You are on page 1of 116

저작자표시-비영리-변경금지 2.

0 대한민국

이용자는 아래의 조건을 따르는 경우에 한하여 자유롭게

l 이 저작물을 복제, 배포, 전송, 전시, 공연 및 방송할 수 있습니다.

다음과 같은 조건을 따라야 합니다:

저작자표시. 귀하는 원저작자를 표시하여야 합니다.

비영리. 귀하는 이 저작물을 영리 목적으로 이용할 수 없습니다.

변경금지. 귀하는 이 저작물을 개작, 변형 또는 가공할 수 없습니다.

l 귀하는, 이 저작물의 재이용이나 배포의 경우, 이 저작물에 적용된 이용허락조건


을 명확하게 나타내어야 합니다.
l 저작권자로부터 별도의 허가를 받으면 이러한 조건들은 적용되지 않습니다.

저작권법에 따른 이용자의 권리는 위의 내용에 의하여 영향을 받지 않습니다.

이것은 이용허락규약(Legal Code)을 이해하기 쉽게 요약한 것입니다.

Disclaimer
의학박사 학위논문

Bank Vole 프리온 단백 발현 형질 전환 마우스의

신경세포에서 다양한 프리온 감염 세포주 확립

Establishment of advanced in vitro model from Bank Vole

transgenic mice for susceptibility to various prion strains

Choi, Hong Seok (최 홍 석)

의학과 (Department of Medical Science)

미생물학 전공 (Major in microbiology)

한림대학교 대학원
Graduate School, Hallym University

2016년도
김용선 교수지도

의학 박사 학위논문

최홍석의 박사 학위논문을 합격으로 판정함.

2016 년 12 월

심사위원장 권 형 주 ________________

심사위원 최 경 찬 ________________

심사위원 박 재 봉 ________________

심사위원 김 재 일 ________________

심사위원 김 용 선 ________________
CONTENTS

I . Chapter 1

Dysfunction of Mitochondrial Dynamics in the Brains of Scrapie-Infected Mice.....1

1.1 Intoduction..................................................................................................... 3

1.2 Materials and methods..........................................................................................5

1.3 Result....................................................................................................................8

1.4 Discussion...........................................................................................................11

1.5 References..........................................................................................................19

II . Chapter 2

Upregulation of Connexin 43 Expression Via C-Jun N-Terminal Kinase Signaling in

Prion Disease..............................................................................................................23

2.1 Intoduction..................................................................................................... 25

2.2 Materials and methods........................................................................................27

2.3 Result..................................................................................................................34

2.4 Discussion...........................................................................................................39

2.5 References..........................................................................................................53
III . Chapter 3

Establishment of Advanced In Vitro Model from Bank Vole Transgenic Mice for

Susceptibility to Various Prion Strains..................................................................62

3.1 Intoduction.....................................................................................................63

3.2 Materials and methods........................................................................................66

3.3 Result..................................................................................................................70

3.4 Discussion...........................................................................................................73

3.5 References..........................................................................................................81

3.6 Abstract...............................................................................................................85

3.7 Abstract in korea.................................................................................................86

IV. Appendix...................................................................................................................88
LIST OF FIGURES

Figure 1.1. Deposition of PK-resistant prion isoforms in ME7-infected mice.................15

Figure 1.2. Differential expression of mitochondrial fusion and fission proteins in

hippocampi of ME7-infected mice...................................................................................16

Figure 1.3. Subcellular localization of mitochondrial fusion and fission proteins in

hippocampi of ME7-infected mice...................................................................................17

Figure 1.4. Electron microscopic analysis of the mitochondria in hippocampal neurons

of ME7-infected mice……………………….................….............................................18

Figure 2.1. Increased expression of Cx43 protein and mRNA in the brains of scrapie

infected mice....................................................................................................................43

Figure 2.2. Immunohistochemical staining of Cx43 in brain sections from control and

scrapie-infected mice.......................................................................................................45

Figure 2.3. Increased Cx43 protein in membrane fractions of scrapie infected

brains………………………………………………………………………………….46

Figure 2.4. Subcellular distribution of Cx43 and PrP in hippocampal cell lines with or

without scrapie infection..................................................................................................47

Figure 2.5. JNK activation-mediated Cx43 upregulation in scrapie-infected cells and

mic………………………………………………………………………………….......48

Figure 2.6. Increased EtBr uptake in scrapie-infected cells.............................................49

Figure 2.7. Increased scrape-loading dye transfer in scrapie-infected cells.....................50

Figure 2.8. Attenuation of JNK signaling-mediated Cx43 upregulation by inhibiting the

accumulation of PrPSc......................................................................................................52

Figure 3.1. Cell-type characterization and expressions of the PrP in the BV neuronal cell
lines………………………………………......................................................................76

Figure 3.2. Analysis of PK and TL-resistant PrPSc in the BV neuronal cell lines infected

with different prion strains.............................................................................................78

Figure 3.3. Accumulation of PK-resistant prion isoforms in ZW-22L and BV-22L

scrapie-infected ICR mice………………………………………...............................79

LIST OF TABLES

Table 1. Accumulation of PK-resistant prion isoforms in ZW-22L and BV-22L scrapie-

infected ICR mice………………………………………………………………………79


I. Chapter 1

Dysfunction of mitochondrial dynamics in the brains of scrapie-

infected mice

Abstract

Mitochondrial dysfunction is a common and prominent feature of many

neurodegenerative diseases, including prion diseases; it is induced by oxidative stress in

scrapie-infected animal models. In previous studies, we found swelling and dysfunction

of mitochondria in the brains of scrapie-infected mice compared to brains of controls, but

the mechanisms underlying mitochondrial dysfunction remain unclear. To examine

whether the dysregulation of mitochondrial proteins is related to the mitochondrial

dysfunction associated with prion disease, we investigated the expression patterns of

mitochondrial fusion and fission proteins in the brains of ME7 prion-infected mice.

Immunoblot analysis revealed that Mfn1 was up-regulated in both whole brain and

specific brain regions, including the cerebral cortex and hippocampus, of ME7-infected

mice compared to controls. Additionally, expression levels of Fis1 and Mfn2 were

elevated in the hippocampus and the striatum, respectively, of the ME7-infected brain. In

contrast, Dlp1 expression was significantly reduced in the hippocampus in the ME7-

infected brain, particularly in the cytosolic fraction. Finally, we observed abnormal

mitochondrial enlargement and histopathological change in the hippocampus of the ME7-

infected brain. These observations suggest that the mitochondrial dysfunction, which is

1
presumably caused by the dysregulation of mitochondrial fusion and fission proteins, may

contribute to the neuropathological changes associated with prion disease.

Key words : Prion disease, Mitochondrial fusion, Fission, Scrapie, Mitochondrial

dysfunction

2
1.1 Introduction

Prion diseases, also known as transmissible spongiform encephalopathies (TSEs), are

fatal neurodegenerative disorders that affect both humans and animals [1, 2]. Scrapie is a

prototypical prion disease that affects sheep and goats. Clinically, scrapie is characterized

by a long latent period, progressive ataxia, tremor, wasting and ultimately death [3]. Many

scrapie strains have been isolated from sheep and goats and used to examine not only the

range of various pathogenesis pathways but also the neuropathological mechanisms

induced by prion disease [4]. Typical features of the disease include the formation of

spongiform vacuoles and astrocytosis, the formation of amyloid plaques in some cases

and neuronal loss in the brain [5, 6]. A key event in prion disease is the conformational

misfolding of the endogenously expressed cellular prion protein (PrPC) into the scrapie

form of the pathogenic prion protein (PrPSc) [5].

A number of recent studies have demonstrated that mitochondria are dynamic organelles

that continually undergo fission and fusion with one another [7–9]. Mitochondria can

change in number and morphology within a cell during development, throughout the cell

cycle and when challenged with various cytotoxic stimuli [9]. In mammals, the key

molecules involved in mitochondrial fission are dynamin-like protein 1 (Dlp1, also

referred to as Drp1) and fission 1 (Fis1). The process opposing fission, i.e., mitochondrial

fusion, is controlled in mammalian cells by mitofusin 1 (Mfn1), mitofusin 2 (Mfn2) and

optic atrophy 1 (Opa1) [7]. The sizes, shapes and interconnectivities of mitochondria are

determined by their fusion and fission [9]. It has been suggested that the mitochondrial

defects associated with Alzheimer’s disease (AD), Parkinson’s disease (PD) and

Huntington’s disease (HD) may result, at least in part, from a disruption of the fusion and

3
fission mechanisms of mitochondria [8, 9]. A recent study reported that the expression of

Dlp1 is decreased and that mitochondria are abnormally elongated in the fibroblasts of

AD patients and in neuronal cell lines overexpressing amyloid precursor protein (APP)

[10]. The study suggests that APP causes an imbalance between mitochondrial fusion and

fission that results in an abnormal distribution of mitochondria, which in turn contributes

to mitochondrial and neuronal dysfunction. Several observations suggest a link between

mitochondrial dysfunction and PD. Pink1 and parkin, which are PD-related genes,

promote mitochondrial fission and inhibit mitochondrial fusion in Drosophila [11].

Additionally, HD research has focused on mitochondrial dysfunction. A mouse model of

HD exhibits early defects in respiration and ATP production [12]. Moreover, mutant

huntingtin seems to disrupt mitochondrial Ca2+ buffering [13] and to cause mitochondrial

ultrastructural changes in the lymphoblasts of HD patients [14]. Previous studies reported

that dysfunction and enlargement of the mitochondria occur by oxidative stress in animal

models of prion disease [15, 16]. However, the underlying mechanism responsible for

this mitochondrial dysfunction remains unclear.

In the present study, we investigated the mitochondrial fusion and fission proteins that

may be involved in the mitochondrial dysfunction observed in prion disease. We show

that mitochondrial fusion and fission proteins are differentially modulated in the terminal

stage of an experimental mouse model of prion disease and that this modulation may

contribute to the morphological damage and reduction in number of mitochondria in

infected neuronal cells.

4
1.2 Materials and methods

1) Antibodies

The following monoclonal and polyclonal antibodies were used mouse monoclonal

anti-PrP (3F10) [17], mouse monoclonal anti-COX IV (Abcam), goat polyclonal anti-

enolase (Santa Cruz), mouse monoclonal to β-actin (Sigma–Aldrich), mouse

monoclonal anti-Opa1 and mouse monoclonal anti-Dlp1 (BD Transduction

Laboratories), mouse monoclonal anti-Mfn2, chicken polyclonal anti-Mfn1 (Novus

Biologicals) and rabbit polyclonal anti-Fis1 (Santa Cruz).

2) Animals and scrapie strains

Six-week-old C57BL/6 mice were obtained from the Central Laboratory Animal

(Republic of Korea) and divided into two groups: one group was infected with the ME7

scrapie strain, and the other included age-matched controls. The ME7 scrapie strain was

kindly provided by Dr. Alan Dickinson (Neuropathogenesis Unit, Edinburgh, UK). This

scrapie strain was maintained by serial intracerebral passages of brain homogenate from

a terminally affected mouse. The mice were intracerebrally inoculated with 30 μl of 1%

(w/v) brain homogenate in 0.01 M phosphate-buffered saline (PBS, pH 7.4) from either

a normal brain or an ME7-infected C57BL/6 mouse brain at the terminal stage of the

disease. When the clinical signs of prion disease were evident in the terminal stage (160

days post inoculation, dpi), the mice were sacrificed.

3) Western blot analysis

Whole brains or hippocampal regions were homogenized gently in 10-fold greater

5
volumes (w/v) of 50 mM Tris–HCl (pH 7.4) containing 150 mM NaCl, 1 mM EDTA,

0.25% Na-deoxycholate, 1% NP-40 and protease inhibitor cocktails (Roche). The

protein concentration was determined using the BCA assay (Thermo Scientific). The

homogenates were treated with Proteinase K (PK) at a concentration of 50 μg/ml. Equal

amounts of protein (10–50 lg in all assays) were separated by SDS–PAGE using 10%,

12% or 15% acrylamide gels and then transferred to nitrocellulose membranes (Thermo

Scientific). After blocking with 5% skim milk for 1 h, the membranes were incubated

with the individual antibodies overnight at 4 °C and then incubated with horseradish

peroxidase-conjugated secondary antibody in Tris-buffered saline with 0.05% Tween-20

(TBST) containing 5% skim milk for 1 h at room temperature. The blots were visualized

with the SuperSignal West Pico Chemiluminescent Substrate (Thermo Scientific). The

expression levels of each protein were quantified using ImageJ software (NIH).

4) Transmission electron microscopy (TEM)

The animals were perfused with 0.1 M PBS (pH 7.4) containing 4% paraformaldehyde

and 2.5% glutaraldehyde under deep anesthesia with 16.5% urethane. The brains were

removed and fixed in the 0.1 M PBS fixative that was used for perfusion. The bilateral

hippocampal regions were trimmed into small pieces immediately after their surgical

removal and kept in the fixative for 2 h at 4 °C. Post-fixation was performed in 0.1 M

PBS with 1% osmium tetroxide followed by dehydration through a graded ethanol series

and embedding in Epon 812. Ultra-thin sections (75 nm) prepared by using an

ultramicrotome (RMC MTXL) were stained with uranyl acetate and lead citrate and were

subsequently observed with a transmission electron microscope (JEM-1011, JEOL). The

numbers of total and damaged mitochondria were counted in the hippocampal neurons

6
in the control and ME7-infected brains and then calculated based on fifteen arbitrarily

selected hippocampal neurons. The sizes of the mitochondria were measured under high

magnification (x50,000) using TEM (iTEM, Olympus Soft Imaging Solutions, GmbH),

and the values were calculated as the means ± SDs of the lengths and widths of thirty

arbitrarily selected mitochondria from each group. Statistical analyses of the numbers

and sizes of the mitochondria were performed using Jandel SigmaStat software (V 3.5).

5) Statistical analyses

The compared values were calculated as the mean ± SD of three brains from each group,

and statistical significance was determined using Student’s t-tests.

7
1.3 Result

1) Imbalanced expression of mitochondrial fusion and fission proteins in infected brains

First, we identified deposits of PK-resistant PrPSc in ME7-infected brains in the end

stage; the normal PrPC proteins were completely degraded by the PK treatment, as shown

in Fig. 1. To determine whether the mitochondrial fusion and fission proteins are affected

by prion infection, their expression patterns were investigated in whole brains of the

control and ME7-infected mice at the end stage of disease (160 dpi) (Fig. 2). Of the

mitochondrial fusion and fission proteins (fusion proteins: Opa1, Mfn1 and Mfn2; fission

proteins: Dlp1 and Fis1), Western blot analysis revealed that only Mfn1 was differentially

expressed in the infected whole brains compared to the age-matched control brains (Fig.

2A and B). It has previously been reported that the hippocampal regions are more severely

damaged than any other regions in brains infected with the ME7 scrapie strain [18]. Thus,

to compare the expression levels of the mitochondrial fusion and fission proteins in

various brain regions, we dissected both the control and ME7-infected brains into the

following regions: cerebral cortex, hippocampus, cerebellum, striatum and brainstem.

We found that the levels of Mfn1 and Fis1 were significantly increased in the

hippocampi of the infected brains, whereas the Dlp1 levels were significantly decreased

at the end stage (Fig. 2C and D). Additionally, the levels of Mfn1 and Mfn2 were

increased in the cerebral cortex and striatum, respectively, of infected brains (data not

shown). In the cerebellum and brain stem, none of the five mitochondrial fusion and

fission proteins was significantly differentially expressed between the control and

infected mice (data not shown). These results indicate that the differential regulation of

mitochondrial fusion and fission proteins may be involved in the mitochondrial

dysfunction of the ME7-infected brains, particularly in the hippocampus [16].

8
2) Analysis of fission proteins in the hippocampal cytosol of infected brain

To examine the localization of the mitochondrial fusion and fission proteins in the

organelles and the expression levels of these proteins in the cytosol and mitochondria,

mitochondrial and cytosolic fractions were isolated from the hippocampi of control and

ME7-infected mice; again the hippocampus was chosen because this region is known to

be severely damaged in ME7-infected brains [18]. Immunoblot analyses revealed that

Dlp1 levels in the mitochondrial fractions from the hippocampi were not altered by ME7

infection (Fig. 3A and B) but were significantly decreased in the cytosolic fractions of the

infected hippocampi; this finding is consistent with the decrease in Dlp1 levels in the

affected hippocampi that is illustrated in Fig. 2C and D. The concentration of Opa1, which

is primarily localized to the inner membrane of the mitochondria, was not altered in the

mitochondria or cytosol isolated from the hippocampi of the infected brains (Fig. 3C and

D).

