mechanism of cholesterol to the mitochondria is still not fully understood, an increased amount of cholesterol in the mitochondria of NPC1-deficient cells has been demonstrated [3,4]. This leads to mitochondrial dysfunction and alterations in the electron transport chain [3,5–8], promoting the production of reactive oxygen species (ROS). ROS directly attack biomolecules and induce permanent cellular damage, often leading to apoptosis [10–12] In this context, increased ROS levels were found in a number of NPC1 in vivo and in vitro models, ranging from rats treated with NPC1 inhibitor U18666A, BALB/c mice carrying either different NPC1 mutations or a knockout, a yeast homolog of a NPC1 model and CHO cells to human neuroblastoma cells and NPC1 patient-specific fibroblasts [13–17]

Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call