Abstract

Ceruloplasmin (Cp) is a ferroxidase that also plays a role in iron efflux from cells. It can thus help to regulate cellular iron homeostasis. In the CNS, Cp is expressed as a membrane-anchored form by astrocytes. Here, we assessed the role of Cp in permanent middle cerebral artery occlusion (pMCAO) comparing wildtype and Cp null mice. Our studies show that the lesion size is larger and functional recovery impaired in Cp null mice compared to wildtype mice. Expression of Cp increased ninefold at 72 h after pMCAO and remained elevated about twofold at day 14. We also assessed changes in mRNA and protein expression of molecules involved in iron homeostasis. As expected there was a reduction in ferroportin in Cp null mice at 72 h. There was also a remarkable increase in DMT1 protein in both genotypes at 72 h, being much higher in wildtype mice (19.5-fold), that then remained elevated about twofold at 14 days. No difference was seen in transferrin receptor 1 (TfR1) expression, except a small reduction in wildtype mice at 72 h, suggesting that the increase in DMT1 may underlie iron uptake independent of TfR1-endosomal uptake. There was also an increase of ferritin light chain in both genotypes. Iron histochemistry showed increased iron accumulation after pMCAO, initially along the lesion border and later throughout the lesion. Immunofluorescence labeling for ferritin (a surrogate marker for iron) and GFAP or CD11b showed increased ferritin in GFAP+ astrocytes along the lesion border in Cp null mice, while CD11b+ macrophages expressed ferritin equally in both genotypes. Increased lipid peroxidation assessed by 4HNE staining was increased threefold in Cp null mice at 72 h after pMCAO; and 3-nitrotyrosine labeling showed a similar trend. Three key pro-inflammatory cytokines (IL-1β, TNFα, and IL-6) were markedly increased at 24 h after pMCAO equally in both genotypes, and remained elevated at lower levels later, indicating that the lack of Cp does not alter key inflammatory cytokine expression after pMCAO. These data indicate that Cp expression is rapidly upregulated after pMCAO, and loss of Cp results in dysregulation of iron homeostasis, increased oxidative damage, greater lesion size and impaired recovery of function.

Highlights

  • Dysregulation of cellular iron homeostasis is considered to contribute to neural damage after ischemic stroke (MillerotSerrurot et al, 2008; Texel et al, 2011; Garcia-Yebenes et al, 2012)

  • We examined the role of Cp on lesion size and functional recovery, as well as changes in the expression of various molecules involved in iron homeostasis in the ischemic brain

  • The latter may be due to neural plasticity in wildtype mice, which appears to be impaired in Cp null mice, possibly due to free radical mediated secondary damage to neurons and glia

Read more

Summary

Introduction

Dysregulation of cellular iron homeostasis is considered to contribute to neural damage after ischemic stroke (MillerotSerrurot et al, 2008; Texel et al, 2011; Garcia-Yebenes et al, 2012). Iron plays an important role in many physiological processes by catalyzing a variety of enzymatic reactions (Rouault and Tong, 2005) The reason for these various functions is the redox active nature of iron, which can transition between the ferrous (Fe2+) and ferric (Fe3+) states. We and others have reported that Cp gene knockout mice show disruption of iron homeostasis in the CNS and other tissues such as the liver (Harris et al, 1999; Patel et al, 2002) and lead to iron accumulation in the CNS with age (>18 months of age), resulting in free-radical mediated neuronal loss (Patel et al, 2002; Jeong and David, 2006). We have reported a slow accumulation of iron in the brain in Cp null mice with age (>18 months of age) (Patel et al, 2002; Jeong and David, 2006)

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