3) Morphological changes in the mitochondria in the hippocampal region of infected brain

The modulation of mitochondrial dynamics due to fusion and fission protein

concentration probably contributed to alterations in mitochondrial shape [9]. Electron

microscopic analyses were performed to assess the relationship between mitochondrial

fusion and fission proteins and mitochondrial shapes in the hippocampal region. As

shown in Fig. 4, the mitochondria in the normal hippocampal neurons were present in a

variety of shapes, including rod, round and elliptical shapes. The cristae were normal in

appearance. In contrast, enlarged mitochondria with damaged cristae were clearly

observed in many of the hippocampal neurons in infected brains (Fig. 4A). The numbers

9
of total neuronal mitochondria were significantly decreased in the hippocampi of the

infected mice compared to those of controls (Fig. 4B). Additionally, the lengths and

widths of the mitochondria in the hippocampal neurons of infected brains were

significantly increased compared to those of the controls, which is consistent with the

observation of enlarged hippocampal mitochondria visualized in Fig. 4B. The

morphological changes seen in mitochondria may be involved in the mitochondrial

dysfunction in the hippocampi of ME7-infected mice.

10
1.4 Discussion

In this study, we demonstrated for the first time that the expression patterns of

mitochondrial fusion and fission proteins were altered in an experimental mouse model

of prion disease; i.e., ME7 scrapie-infected mice. Of the mitochondrial fusion and fission

proteins examined, the levels of Mfn1 were found to be significantly increased in whole

brains of the ME7-infected mice. Within the different dissected regions of the ME7-

infected brains, we found differences in the expression levels of various mitochondrial

fusion and fission proteins compared to controls. Particularly notable were the alterations

in the expression of Mfn1 in the cerebral cortex; Mfn1, Dlp1 and Fis1 in the hippocampus;

and Mfn2 in the striatum.

A recent study suggested that most neurodegenerative diseases may be associated with

the dysregulation of mitochondrial fusion and fission proteins [19]. Mitochondrial fusion

and fission are regulated by large dynamin-related GTPases. Mitochondrial fusion, which

is regulated by three large GTPases (Mfn1, Mfn2 and Opa1), involves the coordinated

fusion of both the outer and inner mitochondrial membranes [20]. The overexpression of

mitofusins causes the normally punctate mitochondria to become elongated, whereas the

repression of mitofusins causes the fragmentation of the mitochondrial network in tissue

culture cells [21]. Interestingly, we found increased level of Mfn1, which is a

transmembrane protein that localizes to the outer membranes of mitochondria, in the

homogenates of whole brain, hippocampus and cerebral cortex of the infected brain. In

addition the expression level of Mfn2 was increased in the striatum of the infected brain

(Data not shown). Although electron microscopy analyses did not reveal the detailed

membrane structures of the mitochondria in this study, initial outer membrane fusion may

11
have occurred in the brains of the infected mice due to increases in the Mfn1 and Mfn2

proteins. Moreover, although there may be distinct pathways or mechanisms for Mfn1

and Mfn2, a functional interplay between the two proteins may be involved in the control

of mitochondrial fusion [22].

Different strains of mouse-adapted scrapie have been reported to preferentially target

different specific brain regions; as noted previously, the ME7 strain is known to be

particularly associated with hippocampal damage [23]; therefore, we focused on the

hippocampal region of the ME7-infected brains. Our study demonstrated that ME7

infection led to significant increases in the levels of Mfn1 and Fis1 in the hippocampi of

the ME7-infected brains, whereas Dlp1 was significantly decreased. Dlp1 localizes in

several organelles, including the endoplasmic reticulum, microtubules and peroxisomes,

but primarily resides in the mitochondria and cytosol [24]. We isolated the cytosolic

fraction of the hippocampal region and investigated the changes in the expression of

mitochondrial fusion and fission proteins in both the purified mitochondria fraction and

the cytosol. Dlp1 levels were decreased in cytosolic fractions but not in mitochondria

fractions. These observations imply that imbalances in mitochondrial dynamics may

contribute to the enlargement and the degeneration of mitochondria that occurs in the

hippocampi of scrapie-infected mice.

Changes in the shape of the mitochondria have been observed in ME7 scrapie-infected

brain even in the preclinical stage of infection [25]. The mitochondrial enlargement

demonstrated during the end stage of prion disease in this study was also observed in a

previous study that demonstrated structural abnormalities of the mitochondria in the

neurons of the hippocampi and cerebral cortices of brains from scrapie-infected hamsters

[15]. Fragmentation and clustering of mitochondria in the soma have been observed in

12
the brains of patients with sporadic AD, and alterations in the expressions of several

fusion and fission proteins have been observed in these brains [26]. Thus, we sought to

determine the relationship between expression levels of the mitochondrial fusion and

fission proteins and the alterations in the shapes, numbers and sizes of the mitochondria

in a prion disease model. In the ME7 mouse model, we found that the total number of

neuronal mitochondria was reduced significantly and that a number of enlarged and

degenerated mitochondria appeared. These mitochondria had damaged cristae and matrix.

The relationship between the mitochondrial damage and the induction of

neurodegeneration and disease is uncertain. Clearly the fact that the number of

mitochondria was reduced combined with degeneration of the remaining mitochondria

would lead to a loss of neuronal function. It needs to be noted that the loss of mitochondria

could affect the quantity of proteins observed; this is particularly important in instances

where the level of protein decreased (e.g., Dlp1 in the cytosol). Furthermore, in those

instances where there was no change in protein levels (e.g., Opa1), the reduced number

of mitochondria could mask an increase.

Growing evidence suggests that the delicate balance between mitochondrial fission and

fusion is critical for mitochondrial functions, including energy production, Ca2+

signaling, reactive oxygen species (ROS) production, apoptosis and senescence [7,27].

Dysregulation of the mitochondrial fusion and fission proteins in neurons may be a

common pathway leading to the mitochondrial and neuronal dysfunctions that are critical

in the pathogenesis of prion disease. It remains unclear, however, how the alterations in

mitochondrial fusion and fission proteins that were observed in this study contribute to

neurodegeneration in prion diseases. Taken together, the findings of this study suggest

that the disruption of the balance of mitochondrial fusion and fission proteins, which may

13
be a common feature of prion diseases, is involved in the mitochondrial and neuronal

dysfunction in the brains of ME7-infected mice.

14
Fig. 1.1. Deposition of PK-resistant prion isoforms in ME7-infected mice. Whole

brains of control (n = 3) and infected mice (n = 3) in the end stage (160 dpi) were

homogenized and blotted with anti-PrP antibody (3F10). A portion of each sample was

treated with PK (PK 50 ug/ml). β-Actin was used as a loading control.

15
Fig. 1.2. Differential expression of mitochondrial fusion and fission proteins in

hippocampi of ME7-infected mice. The mitochondrial dynamic proteins of the whole

brains and hippocampal regions of the control (n = 3) and infected mice (n = 3) in the end

stage of disease were blotted. β-Actin was used as a loading control for A and C. The

intensities of the bands in panels A and C were measured and quantified (B and D). The

values are expressed as the mean ± SD (n = 3). CON: control (white bars); ME7: ME7-

infected (black bars). Statistically significant differences are indicated (*p < 0.05 and **p

< 0.01).

16
Fig. 1.3. Subcellular localization of mitochondrial fusion and fission proteins in

hippocampi of ME7-infected mice. Opa1 (fusion) and Dlp1 (fission) from the

mitochondria (A) and cytosolic (C) fractions of the hippocampi of the control (n = 3) and

ME7-infected mice (n = 3) at the end stage of the disease were blotted (160 dpi). β-Actin

was used as a loading control. The intensities of the bands in panels A and C were

measured and quantified (B and D). The values are expressed as the mean ± SD (n = 3).

CON: control (white bars); ME7: ME7-infected (black bars). Note that both of the Opa1

bands are primarily localized to the mitochondria of hippocampi in both the control and

infected specimens, whereas substantial amounts of Dlp1 were localized to the cytosol.

Statistically significant differences are indicated (*p < 0.05).

17
Fig. 1.4. Electron microscopic analysis of the mitochondria in hippocampal neurons

of ME7-infected mice. (A) Control mitochondria were elliptical in shape and exhibited

compact cristae, whereas the infected mitochondria were enlarged and exhibited partially

swollen cristae. Scale bar, 1 ㎛. (B) The total number of mitochondria in the hippocampal

neurons of the control and ME7-infected mice (the total number of mitochondria were

counted from 15 neuronal cells from each group). (C) The lengths (white bar) and widths

(black bar) of the mitochondria in the hippocampal neurons of the control and ME7-

infected mice (the lengths and widths of 30 mitochondria in each group were measured).

Statistically significant differences are indicated (*p < 0.01). CON: control; ME7: ME7-

infected.

18
1.5 References

[1] A. Aguzzi, C. Weissmann, Prion research: the next frontiers, Nature 389 (1997) 795–

798.

[2] D.C. Bolton, M.P. McKinley, S.B. Prusiner, Identification of a protein that purifies

with the scrapie prion, Science 218 (1982) 1309–1311.

[3] R.H. Kimberlin, C. Walker, Characteristics of a short incubation model of scrapie in

the golden hamster, J. Gen. Virol. 34 (1977) 295–304.

[4] R.L. Chandler, B.A. Turfrey, Inoculation of voles, Chinese hamsters, gerbils and

guinea-pigs with scrapie brain material, Res. Vet. Sci. 13 (1972) 219–224.

[5] R.I. Carp, X. Ye, R.J. Kascsak, R. Rubenstein, The nature of the scrapie agent.

Biological characteristics of scrapie in different scrapie strain–host combinations, Ann.

N. Y. Acad. Sci. 724 (1994) 221–234.

[6] P.J. Lucassen, A. Williams, W.C. Chung, H. Fraser, Detection of apoptosis in murine

scrapie, Neurosci. Lett. 198 (1995) 185–188.

[7] B. Su, X. Wang, L. Zheng, G. Perry, M.A. Smith, X. Zhu, Abnormal mitochondrial

dynamics and neurodegenerative diseases, Biochim. Biophys. Acta 2010 (1802) 135–142.

[8] M.T. Lin, M.F. Beal, Mitochondrial dysfunction and oxidative stress in

neurodegenerative diseases, Nature 443 (2006) 787–795.

[9] H. Chen, D.C. Chan, Mitochondrial dynamics-fusion, fission, movement, and

mitophagy-in neurodegenerative diseases, Hum. Mol. Genet. 18 (2009) R169–R176.

[10] X. Wang, B. Su, S.L. Siedlak, P.I. Moreira, H. Fujioka, Y. Wang, G. Casadesus, X.

Zhu, Amyloid-beta overproduction causes abnormal mitochondrial dynamics via

19
differential modulation of mitochondrial fission/fusion proteins, Proc. Natl. Acad. Sci.

U.S.A. 105 (2008) 19318–19323.

[11] H. Deng, M.W. Dodson, H. Huang, M. Guo, The Parkinson’s disease genes pink1

and Parkin promote mitochondrial fission and/or inhibit fusion in Drosophila, Proc. Natl.

Acad. Sci. U.S.A. 105 (2008) 14503–14508. 2.

[12] T. Milakovic, G.V. Johnson, Mitochondrial respiration and ATP production are

significantly impaired in striatal cells expressing mutant huntingtin, J. Biol. Chem. 280

(2005) 30773–30782.

[13] A.V. Panov, C.A. Gutekunst, B.R. Leavitt, M.R. Hayden, J.R. Burke, W.J.

Strittmatter, J.T. Greenamyre, Early mitochondrial calcium defects in Huntington’s

disease are a direct effect of polyglutamines, Nat. Neurosci. 5 (2002) 731–736.

[14] F. Squitieri, M. Cannella, G. Sgarbi, V. Maglione, A. Falleni, P. Lenzi, A. Baracca,

G. Cislaghi, C. Saft, G. Ragona, M.A. Russo, L.M. Thompson, G. Solaini, F. Fornai,

Severe ultrastructural mitochondrial changes in lymphoblasts homozygous for

Huntington disease mutation, Mech. Ageing Dev. 127 (2006) 217–220.

[15] S.I. Choi, W.K. Ju, E.K. Choi, J. Kim, H.Z. Lea, R.I. Carp, H.M. Wisniewski, Y.S.

Kim, Mitochondrial dysfunction induced by oxidative stress in the brains of hamsters

infected with the 263 K scrapie agent, Acta Neuropathol. 96 (1998) 279–286.

[16] J.H. Park, B.H. Kim, S.J. Park, J.K. Jin, Y.C. Jeon, G.Y. Wen, H.Y. Shin, R.I. Carp,

Y.S. Kim, Association of endothelial nitric oxide synthase and mitochondrial dysfunction

in the hippocampus of scrapie-infected mice, Hippocampus 21 (2010) 319–333.

[17] J.K. Choi, S.J. Park, Y.C. Jun, J.M. Oh, B.H. Jeong, H.P. Lee, S.N. Park, R.I. Carp,

Y.S. Kim, Generation of monoclonal antibody recognized by the GXXXG motif (glycine

zipper) of prion protein, Hybridoma (Larchmt) 25 (2006) 271–277.

20
[18] W.K. Ju, K.J. Park, E.K. Choi, J. Kim, R.I. Carp, H.M. Wisniewski, Y.S. Kim,

Expression of inducible nitric oxide synthase in the brains of scrapie-infected mice, J.

Neurovirol. 4 (1998) 445–450.

[19] S. Frank, Dysregulation of mitochondrial fusion and fission: an emerging concept in

neurodegeneration, Acta Neuropathol. 111 (2006) 93–100.

[20] D.C. Chan, Mitochondrial fusion and fission in mammals, Annu. Rev. Cell Dev. Biol.

22 (2006) 79–99.

[21] A. Sante, M.T. Fuller, Control of mitochondrial morphology by a human mitofusin,

J. Cell Sci. 114 (2001) 867–874.

[22] S.A. Detmer, D.C. Chan, Complementation between mouse Mfn1 and Mfn2 protects

mitochondrial fusion defects caused by CMT2A disease mutations, J. Cell Biol. 176

(2007) 405–414.

[23] R.M. Deacon, D. Reisel, V.H. Perry, J. Nicholas, P. Rawlins, Hippocampal scrapie

infection impairs operant DRL performance in mice, Behav. Brain Res. 157 (2005) 99–

105.

[24] Y. Yoon, E.W. Krueger, B.J. Oswald, M.A. McNiven, The mitochondrial protein

hFis1 regulates mitochondrial fission in mammalian cells through an interaction with the

dynamin-like protein DLP1, Mol. Cell. Biol. 23 (2003) 5409–5420.

[25] Z. Sisková, D.J. Mahad, C. Pudney, G. Campbell, M. Cadogan, A. Asuni, V.

O’Connor, V.H. Perry, Morphological and functional abnormalities in mitochondria

associated with synaptic degeneration in prion disease, Am. J. Pathol. 177 (2010) 1411–

1421.

21
[26] X. Wang, B. Su, H.G. Lee, X. Li, G. Perry, M.A. Smith, X. Zhu, Impaired balance

of mitochondrial fission and fusion in Alzheimer’s disease, J. Neurosci. 29 (2009) 9090–

9103.

[27] S. Frank, B. Gaume, E.S. Bergmann-Leitner, W.W. Leitner, E.G. Robert, F. Catez,

C.L. Smith, R.J. Youle, The role of dynamin-related protein 1, a mediator of

mitochondrial fission, in apoptosis, Dev. Cell 1 (2001) 515–525.

22
II. Chapter 2

Upregulation of Connexin 43 Expression Via C-Jun N-Terminal

Kinase Signaling in Prion Disease

Abstract

Prion infection leads to neuronal cell death, glial cell activation, and the accumulation

of misfolded prion proteins. However, the altered cellular environments in animals with

prion diseases are poorly understood. In the central nervous system, cells connect the

cytoplasm of adjacent cells via connexin (Cx)-assembled gap junction channels to allow

the direct exchange of small molecules, including ions, neurotransmitters, and signaling

molecules, which regulate the activities of the connected cells. Here, we investigate the

role of Cx43 in the pathogenesis of prion diseases. Upregulated Cx43 expression, which

was dependent on c-Jun N-Terminal Kinase (JNK)/c-Jun signaling cascades, was found

in prion-affected brain tissues and hippocampal neuronal cells. Scrapie infection induced

Cx43 formed aggregated plaques within the cytoplasmic compartments at the cell-cell

interfaces. The ethidium bromide (EtBr) uptake assay and scrape-loading dye transfer

assay demonstrated that increased Cx43 has functional consequences for the activity of

Cx43 hemichannels. Interestingly, blockade of PrPSc accumulation reduced Cx43

expression through the inhibition of JNK signaling, indicating that PrPSc accumulation

may be directly involved in JNK activation-mediated Cx43 upregulation. Overall, our

findings describe a scrapie infection-mediated novel regulatory signaling pathway of

Cx43 expression and may suggest a role for Cx43 in the pathogenesis of prion diseases.

23
Keywords: Connexin 43, gap junction, JNK, prion protein, scrapie

24
2.1 Introduction

Gap junctions are a complex of membrane channels that allow the diffusion of small

molecules (<1.5 kDa) between adjacent cells to form a functional syncytium. Gap

junctions are composed of two hemichannels (connexons) formed by hexamers of

connexins (Cxs) [1]. Of the Cx genes that code for gap junction proteins, Cx43 is the most

ubiquitous; it is abundantly expressed in the brain and highly expressed during embryonic

development [2, 3]. In the central nervous system (CNS), Cx43 is widely expressed in

neurons, astrocytes, and oligodendrocytes [4–6], and various signal molecules, including

ATP, glutamate, and Ca2+ ions, diffuse through functional Cx43 hemichannels [7–11],

thereby modulating crucial CNS processes [12].

Cx43 has contributed to neuronal death in vitro [11, 13], in a rat cortical ablation model

[14], and in ischemic brain injury [15]. Cx43 plays a protective role against oxidative

stress-induced cell death [16, 17]; deletion or blockade of this protein prevents chronic

neuropathic pain following spinal cord injury [18, 19] and fetal ischemia [20]. The

deletion of Cx43 or expression of a truncated form increases the vulnerability to stroke

[21, 22]. In addition, Cx43 is involved in neuronal differentiation [23–25], cellular

mortality [26–29], hippocampal proliferation, and the survival of newborn cells [30].

Moreover, the role of Cx43 has been implicated in various pathological conditions of

the brain. Increased Cx43 levels have been identified within amyloid plaques in the brains

of Alzheimer’s disease patients and in an animal model of the disease [31, 32], as well as

in the anterior horns of mSOD1-Tg mice, which represent an amyotrophic lateral sclerosis

mouse model [33], the MPTP-lesioned striatum of a Parkinson’s disease model [34], and

the hippocampal regions of patients with mesial temporal lobe epilepsy [35]. In contrast,

25
the loss of Cx43 expression is found in the actively demyelinating lesions of multiple

sclerosis and in the active perivascular lesions of neuromyelitis optica [36]. Therefore,

Cx43 is associated with the pathophysiology of various tissue conditions. However, to

the best of our knowledge, there are no data available regarding the role of Cx43 in prion

diseases.

Transmissible spongiform encephalopathies or prion diseases are infectious and fatal

neurodegenerative diseases. A mutation and an abnormal structure of the prion protein

(PrPC) are associated with a protease-resistant and infectious form (PrPSc), which is

considered a causative factor for prion diseases. Neuropathologically, prion diseases are

characterized by a long incubation period of many months to several decades, neuronal

vacuolation (spongiosis), neuronal cell death, and glial cell activation (microgliosis and

astrocytosis) [37]. These changes lead to the release of inflammatory molecules, such as

proinflammatory cytokines, reactive oxygen species, proteases, and complement proteins

that induce neuronal damage and remove the damaged cells [37].

The aim of the present study was to determine the specific role of Cx43 in prion

pathogenesis using cellular and mouse models of prion disease. In this study, we report

for the first time that Cx43 expression is increased via the c-Jun N-Terminal Kinase (JNK)

signaling pathway in both in vitro and in vivo models of prion disease.

26
2.2 Materials and methods

1) Animals and scrapie strains

C57BL/6J mice and golden Syrian hamsters (4 to 6 weeks of age) were purchased from

Central Lab Animal, Inc. (Seoul, Republic of Korea). The original stocks of the ME7,

22L, 139A, and 263K scrapie strains were kindly provided by Dr. Alan Dickinson of the

Agriculture and Food Research Council and Medical Research Council Institute

(Neuropathogenesis Unit, Edinburgh, UK). For scrapie infection, the mice were

intracerebrally inoculated with 30 μl of 1% w/v brain homogenates of the ME7, 22L, and

139A inocula (for mice) or 50 μl of 1% w/v hamster brain homogenate of the 263K

inoculum (for hamsters) in phosphate-buffered saline (PBS, pH 7.4) using a stereotaxic

apparatus (Stoelting, Wood Dale, IL, USA). The control mice received 30∼50 μl of 1%

w/v normal brain homogenate. The scrapie-infected and uninfected mice were sacrificed

at 10–150 days post-inoculation (dpi) or at the terminal stage (160 dpi for ME7, 140 dpi

for 22L, and 170 dpi for 139A) when the mice displayed typical clinical signs of the

disease. All experiments were performed in accordance with Korean laws and with the

approval of the Hallym Medical Center Institutional Animal Care and Use Committee

(HMC2011-0-0115-07).

2) Subcellular fractionation

Mouse brain tissues were lysed in cold hypotonic buffer (10mM Tris-HCl, pH 7.4; 1mM

DTT; 5mM MgCl2; 10mM KCl; 10mM NaF; and 1mM Na3VO4) with a protease inhibitor

cocktail tablet (Roche, Indianapolis, IN, USA) by passing through a 23-gauge syringe

needle 10 times. The lysates were centrifuged at 500 g for 10 min to remove the nuclei

27
and unbroken cells. The post-nuclear supernatants were subsequently centrifuged at

100,000 g for 1 h at 4°C to separate the membrane pellet from the cytosolic fraction. The

membrane pellets were washed with ice-cold PBS and resuspended in modified RIPA

buffer (50mM Tris-HCl, pH 7.4; 150mM NaCl; 2mM EDTA; 1% Triton X-100; 1%

Nonidet P (NP)-40; 0.25% sodium deoxycholate; 1mM Na3VO4; and 10mM NaF) with a

protease inhibitor cocktail tablet by rocking for 1 h at 4°C, followed by centrifugation at

20,000 g for 10 min at 4°C. The supernatant contained the solubilized membrane proteins.

3) Western blot analysis

The mouse brains and cultured cells were lysed in modified RIPA buffer with 20mM

Tris-HCl (pH 7.5), 150mM NaCl, 1% Triton, 0.5% sodium deoxycholate, 0.1% sodium

dodecyl sulfate (SDS), 1% NP-40, 10mM NaF, 1mM Na3VO4, 1mM EDTA, 1mM EGTA

and 0.1Mphenylmethylsulfonyl fluoride (PMSF), as well as a protease inhibitor cocktail.

The homogenates were centrifuged at 15,000 g at 4°C for 30 min, the supernatants were

collected, and the protein concentrations were determined using a BCA protein assay kit

(Pierce, Rockford, IL, USA). Equal protein amounts were separated by 10 or 12% SDS-

PAGE and then transferred to PVDF membrane using an electrotransfer system (Bio-Rad,

Hercules, CA, USA). To detect the target proteins, mouse monoclonal anti- PrP (3F10,

1:3,000) [38] rabbit polyclonal anti-Cx43 (1:1,000; Cell Signaling Technology, Beverly,

MA, USA), mouse monoclonal anti-JNK (1:1,000; Santa Cruz Biotechnology, Santa Cruz,

CA, USA), mouse monoclonal anti-phospho-JNK (1:1,000, Santa Cruz Biotechnology),

rabbit polyclonal anti-c-Jun (1,000; Santa Cruz Biotechnology), rabbit polyclonal

antiphospho- c-Jun (1:1000; Cell Signaling Technology), and mouse monoclonal anti-β-

actin (1:10,000; Sigma- Aldrich, Saint Louis, MO, USA) antibodies were used with the

28
appropriate secondary antibodies conjugated to horseradish peroxidase. To detect PrPSc,

50 μg/ml or 20 μg/ml of proteinase K (PK) were treated for 30 min at 37°C in cell

lysates or brain homogenates, respectively. The target signals were visualized by digital

images captured with an ImageQuant™ LAS 4000 imager (GE Healthcare Life Sciences,

Piscataway, NJ, USA) using an enhanced chemiluminescence western blot detection

system (Amersham Biosciences, Piscataway, NJ, USA).

4) Maintenance and scrapie infection of cultured cell lines

Mouse hippocampal neuronal cell lines, including ZW13-2 (wild-type PrP) and Zpl2-4

(PrP knockout) cells, were previously established [39]. The cells were cultured in

Dulbecco’s modified Eagle’s medium (Hyclone, Logan, UT, USA) with 10% fetal bovine

serum (FBS, Hyclone), 100 units/ml penicillin, and 100 μg/ml streptomycin in a 37°C

incubator with 5% CO2. The ZW13-2 cells were persistently infected with the 22L and

139A scrapie strains as previously described [40]. The infected cells were maintained in

Opti-MEM (Sigma-Aldrich) with 10% fetal calf serum (Hyclone) and sub-cultured every

3 days at a 1:2 split for the first 10 passages. The infected cells stably produced PrPSc for

over 50 passages. For immunocytochemistry, dye uptake assay, and scrape-loading dye

transfer assay, cells were seeded onto coverslips at 2×104 cells per well in 24-well plates

and allowed to adhere for 2 days.

5) Cell blot assay

Stable scrapie infection was confirmed after five passages as previously described [40].

Briefly, cells were grown to confluence on Thermanox plastic cover slips (Nalgene Nunc

International, Rochester, NY, USA) and transferred to nitrocellulose membrane. After

29
drying for 1 h at 37°C, the membrane was incubated in lysis buffer (50mM Tris-HCl, pH

8.0; 150mM NaCl; 0.5% sodium deoxycholate; 0.5% Triton X-100; 2mM PMSF)

containing PK (5 ㎍/ml) at 37°C for 10 min. The membrane was placed into 3 M

guanidinium thiocyanate (Sigma-Aldrich), 10mM Tris·HCl (pH 8.0) for 10 min followed

by immunostaining with anti-PrP antibody 3F10 (1:3,000).

6) Semi-quantitative RT-PCR

Total RNA was extracted from the brain samples using TRI reagent (Sigma-Aldrich)

according to the manufacturer’s protocols. cDNA was synthesized using the Moloney

murine leukemia virus reverse transcriptase (Promega, Madison, WI, USA). The primer

sequences were as follows: connexin43 (127 bp), sense, 5′-

CTGAGTGCGGTCTACACCTG-3′, antisense, 5′-GAGCGAGAGACACCAAGGAC-

3′; β-actin (196 bp), sense, 5′-TGTGATGGACTCCGGTGACGG-3′, antisense, 5′-

ACAGCTTCTCTTTGATGTCACGC-3′. The PCR products were separated by

electrophoresis on a 1% agarose gel and visualized under UV light.

7) Immunohistochemistry

Immunohistochemical procedures were performed as previously described [41]. The

sections were blocked with 10% normal donkey serum in Tris-buffered saline (50mM

Tris-HCl and 150mM NaCl, pH 7.6) and then incubated overnight at 4°C with rabbit

polyclonal anti-Cx43 antibody (1:100). After washing in PBS, the sections were first

incubated with biotinylated donkey anti-rabbit IgG antibody (1:500; Vector Laboratories,

Burlingame, CA, USA) for 1 h and then with avidin-biotin peroxidase complex (ABC Kit,

Vector Laboratories). The sections were mounted in Permount (Thermo Fisher Scientific,

30
Inc., Waltham, MA, USA). The control sections were stained without primary antibody.

8) Immunocytochemistry

The cells were fixed with 4% paraformaldehyde in PBS for 15 min at room temperature

and then treated with 0.1% Triton X-100 in PBS. After a brief wash with PBS, the cells

were incubated with blocking buffer (5% normal goat serum and 0.1% Tween 20 in PBS)

for 1 h, followed by incubation with rabbit polyclonal anti-Cx43 antibody (1:200, Cell

Signaling Technology) and mouse monoclonal anti-PrP (3F10, 1:200) [38] overnight at

4°C. The cells were subsequently incubated with either Alexa Fluor 568 goat anti-rabbit

IgG or Alexa Fluor 488 goat anti-mouse IgG antibodies (Invitrogen, Carlsbad, CA, USA)

for 1 h. Control reactions omitting the primary antibodies resulted in no labeling with the

secondary antibodies (data not shown). After rinsing with PBS, the cells were then

mounted in 4′,6-diamidino-2-phenylindole (DAPI)-containing Vectashield Mounting

Medium (Vector Laboratories) to label nuclei and visualized using a confocal laser

scanning microscope (LSM 700; Carl Zeiss, Oberkochen, Germany).

9) Dye uptake assay

To evaluate the functional Cx43 hemichannel, an uptake assay using the hemichannel,

permeable reporter dye ethidium bromide (EtBr) was performed as previously described

[42]. The cells were incubated with 5 μM EtBr in PBS in the presence or absence of a

connexin hemichannel blocker LaCl3 (500 μM) for 5 min at 37°C. For JNK inhibition,

the cells were pretreated with the JNK inhibitor SP600125 (50 μM) for 6 h and then

incubated with 5 μM EtBr for 5 min at 37°C. The cells were then washed with Hank’s

balanced salt solution (HBSS), fixed with 4% paraformaldehyde in PBS for 15 min at

31
room temperature and washed with HBSS. Dye uptake was examined with a confocal

laser scanning microscope.

10) Scrape-loading dye transfer assay

To determine the functional Cx43 hemichannels, an uptake assay using the gap junction

permeable reporter dye Lucifer yellow (LY) (Sigma-Aldrich) was performed as

previously described [43]. The cells were incubated with 0.01% LY in the presence or

absence of the Cx channel blocker lanthanum chloride (LaCl3, 500 μM ) for 5 min at

37°C. For JNK inhibition, the cells were pretreated with a JNK inhibitor SP600125 (50

μM) for 6 h and then incubated with 0.01% LY in PBS for 5 min at 37°C. The cells were

washed with HBSS and fixed with 100% MeOH for 15 min at –20°C. The cells were

observed under a confocal laser scanning microscope. The distances of LY diffusion after

scrape loading were measured in at least twenty random areas from each sample, and the

fluorescence intensity was assessed using Image J software and compared between the

control and infected cells with or without each treatment.

11) Brilliant blue G (BBG) treatment

Uninfected and 22L scrapie-infected cells (1×106 cells/100 mm dish) were treated or not

treated with BBG (0.6–60 μM) for 3 days and then lysed. The expression levels of PrPSc,

JNK, phospho-JNK, and Cx43 were determined by western blotting as previously

described.

12) Statistical analysis

The data are expressed as the mean±SEM. Significant differences between the

32
experimental groups were evaluated using one-way analysis of variance (ANOVA).

Statistical significance was defined as ∗p < 0.05, ∗∗p < 0.01, and ∗∗∗p < 0.001.

33
2.3 Results

1) Upregulation of Cx43 expression in scrapie-infected mouse brains

We first investigated the expression levels of the Cx43 protein in the brain tissues of

control mice and mice infected with one of three scrapie strains (ME7, 22L, or 139A).

These strains possess distinct incubation periods and neuropathological features [44]. In

the whole brain and dissected brain tissues, including the cerebral cortex, hippocampus,

striatum, cerebellum, and brain stem, we demonstrated that Cx43 protein Fig. 1A, B) and

mRNA (Fig. 1C) were highly expressed in animals with scrapie infection compared with

the controls. These results suggest that both the mRNA and protein levels of Cx43 are

upregulated after scrapie infection, and the upregulated protein levels of Cx43 in most

brain regions are a result of increased gene expression in scrapie-infected mice.

To further determine whether the increased Cx43 protein expression is correlated with

disease progression, we performed western blotting using whole brains obtained at 50,

100, and 150 dpi for the ME7 strain (for mice) and at 10, 30, 60, and 90 dpi for the 263K

strain (for hamsters) (Fig. 1D). No difference between the control and scrapie-infected

brains was identified at 50 dpi in ME7 mice or in 263K hamsters at 30 dpi. However,

Cx43 protein expression tended to increase relative to the stage of scrapie development

and was significantly increased at 90 dpi (for hamsters) and 150 dpi (for mice). These

time points were correlated with marked accumulations of PrPSc, indicating that Cx43

protein increased at the end stage of scrapie infection.

To confirm the increase in Cx43 after scrapie infection, we subsequently performed

immunohistochemical staining of Cx43 in various brain regions of the control and

scrapie-infected mice. Cx43 was prominent in most brain regions, including the cerebral

34
cortex, hippocampus, striatum, cerebellum, and brain stem, at the end stage of scrapie

infection compared with levels in the controls (Fig. 2). A marked increase in Cx43

immunoreactivity was seen in the hippocampus and the Purkinje layer of the cerebellum

in the scrapie-infected mice. In addition, Cx43, which showed a diffuse staining pattern,

localized to regions of cell-cell contact or blood vessels (arrows in Fig. 2). Overall, these

findings suggest that scrapie infection increases Cx43 expression in the brain.

2) Increased Cx43 in the membrane fraction from the brains of scrapie-infected mice

Because Cx43 is an integral plasma membrane protein, we subsequently examined

whether the increased Cx43 is related to membrane localization using cytosolic and

membrane fractions prepared from control brains and from ME7, 22L, and 139A scrapie-

infected brains as described in the Materials and Methods. Calnexin and enolase were

used as markers of the membrane and cytosolic fractions, respectively. As expected, the

Cx43 protein was preferentially detected in the membrane fraction of all scrapie-infected

brains compared with levels in the controls (Fig. 3), which is consistent with the

immunoblot analysis data (Fig. 1). These data suggest that increases in membrane

associated Cx43 protein result from scrapie-induced pathological conditions.

3) Increased Cx43 expression in scrapie-infected hippocampal cell lines

Gap junctions are complexes of intercellular channels between adjacent cells [1]. We

subsequently examined whether a scrapie infection-induced increase in Cx43 expression

is associated with gap junction plaque formation in hippocampal neuronal cell lines, as

previously established [39]. Although Cx43 is a primary gap junction protein in astrocytes,

we demonstrated that the ZW13-2 and Zpl2-4 hippocampal neuronal cell lines

35
endogenously express Cx43. Thus, these cell lines were used as an in vitro model of prion

replication after infection with either of two mouse derived scrapie strains (22L or 139A).

As shown in Fig. 4, Cx43 (red) was detected in both ZW13-2 and Zpl2-4 cells, and intense

expression of Cx43 was mainly localized within plaques at the cell-cell contact areas. In

the presence of PrP (ZW13-2), Cx43 staining consisted of sparse but large puncta

distinctly separated from the nucleus (blue). Interestingly, Cx43 was clearly detected with

more aggregates between neighboring scrapie-infected ZW13-2 cells but not between

Zpl2-4 cells or uninfected cells. Uptake of infection was confirmed by the PK-resistant

PrPSc levels in cultures after multiple passages (Supplementary Figure 1). These results

demonstrated that pathogenic PrP or scrapie infection induce the upregulation of Cx43

expression and the formation of gap junction-like plaques in neuronal cells.

4) Association of JNK activation and Cx43 upregulation in scrapie-infected cells and

mice

JNK is a stress-activated kinase reported to be important in intracellular signaling

pathways that regulate Cx43 expression [45–47]. To determine whether JNK signaling is

involved in the induction of Cx43, we examined the activation of JNK and its downstream

molecule c-Jun in scrapie-infected neuronal cells and mice. Scrapie infection was

confirmed by the detection of PK-resistant PrPSc (Fig. 5A, second panels). Interestingly,

Cx43 was upregulated and associated with JNK activation, which was demonstrated by

increased phospho-JNK (P-JNK) in scrapie-infected neuronal cells and in infected brains

(Fig. 5A). The levels of P-c-Jun were also significantly increased in association with JNK

activation; however, the total levels of JNK and c-Jun were not altered. To further

elucidate the functional roles of JNK in the induction of Cx43 by scrapie infection, control

36
and infected cell lines were treated with various concentrations of an ATP-competitive

inhibitor of JNK, SP600125 (0–100 μM), for 6 h. As shown in Fig. 5B and C, increased

Cx43 and P-JNK/P-c-Jun by scrapie infection were effectively downregulated by

SP600125 treatment in a dose- and time-dependent manner. These findings suggest that

JNK-c-Jun signaling is essential for the Cx43 overexpression induced by scrapie infection.

5) Increased dye uptake by scrapie-induced Cx43 expression

The uptake of the fluorescent dye EtBr has been used as an indicator of hemichannel

opening [42, 48]. To determine whether Cx43 upregulation by scrapie infection affects

the opening of Cx43 hemichannels, we performed EtBr uptake assays in scrapie-infected

and un-infected ZW13-2 neuronal cells. As shown in Fig. 6 enhanced EtBr uptake was

observed in both the 22L and 139A scrapie-infected cells compared with the control cells.

The scrapie infection-enhanced EtBr uptake was inhibited in the presence of LaCl3 (500

μM; a known connexin hemichannel blocker) and the JNK inhibitor SP600125, indicating

that scrapie infection mediated upregulation of Cx43 controls the functional state of

hemichannels.

The scrape-loading dye transfer technique is used to determine the intrinsic gap junction

intercellular communication (GJIC) [43, 49]. Next, scrapie-infected and un-infected

ZW13-2 neuronal cells were subjected to a scrape-loading dye transfer assay in the

absence or presence of LaCl3 and a JNK inhibitor as described in the Materials and

Methods. As shown in Fig. 7A (top panels), gap junction-mediated intercellular transfer

of LY was increased in the 22L and 139A scrapie-infected cells compared with the control

cells. This increase was abolished by either hemichannel inhibition (LaCl3) or JNK

inhibition (SP600125) (Fig. 7A, middle and bottom panels, respectively). Hemichannel

37
inhibition almost blocked GJIC in either control or infected cells (Fig. 7A middle panels),

whereas JNK inhibition maintained basal levels of GJIC in control and infected cells (Fig.

7A ottom panels) without a significant difference between control and infected cells. The

distances of LY diffusion were measured and compared with those in the controls and

infected cells with or without treatments (Fig. 7B). These results indicate that the

upregulation of Cx43 expression by scrapie infection represents the efficient

communication capacity of Cx43 hemichannels and GJIC and likely occurs via JNK

activation.

6) Suppression of Cx43 expression with decreased JNK activity by BBG-mediated

blockade of prion conversion

To further investigate whether Cx43 induction is regulated by PrPSc accumulation, we

conducted a blocking experiment of PrPSc accumulation using BBG, which has anti-prion

activity through the inhibition of PrPSc accumulation [50]. Un-infected and 22L scrapie-

infected ZW13-2 neuronal cells were incubated with various concentrations of BBG (0–

60 μM) for 3 days, and the expression levels of Cx43, PrPSc, and p-JNK were

subsequently measured. As shown in Fig. 8 BBG treatment efficiently inhibited PrPSc

accumulation in a dosedependent manner. In correlation with the decrease in PrPSc, the

expression levels of Cx43 and p-JNK were gradually reduced following BBG treatment

of 22L scrapie-infected cells. In contrast, no changes were observed in the uninfected

cells treated with BBG. These results suggest that PrPSc accumulation may be responsible

for JNK signaling-mediated Cx43 upregulation.

38
2.4 Discussion

In the present study, we demonstrated that the upregulation of Cx43 expression, which

controls functional properties of hemichannels, is mediated by JNK activation and

correlated with PrPSc accumulation in both in vivo and in vitro systems of prion diseases.

These findings represent the first demonstration of the link between the Cx43 protein and

prion pathogenesis.

Cx43 hemichannels contribute to cell death and are responsible for tissue damage [11,

13, 15, 18, 20, 33, 35, 51]. Several signaling molecules are linked to brain inflammation

and unwarranted cell death. It is conceivable that the excessive diffusion of

neurotransmitters or ions, such as glutamate, ATP, and Ca2+, from damaged cells to

adjacent normal cells via Cx43-promoted gap junctions may accelerate this

pathophysiological process [8–10]. Astrocytes and microglia release glutamate via gap

junctions [8, 52–54], which then induces neuronal cell death [54, 55].

Under various pathological conditions, astrocytes become reactive, triggering a long-

lasting process that comprises complex phenotypic changes [56]. Astrocyte activation, in

which there is both hypertrophy and proliferation, involves changes in the cellular

phenotype and in the expression of transporters, receptors and ion channels, which can be

deleterious for neurons and thus lead to neuronal cell death [57]. These changes can

explain how connexins can undergo both up- and downregulation depending on the type

of brain pathology, nature of the injury, time scale and distance from the lesioned area

[58, 59].

Continuous astrocyte-neuron interactions are required to maintain Cx30 expression and

increased Cx43 levels in astrocytes [60]. The expression level of Cx43 and its

39
hemichannel activity were significantly increased in scrapie-infected brains and in

cultured hippocampal cells, even though the neuronal loss associated with prion diseases

reduces the level of Cx43 in astrocytes associated with the lost neurons. In our study, it is

difficult to identify which cell type(s) is mainly stained for Cx43. However, it is possible

that astroglial Cx43 upregulation may be caused by scrapie infection-mediated

pathological changes since reactive astrocytes are one of the major pathological changes

associated with prion diseases. Using previously established hippocampal neuronal cell

lines [39], increased Cx43 was observed following inoculation with two independent

scrapie strains. Although Cx43 is primarily expressed in astrocytes [4, 60, 61] and

astrocyte gap junctions coupled with CNS cells [59], these neuronal cell lines are a useful

in vitro system to study the functional role of Cx43 in prion diseases because they express

endogenous Cx43. In addition, our data suggest that scrapie infection-enhanced Cx43

proteins may consistently exacerbate the pathogenesis of prion diseases despite the

limited involvement of functional Cx43 expression in the glial cells from this study.

According to previous findings, the increased production of reactive oxygen species and

proinflammatory cytokines, including IL-1α, IL-1β, and TNF-α, in the brains of scrapie-

infected mice can lead astrocytes to become reactive [62]. These astrocytes can be well-

coupled via gap junctions, leading to increases in hemichannel activity because of the

increased Cx43 mRNA and protein levels in scrapie infected brains. Consistent with this

phenomenon, Cx43 was associated with gap junctional plaques via distribution to

neighboring cells after scrapie infection (Fig. 4). Although increases in Cx43 and gap

junctional communication and their effects on neuronal death and inflammatory

responses have been linked to chronic and progressive neurodegenerative diseases such

40
as stroke, Alzheimer’s disease, Parkinson’s disease, and Huntington’s disease [57], it

remains a matter of debate whether Cx43 is neuroprotective or deleterious.

Accumulating evidence has indicated that connexins act as phosphoproteins via a shift

in their electrophoretic mobility or direct incorporation of 32P [63]. The JNK signaling

cascade is an important intracellular signaling pathway that regulates Cx43 [47]. Our data

demonstrated that upregulated Cx43 was a result of JNK activation and was blocked by

SP600125, a potent inhibitor of JNK. In this study, the assays used to measure Cx43

predominately identified a single band, and we were unable to distinguish the

phosphorylation status of Cx43. Because Cx43 can be phosphorylated by various kinases,

the activation of a specific kinase may not correlate with Cx43 phosphorylation in our

models. A recent study has suggested that astroglial Cx43 plays a protective role in

oxidative stress-induced cell death, which depends on the phosphorylation state of Cx43

[17]. In addition, the opening of hemichannels formed by pannexin 1, the mammalian

ortholog of the invertebrate gap junction protein innexin, has also been implicated in

neuronal death after ischemia [64]. Moreover, during the astrogliosis response observed

24 h after reperfusion, de novo synthesis of pannexin 2 occurs in hippocampal rat

astrocytes [65]. Thus, the functional role of Cx43 and pannexin in the pathogenesis of

prion diseases remains unresolved. In addition, previous reports have demonstrated that

JNK signaling activation is involved in the pathogenesis of scrapie-infected hamster

brains [66] and JNK signaling cascades are necessary for Cx43 expression [45–47]. JNK

regulates downstream gene expression through the activation or inactivation of various

transcriptional factors, such as c-Jun, c-fos, and SP1 [67]. We also demonstrated that the

activated JNK/c-Jun cascade is responsible for Cx43 expression in both in vitro and in

vivo models of prion disease. Surprisingly, the accumulation of pathogenic PrPSc appears

41
to occur upstream of JNK activation, which is then followed by increased Cx43

expression. Although PrP did not co-localize in Cx43 plaque formation, scrapie infection

increased Cx43 aggregates at the cell-cell contact areas. Therefore, PrPSc accumulation

may be responsible for the upregulation of Cx43 expression and gap junction formation.

Overall, for the first time, these results demonstrated that scrapie infection activates

JNK signaling cascades to enhance Cx43 and gap junctions in neuronal cells and in the

brain. Pathogenic PrPSc (or scrapie)-mediated upregulation of Cx43 through JNK

activation may exacerbate prion pathology, including neuronal death. Although blocking

gap junctions, which inhibit the diffusion of neurotoxic molecules, has been proposed as

a candidate therapy for various neurodegenerative diseases, it can also limit essential

physiological signals. Thus, this strategy has both neurotoxic and neuroprotective effects

on disease progression. A previous report demonstrated that a JNK inhibitor reduced

PrP106-126 peptide-induced neuronal apoptosis [68]. Therefore, we suggest that

blocking the gap junctions (functional Cx hemichannels) expressed by neurons,

astrocytes, and microglia in the region of prion propagation might represent a novel

strategy to reduce disease phenotypes in prion diseases.

42
Fig. 2.1 Increased expression of Cx43 protein and mRNA in the brains of scrapie

infected mice. A) Cx43 protein expression in whole brain and in dissected brains of the

control and ME7 (160 dpi), 22 L (140 dpi), and 139A (170 dpi) scrapie-infected mice was

evaluated by western blot with anti-Cx43 antibody. β-actin was used as a loading control.

B) The intensities of the Cx43 bands in panel A were measured and quantified. The values

are expressed as the mean±SEM (n = 3). C) In whole brains, the Cx43 mRNA levels were

43
analyzed by RT-PCR with three individuals per group. The error bars represent the SEM.

D) Cx43 protein expression was analyzed in the whole brains of the control and scrapie-

infected mice by western blot at the indicated time points after inoculation. β-actin was

used as a loading control, and PrPSc was detected using anti-PrP antibody after PK (50

μg/ml) treatment. Each experiment was repeated at least three times, and similar results

were obtained. ∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001.

44
Fig. 2.2 Immunohistochemical staining of Cx43 in brain sections from control and

scrapie-infected mice. Cx43 protein was detected in various brain sections obtained from

the control and ME7, 22 L and 139A scrapie-infected mice at 150 days post-inoculation.

Arrows, Cx43-positive cell-cell cotact or blood vessels. Scale bar, 25 μm.

45
Fig. 2.3 Increased Cx43 protein in membrane fractions of scrapie infected brains.

Whole brains were lysed, separated into cytosol and membrane fractions, and subjected

to western blot analysis as described in the Materials and Methods. Each fraction of the

brain homogenates was evaluated using anti-calnexin as a membrane marker and anti-

enolase as a cytosolic marker.

46
Fig. 2.4 Subcellular distribution of Cx43 and PrP in hippocampal cell lines with or

without scrapie infection. Double immunofluorescence staining was carried using anti-

Cx43 (red) and anti-PrP (green) in the PrP knockout (Zpl2-4) and wild-type (ZW13-2)

hippocampal neuronal cell lines with or without the 22 L or 139A scrapie infection. DAPI

was used for nuclear staining. Note that Cx43 was mainly localized in plaque formations

at the cell-cell contact areas (merged and bright-field images). Arrows indicate parts of

the gap junctions. Scale bars, 20μm.

47
Fig. 2.5 JNK activation-mediated Cx43 upregulation in scrapie-infected cells and

mice. A) Increased Cx43 expression and JNK activation in control and scrapie-infected

ZW13-2 cells (left panels) and mouse brains (right panels). To detect PrPSc, cell lysates

and brain homogenates were treated for 30 min at 37°C with PK at 50 μg/ml or 20 μg/ml,

respectively. B, C) JNK inhibition attenuates Cx43 expression in a dose- (B) and time-

dependent (C) manner. Control, 22L-, and 139A-infected ZW13-2 cells were treated with

0, 25, 50, and 100 μM of the JNK inhibitor SP600125 for 6 h (B) or with 50 μM

SP600125 for the time indicated (C). Each protein was analyzed by western blot with the

indicated antibodies.

48
Fig. 2.6 Increased EtBr uptake in scrapie-infected cells. An EtBr uptake assay was

performed using control or scrapie-infected ZW13-2 cells. Cells were pretreated with 500

μM LaCl3 for 5 min (middle panels) or 50 μM of the JNK inhibitor SP600125 for 6 h

(bottom panels) and then incubated with 5 μM EtBr for 5 min at 37°C; the fluorescent

signal was then analyzed using a confocal microscope.

49
50
Fig. 2.7 Increased scrape-loading dye transfer in scrapie-infected cells. Control or

scrapie-infected ZW13-2 cells were incubated with 0.1% Lucifer yellow (LY) for 5 min

at 37°C (top panels) or with 500 μM LaCl3 treatment (middle panels). For JNK inhibition,

the cells were pretreated with the JNK inhibitor SP600125 (50 μM) for 6 h prior to LY

treatment (bottom panels). LY fluorescence was determined using a confocal microscope

(A) and mean length of LY-filled area (the distance from the center to the edge, n = 20)

was represented by bars (B). The arrow indicates scrape; lines drawn in upper panels

indicate the distance for dye transfer. ∗∗∗p < 0.001. Scale bar, 50 μm.

51
Fig. 2.8 Attenuation of JNK signaling-mediated Cx43 upregulation by inhibiting the

accumulation of PrPSc. Control and 22 L scrapie-infected ZW13-2 cells were treated

with various concentrations of Brilliant blue G (BBG; 0-60 μM) for 3 days, and the

expression levels of Cx43, phosphorylated JNK, and PrPSc were analyzed by western blot

with the indicated antibodies. To detect PrPSc, the total cell lysates were treated with 50

μg/ml of PK for 30 min at 37°C. β-actin was used as a loading control.

52
2.5 References

[1] GoodenoughDA, Paul DL (2009) Gap junctions. Cold Spring Harb Perspect Biol 1,

a002576.

[2] Bruzzone R, Dermietzel R (2006) Structure and function of gap junctions in the

developing brain. Cell Tissue Res 326, 239-248.

[3] Elias LA, Kriegstein AR (2008) Gap junctions: Multifaceted regulators of embryonic

cortical development. Trends Neurosci 31, 243-250.

[4] Dermietzel R, Hertberg EL,Kessler JA, SprayDC(1991) Gap junctions between

cultured astrocytes: Immunocytochemical, molecular, and electrophysiological analysis.

J Neurosci 11, 1421-1432.

[5] Nagy JI, Dudek FE, Rash JE (2004) Update on connexins and gap junctions in neurons

and glia in the mammalian nervous system. Brain Res Brain Res Rev 47, 191-215.

[6] Nualart-Marti A, Solsona C, Fields RD (2013) Gap junction communication in

myelinating glia. Biochim Biophys Acta 1828, 69-78.

[7] Stout CE, Costantin JL, Naus CC, Charles AC (2002) Intercellular calcium signaling

in astrocytes via ATP release through connexin hemichannels. J Biol Chem 277, 10482-

10488.

[8] Ye ZC, Wyeth MS, Baltan-Tekkok S, Ransom BR (2003) Functional hemichannels in

astrocytes: A novel mechanism of glutamate release. J Neurosci 23, 3588-3596.

[9] Lurtz MM, Louis CF (2007) Intracellular calcium regulation of connexin43. Am J

Physiol Cell Physiol 293, C1806-C1813.

[10] Kang J, Kang N, Lovatt D, Torres A, Zhao Z, Lin J, Nedergaard M (2008) Connexin

43 hemichannels are permeable to ATP. J Neurosci 28, 4702-4711.

53
[11] Orellana JA, Froger N, Ezan P, Jiang JX, Bennett MV, Naus CC, Giaume C, Saez JC

(2011a) ATP and glutamate released via astroglial connexin 43 hemichannels mediate

neuronal death through activation of pannexin 1 hemichannels. J Neurochem 118, 826-

840.

[12] Orellana JA, Martinez AD, Retamal MA (2013) Gap junction channels and

hemichannels in the CNS: Regulation by signaling molecules. Neuropharmacology 75,

567-582.

[13] Orellana JA, Shoji KF, Abudara V, Ezan P, Amigou E, S´aez PJ, Jiang JX, Naus CC,

S´aez JC, Giaume C (2011b) Amyloid β-induced death in neurons involves glial and

neuronal hemichannels. J Neurosci 31, 4962-4977.

[14] Ishii Y, Shintani-Ishida K, Yoshida K (2013) Connexin-43 hemichannels contribute

to the propagation of μ-calpainmediated neuronal death in a cortical ablation injury model.

Biochem Biophys Res Commun 441, 457-462.

[15] Frantseva MV, Kokarovtseva L, Perez Velazquez JL (2002) Ischemia-induced brain

damage depends on specific gapjunctional coupling. J Cereb Blood Flow Metab 22, 453-

462.

[16] Giardina SF, Mikami M, Goubaeva F, Yang J (2007) Connexin 43 confers resistance

to hydrogen peroxide-mediated apoptosis. Biochem Biophys Res Commun 362, 747-752.

[17] Le HT, Sin WC, Lozinsky S, Bechberger J, Vega JL, Guo XQ, Saez JC, Naus CC

(2014) Gap junction intercellular communication mediated by connexin43 in astrocytes

is essential for their resistance to oxidative stress. J Biol Chem 289, 1345-1354.

[18] Cronin M, Anderson PN, Cook JE, Green CR, Becker DL (2008) Blocking

connexin43 expression reduces inflammation and improves functional recovery after

spinal cord injury. Mol Cell Neurosci 39, 152-160.

54
[19] Chen MJ, Kress B, Han X, Moll K, PengW, Ji RR, Nedergaard M (2012) Astrocytic

CX43 hemichannels and gap junctions play a crucial role in development of chronic

neuropathic pain following spinal cord injury. Glia 60, 1660-1670.

[20] Davidson JO, Green CR, Nicholson LF, O’Carroll SJ, Fraser M, Bennet L, Gunn AJ

(2012) Connexin hemichannel blockade improves outcomes in a model of fetal ischemia.

Ann Neurol 71, 121-132.

[21] Siushansian R, Bechberger JF, Cechetto DF, Hachinski VC, Naus CC (2001)

Connexin43 null mutation increases infarct size after stroke. J Comp Neurol 440, 387-

394.

[22] Kozoriz MG, Bechberger JF, Bechberger GR, Suen MW, Moreno AP,Maass

K,Willecke K, Naus CC (2010) The connexin43 C-terminal region mediates

neuroprotection during stroke. J Neuropathol Exp Neurol 69, 196-206.

[23] Santiago MF, Alcami P, Striedinger KM, Spray DC, Scemes E (2010) The carboxyl-

terminal domain of connexin43 is a negative modulator of neuronal differentiation. J Biol

Chem 285, 11836-11845.

[24] Lemcke H, Kuznetsov SA (2013) Involvement of connexin43 in the EGF/EGFR

signalling during self-renewal and differentiation of neural progenitor cells. Cell Signal

25, 2676-2684.

[25] Rinaldi F, Hartfield EM, Crompton LA, Badger JL, Glover CP, Kelly CM, Rosser

AE, Uney JB, Caldwell MA (2014) Cross-regulation of Connexin43 and β-catenin

influences differentiation of human neural progenitor cells. Cell Death Dis 5, e1017.

[26] Huang GY, Cooper ES, Waldo K, Kirby ML, Gilula NB, Lo CW (1998) Gap

junction-mediated cell-cell communication modulates mouse neural crest migration. J

Cell Biol 143, 1725-1734.

55
[27] Cina C, Maass K, Theis M, Willecke K, Bechberger JF, Naus CC (2009) Involvement

of the cytoplasmic C-terminal domain of connexin43 in neuronal migration. J Neurosci

29, 2009-2021.

[28] Behrens J, Kameritsch P,Wallner S, Pohl U, Pogoda K (2010) The carboxyl tail of

Cx43 augments p38 mediated cell migration in a gap junction-independent manner. Eur

J Cell Biol 89, 828-838.

[29] Liu X, Sun L, Torii M, Rakic P (2012) Connexin 43 controls the multipolar phase of

neuronal migration to the cerebral cortex. Proc Natl Acad Sci U S A 109, 8280-8285.

[30] Liebmann M, Stahr A, Guenther M, Witte OW, Frahm C (2013) Astrocytic Cx43 and

Cx30 differentially modulate adult neurogenesis in mice. Neurosci Lett 17, 40-45.

[31] Nagy JI, Li W, Hertzberg EL, Marotta CA (1996) Elevated connexin43

immunoreactivity at sites of amyloid plaques in Alzheimer’s disease. Brain Res 717, 178.

[32] MeiX, Ezan P, Giaume C,KoulakoffA(2010) Astroglial connexin immunoreactivity

is specifically altered at β-amyloid plaques in β-amyloid precursor protein/presenilin1

mice. Neuroscience 171, 92-105.

[33] Cui Y, Masaki K, Yamasaki R, Imamura S, Suzuki SO, Hayashi S, Sato S, Nagara Y,

Kawamura MF, Kira J (2014) Extensive dysregulations of oligodendrocytic and astrocytic

connexins are associated with disease progression in an amyotrophic lateral sclerosis

mouse model. J Neuroinflammation 11, 42.

[34] Rufer M, Wirth SB, Hofer A, Dermietzel R, Pastor A, Kettenmann H, Unsicker K

(1996) Regulation of connexin- 43, GFAP, and FGF-2 is not accompanied by changes in

astroglial coupling in MPTP-lesioned, FGF-2-treated parkinsonian mice. J Neurosci Res

46, 606-617.

[35] Fonseca CG, Green CR, Nicholson LF (2002) Upregulation in astrocytic connexin

56
43 gap junction levels may exacerbate generalized seizures in mesial temporal lobe

epilepsy. Brain Res 929, 105-116.

[36] Masaki K, Suzuki SO, Matsushita T, Matsuoka T, Imamura S, Yamasaki R, Suzuki

M, Suenaga T, Iwaki T, Kira J (2013) Connexin 43 astrocytopathy linked to rapidly

progressive multiple sclerosis and neuromyelitis optica. PLoS One 8, e72919.

[37] Prusiner SB (1998) Prions. Proc Natl Acad Sci U S A 95, 13363-13383.

[38] Choi JK, Park SJ, Jun YC,OhJM, Jeong BH, LeeHP, Park SN, Carp RI, Kim YS

(2006) Generation of monoclonal antibody recognized by the GXXXG motif (glycine

zipper) of prion protein. Hybridoma (Larchmt) 25, 271-277.

[39] Kim BH, Kim JI, Choi EK, Carp RI, Kim YS (2005) A neuronal cell line that does

not express either prion or doppel proteins. Neuroreport 16, 465-429.

[40] Harrison CF, Lawson VA, Coleman BM, Kim YS, Masters CL, Cappai R, Barnham

KJ, Hill AF (2010) Conservation of a glycine-rich region in the prion protein is required

for uptake of prion infectivity. J Biol Chem 285, 20213-20223.

[41] Jang B, Jin JK, Jeon YC, Cho HJ, Ishigami A, Choi KC, Carp RI, Maruyama N, Kim

YS, Choi EK (2010) Involvement of peptidylarginine deiminase-mediated post-

translational citrullination in pathogenesis of sporadic Creutzfeldt-Jakob disease. Acta

Neuropathol 119, 199-210.

[42] Orellana JA, Hern´andez DE, Ezan P, Velarde V, Bennett MV, Giaume C, S´aez JC

(2010) Hypoxia in high glucose followed by reoxygenation in normal glucose reduces the

viability of cortical astrocytes through increased permeability of connexin 43

hemichannels. Glia 58, 329-343.

[43] Krutovskikh VA, Troyanovsky SM, Piccoli C, Tsuda H, Asamoto M, Yamasaki H

(2000) Differential effect of subcellular localization of communication impairing gap

57
junction protein connexin43 on tumor cell growth in vivo. Oncogene 19, 505-513.

[44] Bruce ME, McConnell I, Fraser H, Dickinson AG (1991) The disease characteristics

of different strains of scrapie in Sinc congenic mouse lines: Implications for the nature of

the agent and host control of pathogenesis. J Gen Virol 72, 595-603.

[45] Shyu KG, Wang BW, Yang YH, Tsai SC, Lin S, Lee CC (2004) Amphetamine

activates connexin43 gene expression in cultured neonatal rat cardiomyocytes through

JNK and AP-1 pathway. Cardiovasc Res 63, 98-108.

[46] Gao FH,Wang Q,Wu YL, Li X, ZhaoKW, ChenGQ(2007) c- Jun N terminal kinase

mediates AML1-ETO protein-induced connexin-43 expression. Biochem Biophys Res

Commun 356, 505-511.

[47] Li K, ChiY, Gao K,YanQ,Matsue H, Takeda M, Kitamura M, Yao J (2013)

Connexin43 hemichannel-mediated regulation of connexin43. PLoS One 8, e58057.

[48] Contreras JE, S´aez JC, Bukauskas FF, Bennett MV (2003) Gating and regulation of

connexin 43 (Cx43) hemichannels. Proc Natl Acad Sci U S A 100, 11388-11393.

[49] McKarns SC, Doolittle DJ (1992) Limitations of the scrapeloading/ dye transfer

technique to quantify inhibition of gap junctional intercellular communication. Cell Biol

Toxicol 8, 89-103.

[50] Iwamaru Y, Takenouchi T, Murayama Y, Okada H, Imamura M, Shimizu Y,

Hashimoto M, Mohri S, Yokoyama T, Kitani H (2012) Anti-prion activity of Brilliant

Blue G. PLoS One 7, e37896.

[51] Frantseva MV, Kokarovtseva L, Naus CG, Carlen PL, Mac- Fabe D, Perez Velazquez

JL (2002b) Specific gap junctions enhance the neuronal vulnerability to brain traumatic

injury. J Neurosci 22, 644-653.

[52] Parpura V, Scemes E, Spray DC (2004) Mechanisms of glutamate release from

58
astrocytes: Gap junction “hemichannels”, purinergic receptors and exocytotic release.

Neurochem Int 45, 259-264.

[53] Takeuchi H, Jin S, Wang J, Zhang G, Kawanokuchi J, Kuno R, Sonobe Y, Mizuno T,

Suzumura A (2006) Tumor necrosis actor-alpha induces neurotoxicity via glutamate

release from hemichannels of activated microglia in an autocrine manner. J Biol Chem

281, 21362-21368.

[54] Takeuchi H, Jin S, Suzuki H, Doi Y, Liang J, Kawanokuchi J, Mizuno T, Sawada M,

Suzumura A (2008) Blockade of microglial glutamate release protects against ischemic

brain injury. Exp Neurol 214, 144-146.

[55] Ridet JL, Malhotra SK, Privat A, Gage FH (1997) Reactive astrocytes: Cellular and

molecular cues to biological function. Trends Neurosci 20, 570-577.

[56] Yawata I, Takeuchi H, Doi Y, Liang J, Mizuno T, Suzumura A (2008) Macrophage-

induced neurotoxicity is mediated by glutamate and attenuated by glutaminase inhibitors

and gap junction inhibitors. Life Sci 82, 1111-1116.

[57] Orellana JA, S´aez PJ, Shoji KF, Schalper KA, Palacios-Prado N, Velarde V, Giaume

C, Bennett MV, S´aez JC (2009) Modulation of brain hemichannels and gap junction

channels by pro-inflammatory agents and their possible role in neurodegeneration.

Antioxid Redox Signal 11, 369-399.

[58] Rouach N, Avignone E, Mˆeme W, Koulakoff A, Venance L, Blomstrand F, Giaume

C (2002) Gap junctions and connexin expression in the normal and pathological central

nervous system. Biol Cell 94, 457-475.

[59] Giaume C, Theis M (2010) Pharmacological and genetic approaches to study

connexin-mediated channels in glial cells of the central nervous system. Brain Res Rev

63, 160-176.

59
[60] Koulakoff A, Ezan P, Giaume C (2008) Neurons control the expression of connexin

30 and connexin 43 in mouse cortical astrocytes. Glia 56, 1299-1311.

[61] Dermietzel R, Traub O, Hwang TK, Beyer E, Bennett MV, Spray DC,Willecke K

(1989) Differential expression of three gap junction proteins in developing and mature

brain tissues. Proc Natl Acad Sci U S A 86, 10148-10152.

[62] Kim JI, Ju WK, Choi JH, Choi E, Carp RI, Wisniewski HM, Kim YS (1999)

Expression of cytokine genes and increased nuclear factor-kappa B activity in the brains

of scrapieinfected mice. Brain Res Mol Brain Res 73, 17-27.

[63] Lampe PD, Lau AF (2004) The effects of connexin phosphorylation on gap

junctional communication. Int J Biochem Cell Biol 36, 1171-1186.

[64] Thompson RJ, Zhou N, MacVicar BA (2006) Ischemia opens neuronal gap junction

hemichannels. Science 312, 924-927.

[65] Zappal`a A, Li Volti G, Serapide MF, Pellitteri R, Falchi M, La Delia F, Cicirata V,

Cicirata F (2007) Expression of pannexin2 protein in healthy and ischemized brain of

adult rats. Neuroscience 148, 653-667.

[66] Lee HP, Jun YC, Choi JK, Kim JI, Carp RI, Kim YS (2005) Activation of mitogen-

activated protein kinases in hamster brains infected with 263K scrapie agent. J

Neurochem 95, 584-593.

[67] Teunissen BE, Jansen AT, van Amersfoorth SC, O’Brien TX, Jongsma HJ,

Bierhuizen MF (2003) Analysis of the rat connexin 43 proximal promoter in neonatal

cardiomyocytes. Gene 322, 123-136.

[68] Carimalo J, Cronier S, Petit G, Peyrin JM, Boukhtouche F, Arbez N, Lemaigre-

Dubreuil Y, Brugg B, Miquel MC (2005) Activation of the JNK-c-Jun pathway during

the early phase of neuronal apoptosis induced by PrP106-126 and prion infection. Eur J

60
Neurosci 21, 2311-2319.

61
III. Chapter 3

Establishment of Advanced In Vitro Model from Bank Vole

Transgenic Mice for Susceptibility to Various Prion Strains

Abstract

Prions are infectious agents that cause a devastating neurodegenerative disease in both
humans and animals. Unlike other rodents, bank vole (Myodes glareolus, BV) is
susceptible to prions from a diverse range of species, including humans. There are two
lines of BV, one homozygous for methionine (BV109M) and the other for isoleucine at
codon 109 (BV109I) of prion protein (PrP). Here, we established neuronal cell lines from
embryonic brain of BV109I mouse that was immortalized by an introduction of plasmid
DNA encoding for SV40-T antigen. BV109I mice develop age-dependent signs of
spontaneous neurologic illness. The established BV cell lines infected with 22L scrapie
strain (BV-22L) showed a replication and an accumulation of disease-associated forms
of the PrP. BV-22L infected neuronal cells were then intracerebrally injected into ICR
mice showing clinical symptoms of disease after 147 days post-inoculation. We also
confirmed that various prion strains, including ME7, 263K, CWD and 22L, were
successfully infected into BV neuronal cells. The newly established BV cell models may
enhance our understanding on the cellular mechanisms of prion replication and can be a
useful tool for the study of the pathogenic mechanisms of prion disease.

Key words: bank vole, neuronal cell lines, prion disease, scrapie

62
3.1. Introduction

Prions are infectious agents that cause a devastating neurodegenerative disease in both

humans and animals. The prion-associated diseases including transmissible spongiform

encephalopathies (TSE) or CJD in human were characterized as “infectivity” which are

distinguish from other neurodegenerative disease such as Alzheimer’s and Parkinson’s

disease; this property seems to reside, at least in part, in an aberrant scrapie form of prion

protein (PrPSc) of a constitutively expressed cellular prion protein (PrPC) [1]. It has been

suggested that the conformer protein, PrPC interacted with abnormal prion protein, PrPSc

and then generated newly formed PrPSc through undefined process, including post-

translational modification, combination with factor X, and mutations and so on (Prusiner,

1998). PrPC is constitutively expressed in most cells, including neuronal cells, in particular

with highest expression level in subset of neurons of central nervous system (CNS). Despite

the distribution of PrPC in various tissues, there are still many arguing on the cellular

functions of PrPC [2, 3, 4].

Scrapie is one of the prion diseases developed in sheep and goat, and its clinical

symptoms characterized by a long latent period, progressive ataxia, tremor, wasting and

death [5]. Due to the property of long latent period, scrapie is regarded as a slow infectious

disease. It is successfully transmitted to several species of rodents, including mice,

hamsters and rats. Many kinds of scrapie strains were isolated and used to study

pathogenesis of prion diseases [6, 7]. The typical features of the prion diseases are

histopathological spongiform degeneration in CNS, reactive gliosis or astrocytosis and

neuronal loss observed in some cases, and formation of amyloid plaques. It has been

reported that a common cause of prion diseases including scrapie is the conformational

63
misfolding of the endogenously expressed prion protein [8]. A predominant α-helical-rich

prion protein PrPC have changed into a disease-associated β-sheet-rich conformation in

terms PrPSc [9], but the mechanism is still unknown. PrPSc has two different biochemical

features compared to PrPC which is ability to formation of amyloid fibrils and relatively

resistant to proteolysis with proteinase K (PK). These features are used to diagnostically

distinguish PrPSc from PrPC. The mice presented clinical signs of scrapie agent has shown

the microglial activation in the regions of brain showing vacuolation, PrP deposition and

neuronal apoptosis. [10, 11]. Under the same condition for proteolysis with PK, PrPSc

produces a smaller protease-resistant molecule of 142 amino acids whereas PrPC is

completely hydrolyzed [12]. Prion rods formed by limited proteolysis, and detergent

extraction are indistinguishable from the filaments that aggregate to form PrP amyloid

plaques in the CNS [13].

Study for transmission barrier needs to elucidate the basis of prion replication and

acquiring knowledge to decipher the risk of interspecies transmission [14]. For the study

of transmission activity between species barrier, the animal models that are susceptible to

various prion strains are quite available. Recently bank voles (Myodes glareouls, BV)

were introduced as an animal model for the research of prion diseases. This animal model

has an important difference compared to a model of mouse and Syrian hamster. A

transgenic (Tg) mice expressing BVPrP showed a high susceptibility to prion strains

originated from various species, including sheep, goats and humans [15]. In addition, BV

mice shows a relatively short incubation period compared to a conventional prion-animal

model [16]. Two lines of BV mouse that is homozygous phenotype with either methionine

or isoleucine at codon 109 on BV Prnp and in termed as Tg mouse BV109M and BV109I

were investigated, respectively [17]. BV109M was shown to be susceptible to sporadic

64
and genetic Creutzfeldt-Jakob disease (CJD) [18], sheep scrapie [19], mouse- and

hamster-adapted scrapie strains [20], cattle and sheep BSE [18] and atypical BSE [14].

The studies with BV109I was revealed that the susceptibility of PrP109I phenotype is

similar to the methionine-carrying line, although the differences depends on the specific

prion strains [21].

In this study we investigated whether BVPrP-expressing neuronal cell lines (BV cells)

are susceptible to a wide range of prion sources. Taking advantage of prion strains adapted

BV cell lines, this study aimed to investigate which prion strains are actually associated

with distinctive PrPSc types using this biochemical phenotypes. Firstly we established the

neuronal cell line derived from BV109I mouse and then assessed the susceptibility of the

cells to 22L scrapie strains. We examined the characterization of 22L strains-adapted BV

neuronal cells, resulting in observed high localization titers of PrPSc accumulation in

cytoplasm.

These wild-type BV, BV-/- and 22L-adapted BV neuronal cell lines will be useful

experimental systems to reveal the physiological functions of both PrPC and PrPSc, and to

investigate the conformational change of prion isoforms and the pathological mechanisms

in prion diseases.

65
3.2. Materials and Methods

1) Animals and prion strains

The prion strains, 22L, ME7, and 263K were kindly provided by Alan Dickinson

(Neuropathogenesis Unit, UK): 22L prion strain was maintained by serial intracerebral

passage of brain homogenate from terminally affected mice. Six-week-old male ICR mice

were obtained from the Central Lab. Animal Inc. (Republic of Korea) and the inbred ICR

mice divided into age-matched controls and experimental group infected with 22L prion

strain. CWD prion strain was passaged to the Tg PrPElk mice [22]. (Jeon et al., 2013).

Mice were inoculated intracerebrally with 30 μl of 1 % (w/v) brain homogenates in 0.01

M phosphate-buffered saline (pH 7.4) from either normal brain or prion-infected ICR

mouse brain at the terminal stages of the disease. Animals were sacrificed when clinical

signs of the disease were manifested and then the brain of both control and prion-infected

mice was harvested for protein analysis and histological pathology of prion disease. All

animal experiment has been conducted in accordance with the guidelines laid down by

the Hallym Medical Center for the Institutional Animal Care and Use Committee.

2) Establishment of neuronal cell lines from BV109I mice

Primary neuronal cells were isolated from 14-days-old embryos of BVPrP-expressing

Tg mice (BV109I). This BV109I mice produced from PrP knockout mouse were kindly

provided by Dr. Joaquin Castilla (Proteomics Unit, Spain). The cells were cultured on

poly-L-lysine (P-L-L) (Sigma-Aldrich, USA) coated dishes with culture media DMEM

(Hyclone, USA) containing 20% FBS, 100 unit/ml of penicillin and 100 unit/ml of

streptomycin (Gibco BRL, USA) at 37℃, 5% CO2. After one week, medium was replaced

66
by selective medium (DMEM with 0.25 mg/ml G418, Hyclone), and changed in every 3

days. When a single colony formed, it was transferred to 24-well plate dish and examined.

At last the cells from a single colony were transferred to 6-well dishes and maintained in

culture medium, DMEM with 10% FBS and 1% antibiotics.

3) Scrapie-infected cell lines

The cells including BV and mouse hippocampal neuronal ZW13-2 (PrP wild-type) [23]

were cultured in DMEM with 10% FBS (Hyclone), 100 units/ml of penicillin and 100

µg/ml of streptomycin in incubator. The BV cells were persistently infected with 22L

prion strain as previously described [24]. The infected cells maintained in Opti-MEM

(Gibco BRL) with 10% FCS (Hyclone). Stable scrapie infection was confirmed after 10

passages by a cell blot assay as previously described [24]. The infected cells stably

produced PrPSc over 50 passages.

4) Reverse transcription-polymerase chain reaction (RT-PCR)

To assess the expression levels of SV40 and PrP, total RNA was extracted using Trizol

reagent (Invitrogen), and synthesized cDNA by AMV transcriptase (Promega, USA). To

determine the integration of SV40 large T antigen, genomic DNA was extracted from the

primary cultured cells using DNA extraction kit (Qiagen, USA) according to the

Manufacture’s instruction. PCR was performed with the following primers (Bioneer,

Republic of Korea).

PrP,

sense: 5’ ATGGCGAACCTTGGCTACTGG-3’

antisense: 5’-CCCTCATCCCACGATCAGGAAGATG-3’

67
SV40 large T antigen,

sense: 5’-TGAGGCTACTGCTGACTCTCAACA-3’

antisense: 5’-GCATGACTCAAAAAACTTAGCAATTCTG-3’

GAPDH,

sense: 5’-TGGTATCGTGGAAGGACTCATGAC-3’

antisense: 5’-ATGCCAGTGAGCTTCCCGTTCAGC-3’ (reference 2004??).

The obtained PCR products were examined by electrophoresis with 1 % TBE-agarose gel.

5) Western blot analysis.

The mouse brains and neuronal cells were lysed in 10 volumes of RIPA buffer (Thermo

Fisher Scientific, USA) containing protease inhibitor cocktails (Roche, Germany). The

brain homogenates and cell lysates were centrifuged at 10,000 g for 15 min at 4℃ and

then the supernatant were taken. The protein concentration was determined by BCA assay

(Pierce, USA). To detect PrPSc, 5 to 50 μg/ml of proteinase K (PK) and 25 to 100 μg/ml

of thermolysin (TL) was treated for 30 min at 37°C in brain homogenates and cell lysates.

Equal amounts of proteins were boiled with 5X SDS Gel-loading buffer [100mM Tris-

HCl (pH 6.8), 4% SDS, 0.2% bromophenol blue, 20% glycerol, and 10% -

mercaptoethanol] for 10 min, separated by 15% SDS-gel. The separated proteins were

transferred to nitrocellulose membrane (Amersham, USA) at 260 mA for 1.5 hr. After

blocking with 5% skim milk in TBST containing 0.05% for 1 hr, the membranes were

incubated with either one of following antibodies at 4°C for overnight : mouse anti-PrP

antibody (3F10; 1:3,000) [25], mouse monoclonal anti-NeuN antibody (1:5,000,

Chemicon, USA) as a neuronal marker and mouse monoclonal anti-GFAP (1:3,000,

68
Chemicon) as an astrocyte marker. Incubation with appropriate secondary antibodies

conjugated with horseradish peroxidase (Sigma-Aldrich) carried out for 1 hr at room

temperature. The membranes were visualized by chemiluminescence substrate as

described in Manufacture’s instruction. (Amersham). The target signals were visualized

by digital images and captured with an ImageQuant™ LAS 4000 imager (GE Healthcare

Life Sciences, USA) using an enhanced chemiluminescence Western blot detection

system (Amersham). Western blot analysis (n=3 for each group) was performed at least

three times.

69
3.3. Result

1) Neuronal cell lines established from BV109I mice

Primary neuronal cells were isolated from the cortical region of 14-days-old embryos of

BV109I mice and transfected with retroviruses expressing SV40 large-T-antigen to be

immortalized. The cells were cultured in selective media until the establishment of cell

lines as described in Materials and Methods, and four distinct neuronal cell lines were

established: BV5, BV6, BV7, and BV8. As shown in Fig. 1A, all of cell lines were

characterized by RT-PCR with a SV40 large T antigen-specific primers showing similar

expression levels. To further confirm the cell type of each cell lines, Western blot and

immunofluorescence analyses were carried out using different cell markers. In both

experiments, four cells were positively reacted with a neuronal cell marker, NeuN, but

not with an astroglial cell marker, GFAP. Characterization of the cell type yielded similar

outcome in both Western blot (Fig 1B) and immunofluorescence analysis (data not

shown). For the positive controls, brain tissues of BV109I mice and ZW13-2 cell line

were examined.

To examine the expression levels of BVPrP mRNA and protein in both BV neuronal cell

lines and the brain of BV109I mice, we performed RT-PCR and Western blot analysis,

respectively. The expression of prnp gene was detected in BV5 and BV 6 cell lines, but

not in BV 7 and BV 8 (Fig. 1C). In consistent with the mRNA expression level of BVPrP,

the BVPrP protein was strongly detected as three typical glycoforms in BV5 and BV6

cell lines, however BV7 and BV8 cell lines showed no detectable BVPrP. Taken together,

these newly established cells originated from BV109I mice were characterized as

neuronal cell lines, which can be used as a model of BVPrP-expressing (BV5 and BV6)

70
and BVPrP-deficient (BV7 and BV8) neuronal cells.

2) BV neuronal cells are susceptible to infection with different prion strains

Unlike other rodents, BVs are highly susceptible to prions from many different species,

suggesting that BVs do not impose a species barrier, which normally restricts the

transmission of prions from one species to another [26]. We intracerebrally inoculated

various prion strains into mouse or hamster to use as prion inoculums, which lastly were

used to establish prion-infected BV neuronal cell lines. When the established BV neuronal

cells expresses PrPC, we assessed their susceptibility to various prion infection. As shown

in Fig. 2, the PK-resistant PrP were detected up to 30 passages in various prion-infected

BV cells, and 22L-infected BV6 cells (Bv-22L) consistently produced stronger

expression of PrPSc than ME7-, 263K- or CWD-infected BV cells (BV-ME7, BV-263K,

or BV-CWD, respectively) (Fig 2A). The levels of PrPSc in BV-22L cells were compared

with the PrPSc in the brain of 22L-infected ICR mice (Fig. 2B). The detected PrP protein

by anti-PrP antibody (3F10) has shown the typical migration pattern of PrPC towards

PrPSc under the PK treatment in different prion-infected BV neuronal cells. We next

confirmed the BV cells-adapted prion strains by the detection of thermolysin (TL)-

resistant PrPSc. As shown in Fig. 2C, the TL-resistant prion isoforms were detected in the

brain of 22L-infected ICR mouse and BV cells following TL treatment, whereas normal

prion isoforms in the BV6 cell line inoculated with the wild-type ICR mouse brain

homogenates was completely degraded by TL treatment. Taken together, our data indicate

that BV neuronal cells are a new, highly susceptible model of prion strain infection.

71
3) Transmission of BV-22L neuronal cells to ICR mice.

Intracerebral inoculation of ICR mice with cell lysates from 22L-infected ZW or BV

neuronal cells resulted in prion disease with clearly evident, progressive clinical signs. In

this studies on two independent cell-passaged scrapie strains, ZW-22L and BV-22L,

showed a different survival rates at first passage, however the survival rate became the

similar in second passage with a scrapie strain (Table 1). The incubation period at first

passage in mice was 149.2 ± 6.14 days (ZW-22L) and 185.3 ± 5.76 days (BV-22L)

decreasing to 145.5 ± 2.7 days (ZW-22L) and 147.8 ± 4 days (BV-22L), respectively, at

the second passages.

To further determine the accumulation of PK-resistant PrPSc in the brains of ZW-22L

and BV-22L inoculated mice at the end stage of the disease, we performed

immunoblotting using whole brains obtained from control and ZW-22L or BV-22L

inoculated mice. As shown in Fig. 3, we found no difference the accumulation of PK-

resistant PrPSc between the brains of ZW-22L and BV-22L inoculated mice (Fig. 3A)

although the total PrP proteins were slightly increased in infected brains. The PrPC in

control mice was completely degraded by the PK treatment at the end stage. Consistent

with these results, the levels and the rate of PK-resistant PrPSc accumulation in the brain

homogenates of second passaged ZW-22L and BV-22L mice were very similar. Taken

together, our data demonstrated that BV neuronal cells are a new, highly susceptible

model of various prion strains infection.

72
1.4. Discussion

In this study we hypothesized that PrPC protein expressing in BV in termed BVPrP mice

may improves the sensitivity of cells to various prion agents. To address this, we

established the neuronal cell lines expressing BVPrP and subjected the cells to in vivo

transmission of mouse-adapted prion strains. It is first time to establish the neuronal cells

originated from BV Tg mice. The characteristics of BV cells infected with 22L scrapie

were similar to scrapie-infected ZW13-2 cells which showed a high level of PK-resistant

PrPSc and the ability to induce scrapie in mice upon intracerebral inoculation. In vivo and

in vitro transmission studies with transgenic mice have clearly demonstrated an inverse

correlation between the expression level of PrPC and the incubation time of experimental

prion diseases.

It has been reported that several cell systems permissive to the replication of different

mouse-adapted prion strains have been developed [27]. They could be a suitable tool for

defining the mechanisms underlying prion-induced brain damage; their simplicity, in fact,

allows analyzing the events during prion pathogenesis better than in vivo models. Most

PrPC expressing cell lines are refractory to mammalian prion infection with unknown

reasons [28]. Prion strains have an exquisite host cell tropism in vivo [29] as well as in

tissue cultured cells [30].

Prions that are fully infectious between individuals of the identical mammalian or yeast

species may transmit poorly or not at all across species, which is called the species barrier.

It has been widely accepted that the species barrier may be largely due to the difference

of the sequence and conformation of PrP protein [31]. This difference from the variants

of PrP sequence has shown in the studies using Tg mice [32]. The characterization of Tg

73
mice expressing wild-type BVPrP showed remarkable finding (Watts et al., 2012).

Different from mice, BVs are highly susceptible to a wide range of prion strains isolated

from numerous species, including humans (Agrimi et al., 2008). Tg mice expressing the

109I polymorphic variant of BVPrP develop a spontaneous neurodegenerative disease

that recapitulates all of the neuropathological hallmarks of prion disease [26]. This model

was used as the shortest incubation time model for prion diseases because of its

outstanding susceptibility to propagate most of the existing prion strains from different

species. In a previous study, we established PrP hippocampal neuronal cell lines including

ZW 13-2 and showed very limited data that ZW 13-2 cell lines could be infected with the

22L and 139A strains of scrapie [23, 24]. Moreover, since only mouse prions were

produced in the cell lines by inoculating mouse prions, we demonstrated that prions could

be replicated and their biological characteristics were retained in the scrapie-infected

neuronal cell lines. This study showed the prion adaptation of the neuronal cell line

derived from the embryonic brain of BV109I mouse. We found that the scrapie-infected

cell line produced three major protease-resistant and PrP-immunoreactive prion proteins

with apparent molecular masses ranging. It showed that brain homogenate from 22L-

infected mice contained similar levels of total PrP and PK-resistant PrPSc. BV-22L

infected neuronal cells were intracerebrally injected into ICR mice, resulting in develop

the clinical symptoms of disease on incubation period averaged at 147 days dpi. The

incubation period was different between first and second passage in case of BV-22L

scrapie infected ICR mice, which presented on average of 185 days (BV-22L) at first

passage and of 147 days (BV-22L) at second passage, resulting in getting shorter

approximately 30 days in second passage than first passage.

Other alternatives of animal bioassay include both cell-based systems and cell-free

74
methodologies. Although cell lines are not generally suitable for prion infection or

replication, various cell lines, most notably N2a cells, have been used to study prions in

vitro. The cell line used for prion infection is not permissive for the infection of all prion

strains, i.e. N2a cell is resistant to strain ME7, 22A, and 301V, but susceptible to RML

and 22L [31].

In a current study, the various prion strains, including ME7, 263K, CWD, and 22L, were

successfully infected into BV neuronal cells and confirmed the PK-resistant PrPSc by

Western blot analysis. These results are consistent with the results described in Prusiner

et al. [31]. However, PrPSc produced in ME7, 263K, or CWD-infected BV neuronal cells

disappeared after over 50 passages although we determined that PrPSc was detectable in

up to 30 passages. There are many assumptions; the formation of PrP Sc in acute

conversion kills the host cells and the difference in PrP allotype between the host and

inoculum [20] and so on.

It is clear that different prion strains-adapted BV cells, which can be a target for prion

infection in vivo, are established in this study for the first time. The establishment of BV

cell models may enhance our understanding on the cellular mechanisms of prion

replication. From a viewpoint of the applied research and public health, prion-adapted

BV109I cell system will be a very useful model for the screening of prion types and

therapeutic compounds, and for the development of new diagnostic marker that are

urgently needed.

75
Figure 3.1. Cell-type characterization and expressions of the PrP in the BV neuronal

cell lines. (A) Expression levels of SV40 large T antigen mRNA were determined by RT-

PCR analysis. Tails from PrP-expressing wild-type or PrP-knockout mice were used as a

negative control for SV40 T. GAPDH was used as a loading control. (B) The BV5, BV6,

BV7 and BV8 cell lines were positive to NeuN which did react with protein lysates of

ZW13-2 neuronal cells as a positive control. In contrast, GFAP (a ~50 kDa protein band,

an astrocyte marker) was detected only in the brain homogenate of BV109I mouse. (C)

mRNA expressions of PrP (789 bp) was checked. Genomic DNA from the tails of PrP-

expressing BV109I or PrP-knockout mouse were used as a positive or negative control,

76
respectively. BV5 and BV6; Expression of BV109I neuronal cell lines. BV7 and BV8;

PrP knockout neuronal cell lines. (D) The expression of PrPC protein in BV neuronal cell

lines were analyzed by immunoblotting with anti-PrP antibody (3F10).

77
Figure 3.2. Analysis of PK and TL-resistant PrPSc in the BV neuronal cell lines

infected with different prion strains. Various prion-infected cells were used in the

Western blot. (A) and (B) The PK-digested PrPSc isoforms were detected using anti-3F10

PrP antibody. A portion of each sample was treated with PK (PK 5 or 10 μg/ml). (C) 22L-

infected mouse and BV6 cells following digestion with thermolysin (TL, 25 through 100

μg/ml).

78
Table 1. Transmission of 22L scrapie-infected BV neuronal cell lines to ICR mice

Figure 3.3. Accumulation of PK-resistant prion isoforms in ZW-22L and BV-22L

scrapie-infected ICR mice. PK-resistant PrPSc isoforms were identified in the brains of

ICR mice that were intracerebrally inoculated with ZW-22L or BV-22L-infected cell

lysates. (A) Whole brains of control (n = 4), first-infected mice (n = 4) and (B) second-

infected mice (n = 3) at the end stage were homogenized and immunoblotted with anti-

79
3F10 PrP antibody. To detect PK-resistant PrPSc, the total cell lysates were treated with

50 ug/ml of PK for 30 min at 37°C.

80
1.5. References

[1] Collins SJ, Lawson VA and Masters CL, Transmissible spongiform encephalopathies.

Lancet, 363:51–61, 2004.

[2] Shyng SL, Huber MT and Harris DA, A prion protein cycles between the cell surface

and an endocytic compartment in cultured neuroblastoma cells. J Biol Chem, 268:15922–

15928, 1993.

[3] Satoh J, Kuroda Y and Katamine S, Gene expression profile in prion protein-deficient

fibroblasts in culture. Am J Pathol, 157:59–68, 2000.

[4] Collinge J, Whittington MA, Sidle KC, Smith CJ, Palmer MS, Clarke AR and Jeffreys

JG, Prion protein is necessary for normal synaptic function. Nature, 370:295–297, 1994.

[5] Kimberlin RH, Walker C, Characteristics of a short incubation model of scrapie in the

golden hamster. J Gen Virol, 34(2):295-304, 1977.

[6] Chandler RL, Encephalopathy in mice produced by inoculation with scrapie brain

material. Lancet, 1(7191):1378-9, 1961.

[7] Chandler RL, Turfrey BA, Inoculation of voles, Chinese hamsters, gerbils and guinea-

pigs with scrapie brain material. Res Vet Sci, 13(3):219-24, 1972.

[8] Bolton DC, McKinley MP, Prusiner SB, Identification of a protein that purifies with

the scrapie prion. Science, 218(4579):1309-11, 1982.

[9] Pan KM, Baldwin M, Nguyen J, Gasset M, Serban A, Groth D, Mehlhorn I, Huang Z,

Fletterick RJ, Cohen FE, Conversion of alpha-helices into beta-sheets features in the

formation of the scrapie prion proteins. Proc Natl Acad Sci U S A, 90(23):10962-6, 1993.

[10] Lucassen PJ, Williams A, Chung WC, Fraser H., Detection of apoptosis in murine

scrapie. Neurosci Lett, 198(3):185-8, 1995.

81
[11] Williams AE, Lawson LJ, Perry VH, Fraser H, Characterization of the microglial

response in murine scrapie. Neuropathol Appl Neurobiol, 20(1):47-55, 1994.

[12] McKinley MP, Meyer RK, Kenaga L, Rahbar F, Cotter R, Serban A, Prusiner SB,

Scrapie prion rod formation in vitro requires both detergent extraction and limited

proteolysis. J Virol, 65(3):1340-51, 1991.

[13] Prusiner SB, McKinley MP, Bowman KA, Bolton DC, Bendheim PE, Groth DF,

Glenner GG, Scrapie prions aggregate to form amyloid-like birefringent rods. Cell, 35(2

Pt 1):349-58, 1983.

[14] Agrimi U, Nonno R, Dell’Omo G, Di Bari M.A, Conte M, Prion protein amino acid

determinants of differential susceptibility and molecular feature of prion strains in mice

and voles. PLoS Pathog, 25: e1000113, 2008.

[15] Christen B, Pérez DR, Hornemann S, Wüthrich K, NMR structure of the bank vole

prion protein at 20 degrees C contains a structured loop of residues 165-171. J Mol Biol,

383(2):306-12, 2008.

[16] Watts JC, Giles K, Stöhr J, Oehler A, Bhardwaj S, Grillo SK, Patel S, DeArmond SJ,

Prusiner SB, Spontaneous generation of rapidly transmissible prions in transgenic mice

expressing wild-type bank vole prion protein. Proc Natl Acad Sci U S A, 109(9):3498-

503, 2012.

[17] Di Bari MA, Nonno R, Castilla J, D'Agostino C, Pirisinu L, Riccardi G, Conte M,

Richt J, Kunkle R, Langeveld J, Vaccari G, Agrimi U, Chronic wasting disease in bank

voles: characterisation of the shortest incubation time model for prion diseases. PLoS

Pathog, 9(3):e1003219, 2013.

[18] Nonno R, Di Bari MA, Cardone F, Vaccari G, Fazzi P, Efficient transmission and

characterization of creutzfeldt–Jakob disease strains in bank voles. PLoS Pathog, 2: 12,

82
2006.

[19] Di Bari MA, Chianini F, Vaccari G, Esposito E, Conte M, The bank vole (Myodes

glareolus) as a sensitive bioassay for sheep scrapie. J Gen Virol, 89: 2975–85, 2008.

[20] Cartoni C, Schinina` ME, Maras B, Nonno R, Vaccari G, Identification of the

pathological prion protein allotypes in scrapie-infected heterozygous bank voles

(Clethrionomys glareolus) by high-performance liquid chromatography-mass

spectrometry. J Chromatogr A, 1081: 122–6, 2005.

[21] Di Bari MA, Chianini F, Vaccari G, Esposito E, Conte M, The bank vole (Myodes

glareolus) as a sensitive bioassay for sheep scrapie. J Gen Virol, 89: 2975–85, 2008.

[22] Jeon YC1, Choi JK, Choi EK, Carp RI, Kim YS, Pathological characterization of

TgElk mice injected with brain homogenate from elk with chronic wasting disease. J Vet

Sci, 14(1):21-6, 2013.

[23] Kim BH, Kim JI, Choi EK, Carp RI, Kim YS, A neuronal cell line that does not

express either prion or doppel proteins. Neuroreport, 16(5):425-9, 2005.

[24] Lee GH, Jang B, Choi HS, Kim HJ, Park JH1, Jeon YC, Carp RI, Kim YS, Choi EK,

Upregulation of Connexin 43 Expression Via C-Jun N-Terminal Kinase Signaling in

Prion Disease. J Alzheimers Dis, 49(4):1005-19, 2015

[25] Choi JK, Park SJ, Jun YC, Oh JM, Jeong BH, Lee HP, Park SN, Carp RI, Kim YS,

Generation of monoclonal antibody recognized by the GXXXG motif (glycine zipper) of

prion protein. Hybridoma (Larchmt), 25(5):271-7, 2006.

[26] Watts JC, Giles K, Stöhr J, Oehler A, Bhardwaj S, Grillo SK, Patel S, DeArmond SJ,

Prusiner SB, Spontaneous generation of rapidly transmissible prions in transgenic mice

expressing wild-type bank vole prion protein. Proc Natl Acad Sci U S A, 109(9):3498-

503, 2012.

83
[27] Vilette D, Cell models of prion infection. Vet Res, 39(4):10, 2008

[28] Béranger F, Mangé A, Solassol J, Lehmann S, Cell culture models of transmissible

spongiform encephalopathies. Biochem Biophys Res Commun, 289(2):311-6, 2001.

[29] Hcker R, Taraboulos A, Scott M, Pan KM, Yang SL, Torchia M, Jendroska K,

DeArmond SJ, Prusiner SB, Replication of distinct scrapie prion isolates is region specific

in brains of transgenic mice and hamsters. Genes Dev, 6(7):1213-28, 1992.

[30] Bosque PJ, Prusiner SB, Cultured cell sublines highly susceptible to prion infection.

J Virol, 74(9):4377-86, 2000.

[31] Prusiner SB, Scott M, Foster D, Pan KM, Groth D, Mirenda C, Torchia M, Yang SL,

Serban D, Carlson GA, Transgenetic studies implicate interactions between homologous

PrP isoforms in scrapie prion replication. Cell, 63(4):673-86, 1990.

[30] Bosque PJ, Prusiner SB, Cultured cell sublines highly susceptible [32] Collinge J,

Clarke AR, A general model of prion strains and their pathogenicity. Science,

318(5852):930-6, 2007.to prion infection. J Virol, 74(9):4377-86, 2000.

[31] Nishida N, Harris DA, Vilette D, Laude H, Frobert Y, Grassi J, Casanova D, Milhavet

O, Lehmann S, Successful transmission of three mouse-adapted scrapie strains to murine

neuroblastoma cell lines overexpressing wild-type mouse prion protein. J Virol,

74(1):320-5, 2000.

[31] Watts JC, Giles K, Patel S, Oehler A, DeArmond SJ, Prusiner SB, Evidence that

bank vole PrP is a universal acceptor for prions. PLoS Pathog, 3;10(4):e1003990, 2014

84
Establishment of Advanced In Vitro Model from Bank Vole Transgenic
Mice for Susceptibility to Various Prion Strains

2016. 88 page
Doctoral Degree
Hong-Seok Choi
Department of Microbiology
Advisor Prof. Yong-Sun Kim

ABSTRACT

Prions are infectious agents that cause a devastating neurodegenerative disease in both
humans and animals. Unlike other rodents, bank vole (Myodes glareolus, BV) is
susceptible to prions from a diverse range of species, including humans. There are two
lines of BVs, one homozygous for methionine (BV109M) and the other for isoleucine at
codon 109 (BV109I) of prion protein (PrP). Here, we established neuronal cell lines from
embryonic brain of BV109I mouse that was immortalized by an introduction of plasmid
DNA encoding for SV40-T antigen. Bv109I mice develop age-dependent signs of
spontaneous neurologic illness. The established BV cell lines infected with 22L scrapie
strain (BV-22L) showed a replication and an accumulation of disease-associated forms
of the PrP. BV-22L infected neuronal cells were then intracerebrally inoculated into ICR
mice showing clinical symptoms of disease after ~147 days post-inoculation. We also
confirmed that various prion strains, including ME7, 263K, CWD and 22L, were
successfully infected into BV neuronal cells. The newly established BV cell models may
enhance our understanding on the cellular mechanisms of prion replication and can be a
useful tool for the study of the pathogenic mechanisms of prion diseases.

Key words : bank vole, neuronal cell lines, prion disease, scrapie

85
Bank Vole 프리온 단백 발현 형질 전환 마우스의 신경세포에서 다양한

프리온 감염 세포주 확립

2016. 88p.

박사학위논문

최홍석

의학과 미생물학 전공

지도교수: 김용선

국문초록

프리온 질환은 사람과 동물 모두에서 발생하는 감염성 신경질환이다. 다른 설치류와는 달리

bank vole (Myodes glareolus, BV)은 인간을 포함한 다양한 종의 프리온에 감염될 수 있다.

프리온 질병의 발생에서 신경세포는 중요한 역할을 한다. 본 연구에서는 BV 유전자 이식

마우스의 배아뇌조직으로부터 해마부위 신경세포를 분리하고 SV large T antigen 감염에

의한 세포 불멸화 과정을 통해 신경세포주를 확립하였다. BV 신경세포주는 스크래피

스트레인 (22L)을 감염시켰으며, 50번 이상의 계대 배양한 이 세포주에서 지속적으로

비정상적인 프리온 단백의 복제나 축적을 확인하였으며 단백질 분해효소 K에 저항성을

가지는 비정상적인 프리온 단백의 축적을 확인하였다. 또한 22L 스크래피에 감염된 BV

신경세포주를 마우스 뇌에 주입한 결과 평균 147일 후에 임상증상을 확인하였다. BV

신경세포주는 스크래피 뿐만 아니라 다른 여러 종의 프리온 스트레인에 감염되었다. 따라서

본 연구에서 처음으로 확립된 BV 신경세포주는 다양한 프리온 스트레인에 감염될 수 있으며

프리온 질환의 발병원인을 연구하는 유용한 도구로 이용될 수 있을 것이다.

핵심어: 프리온 질환, Bank Vole 형질전환 마우스, 스크래피, 신경세포주

86
Establishment of Advanced In Vitro Model from Bank Vole

Transgenic Mice for Susceptibility to Various Prion Strains


The undersigned certify that they have read, and recommended to the Graduate

school for acceptance, a thesis entitled “Establishment of Advanced In Vitro Model from

Bank Vole Transgenic Mice for Susceptibility to Various Prion Strains” submitted by

Hong-Seok Choi in partial fulfillment or the requirements for the degree of Ph.D. in

Medical Science.

Dr. Hyung-Joo Kwon

Department of Microbiology

___________________

Dr. Kyung-Chan Choi

Department of Pathology

___________________

Dr. Jae-Bong Park

Department of Biochemistry

___________________

Dr. Jae-Il Kim

Department of Food Science & Nutrition

___________________

Dr. Yong-Sun Kim

Department of Microbiology

___________________

December, 2016

87
IV. Appendix

1. Choi HS, Choi YG, Shin HY, Oh JM, Park JH, Kim JI, Carp RI, Choi EK, Kim YS,

Dysfunction of mitochondrial dynamics in the brains of scrapie-infected mice. Biochem

Biophys Res Commun, 448(2):157-62, 2014

2. Lee GH*, Jang B*, Choi HS*, Kim HJ, Park JH1, Jeon YC, Carp RI, Kim YS, Choi

EK, Upregulation of Connexin 43 Expression Via C-Jun N-Terminal Kinase Signaling in

Prion Disease. J Alzheimers Dis, 49(4):1005-19, 2015.

* These authors contributed equally to this work.

88
Biochemical and Biophysical Research Communications 448 (2014) 157–162

Contents lists available at ScienceDirect

Biochemical and Biophysical Research Communications


journal homepage: www.elsevier.com/locate/ybbrc

Dysfunction of mitochondrial dynamics in the brains of scrapie-infected


mice
Hong-Seok Choi a,b, Yeong-Gon Choi b, Hae-Young Shin b, Jae-Min Oh b, Jeong-Ho Park a,b,
Jae-Il Kim c, Richard I. Carp d, Eun-Kyoung Choi b,⇑, Yong-Sun Kim a,b,⇑
a
Department of Microbiology, College of Medicine, Hallym University, 1 Okcheon-dong, Chuncheon, Gangwon-do 200-702, Republic of Korea
b
Ilsong Institute of Life Science, Hallym University, 1605-4 Gwanyang-dong, Dongan-gu, Anyang, Gyeonggi-do 431-060, Republic of Korea
c
Department of Food Science and Nutrition, Pukyong National University, 599-1 Daeyeon-3-dong, Nam-gu, Busan 608-737, Republic of Korea
d
New York State Institute for Basic Research in Developmental Disabilities, 1050 Forest Hill Road, Staten Island, NY 10314, USA

a r t i c l e i n f o a b s t r a c t

Article history: Mitochondrial dysfunction is a common and prominent feature of many neurodegenerative diseases,
Received 7 April 2014 including prion diseases; it is induced by oxidative stress in scrapie-infected animal models. In previous
Available online 19 April 2014 studies, we found swelling and dysfunction of mitochondria in the brains of scrapie-infected mice com-
pared to brains of controls, but the mechanisms underlying mitochondrial dysfunction remain unclear. To
Keywords: examine whether the dysregulation of mitochondrial proteins is related to the mitochondrial dysfunction
Prion disease associated with prion disease, we investigated the expression patterns of mitochondrial fusion and fission
Mitochondrial fusion
proteins in the brains of ME7 prion-infected mice. Immunoblot analysis revealed that Mfn1 was up-reg-
Fission
Scrapie
ulated in both whole brain and specific brain regions, including the cerebral cortex and hippocampus, of
Mitochondrial dysfunction ME7-infected mice compared to controls. Additionally, expression levels of Fis1 and Mfn2 were elevated
in the hippocampus and the striatum, respectively, of the ME7-infected brain. In contrast, Dlp1 expres-
sion was significantly reduced in the hippocampus in the ME7-infected brain, particularly in the cytosolic
fraction. Finally, we observed abnormal mitochondrial enlargement and histopathological change in the
hippocampus of the ME7-infected brain. These observations suggest that the mitochondrial dysfunction,
which is presumably caused by the dysregulation of mitochondrial fusion and fission proteins, may con-
tribute to the neuropathological changes associated with prion disease.
Ó 2014 Elsevier Inc. All rights reserved.

1. Introduction protein (PrPC) into the scrapie form of the pathogenic prion protein
(PrPSc) [5].
Prion diseases, also known as transmissible spongiform enceph- A number of recent studies have demonstrated that mitochon-
alopathies (TSEs), are fatal neurodegenerative disorders that affect dria are dynamic organelles that continually undergo fission and
both humans and animals [1,2]. Scrapie is a prototypical prion dis- fusion with one another [7–9]. Mitochondria can change in number
ease that affects sheep and goats. Clinically, scrapie is character- and morphology within a cell during development, throughout the
ized by a long latent period, progressive ataxia, tremor, wasting cell cycle and when challenged with various cytotoxic stimuli [9].
and ultimately death [3]. Many scrapie strains have been isolated In mammals, the key molecules involved in mitochondrial fission
from sheep and goats and used to examine not only the range of are dynamin-like protein 1 (Dlp1, also referred to as Drp1) and fis-
various pathogenesis pathways but also the neuropathological sion 1 (Fis1). The process opposing fission, i.e., mitochondrial
mechanisms induced by prion disease [4]. Typical features of the fusion, is controlled in mammalian cells by mitofusin 1 (Mfn1),
disease include the formation of spongiform vacuoles and astrocy- mitofusin 2 (Mfn2) and optic atrophy 1 (Opa1) [7]. The sizes,
tosis, the formation of amyloid plaques in some cases and neuronal shapes and interconnectivities of mitochondria are determined
loss in the brain [5,6]. A key event in prion disease is the conforma- by their fusion and fission [9]. It has been suggested that the mito-
tional misfolding of the endogenously expressed cellular prion chondrial defects associated with Alzheimer’s disease (AD), Parkin-
son’s disease (PD) and Huntington’s disease (HD) may result, at
least in part, from a disruption of the fusion and fission mecha-
⇑ Corresponding authors.
nisms of mitochondria [8,9]. A recent study reported that the
E-mail addresses: ekchoi@hallym.ac.kr (E.-K. Choi), yskim@hallym.ac.kr
expression of Dlp1 is decreased and that mitochondria are
(Y.-S. Kim).

http://dx.doi.org/10.1016/j.bbrc.2014.04.069
0006-291X/Ó 2014 Elsevier Inc. All rights reserved.
158 H.-S. Choi et al. / Biochemical and Biophysical Research Communications 448 (2014) 157–162

abnormally elongated in the fibroblasts of AD patients and in neu- (10–50 lg in all assays) were separated by SDS–PAGE using 10%,
ronal cell lines overexpressing amyloid precursor protein (APP) 12% or 15% acrylamide gels and then transferred to nitrocellulose
[10]. The study suggests that APP causes an imbalance between membranes (Thermo Scientific). After blocking with 5% skim milk
mitochondrial fusion and fission that results in an abnormal distri- for 1 h, the membranes were incubated with the individual antibod-
bution of mitochondria, which in turn contributes to mitochondrial ies overnight at 4 °C and then incubated with horseradish peroxi-
and neuronal dysfunction. Several observations suggest a link dase-conjugated secondary antibody in Tris-buffered saline with
between mitochondrial dysfunction and PD. Pink1 and parkin, 0.05% Tween-20 (TBST) containing 5% skim milk for 1 h at room tem-
which are PD-related genes, promote mitochondrial fission and perature. The blots were visualized with the SuperSignal West Pico
inhibit mitochondrial fusion in Drosophila [11]. Additionally, HD Chemiluminescent Substrate (Thermo Scientific). The expression
research has focused on mitochondrial dysfunction. A mouse levels of each protein were quantified using ImageJ software (NIH).
model of HD exhibits early defects in respiration and ATP produc-
tion [12]. Moreover, mutant huntingtin seems to disrupt mito- 2.4. Transmission electron microscopy (TEM)
chondrial Ca2+ buffering [13] and to cause mitochondrial
ultrastructural changes in the lymphoblasts of HD patients [14]. The animals were perfused with 0.1 M PBS (pH 7.4) containing
Previous studies reported that dysfunction and enlargement of 4% paraformaldehyde and 2.5% glutaraldehyde under deep anes-
the mitochondria occur by oxidative stress in animal models of thesia with 16.5% urethane. The brains were removed and fixed
prion disease [15,16]. However, the underlying mechanism respon- in the 0.1 M PBS fixative that was used for perfusion. The bilateral
sible for this mitochondrial dysfunction remains unclear. hippocampal regions were trimmed into small pieces immediately
In the present study, we investigated the mitochondrial fusion after their surgical removal and kept in the fixative for 2 h at 4 °C.
and fission proteins that may be involved in the mitochondrial dys- Post-fixation was performed in 0.1 M PBS with 1% osmium tetrox-
function observed in prion disease. We show that mitochondrial ide followed by dehydration through a graded ethanol series and
fusion and fission proteins are differentially modulated in the ter- embedding in Epon 812. Ultra-thin sections (75 nm) prepared by
minal stage of an experimental mouse model of prion disease and using an ultramicrotome (RMC MTXL) were stained with uranyl
that this modulation may contribute to the morphological damage acetate and lead citrate and were subsequently observed with a
and reduction in number of mitochondria in infected neuronal transmission electron microscope (JEM-1011, JEOL). The numbers
cells. of total and damaged mitochondria were counted in the hippocam-
pal neurons in the control and ME7-infected brains and then calcu-
2. Materials and methods lated based on fifteen arbitrarily selected hippocampal neurons.
The sizes of the mitochondria were measured under high magnifi-
2.1. Antibodies cation (50,000) using TEM (iTEM, Olympus Soft Imaging
Solutions, GmbH), and the values were calculated as the
The following monoclonal and polyclonal antibodies were used: means ± SDs of the lengths and widths of thirty arbitrarily selected
mouse monoclonal anti-PrP (3F10) [17], mouse monoclonal anti- mitochondria from each group. Statistical analyses of the numbers
COX IV (Abcam), goat polyclonal anti-enolase (Santa Cruz), mouse and sizes of the mitochondria were performed using Jandel
monoclonal to b-actin (Sigma–Aldrich), mouse monoclonal SigmaStat software (V 3.5).
anti-Opa1 and mouse monoclonal anti-Dlp1 (BD Transduction
Laboratories), mouse monoclonal anti-Mfn2, chicken polyclonal 2.5. Statistical analyses
anti-Mfn1 (Novus Biologicals) and rabbit polyclonal anti-Fis1
(Santa Cruz). The compared values were calculated as the mean ± SD of three
brains from each group, and statistical significance was deter-
2.2. Animals and scrapie strains mined using Student’s t-tests.

Six-week-old C57BL/6 mice were obtained from the Central 3. Results


Laboratory Animal (Republic of Korea) and divided into two
groups: one group was infected with the ME7 scrapie strain, and 3.1. Imbalanced expression of mitochondrial fusion and fission
the other included age-matched controls. The ME7 scrapie strain proteins in infected brains
was kindly provided by Dr. Alan Dickinson (Neuropathogenesis
Unit, Edinburgh, UK). This scrapie strain was maintained by serial First, we identified deposits of PK-resistant PrPSc in ME7-infected
intracerebral passages of brain homogenate from a terminally brains in the end stage; the normal PrPC proteins were completely
affected mouse. The mice were intracerebrally inoculated with degraded by the PK treatment, as shown in Fig. 1. To determine
30 ll of 1% (w/v) brain homogenate in 0.01 M phosphate-buffered whether the mitochondrial fusion and fission proteins are affected
saline (PBS, pH 7.4) from either a normal brain or an ME7-infected by prion infection, their expression patterns were investigated in
C57BL/6 mouse brain at the terminal stage of the disease. When whole brains of the control and ME7-infected mice at the end stage
the clinical signs of prion disease were evident in the terminal of disease (160 dpi) (Fig. 2). Of the mitochondrial fusion and fission
stage (160 days postinoculation, dpi), the mice were sacrificed. proteins (fusion proteins: Opa1, Mfn1 and Mfn2; fission proteins:
Dlp1 and Fis1), Western blot analysis revealed that only Mfn1 was
2.3. Western blot analysis differentially expressed in the infected whole brains compared to
the age-matched control brains (Fig. 2A and B). It has previously been
Whole brains or hippocampal regions were homogenized gently reported that the hippocampal regions are more severely damaged
in 10-fold greater volumes (w/v) of 50 mM Tris–HCl (pH 7.4) than any other regions in brains infected with the ME7 scrapie strain
containing 150 mM NaCl, 1 mM EDTA, 0.25% Na-deoxycholate, 1% [18]. Thus, to compare the expression levels of the mitochondrial
NP-40 and protease inhibitor cocktails (Roche). The protein fusion and fission proteins in various brain regions, we dissected
concentration was determined using the BCA assay (Thermo both the control and ME7-infected brains into the following regions:
Scientific). The homogenates were treated with Proteinase K (PK) cerebral cortex, hippocampus, cerebellum, striatum and brainstem.
at a concentration of 50 lg/ml. Equal amounts of protein We found that the levels of Mfn1 and Fis1 were significantly
H.-S. Choi et al. / Biochemical and Biophysical Research Communications 448 (2014) 157–162 159

3.2. Analysis of fission proteins in the hippocampal cytosol of infected


brain

To examine the localization of the mitochondrial fusion and fis-


sion proteins in the organelles and the expression levels of these
proteins in the cytosol and mitochondria, mitochondrial and cyto-
solic fractions were isolated from the hippocampi of control and
ME7-infected mice; again the hippocampus was chosen because
this region is known to be severely damaged in ME7-infected
brains [18]. Immunoblot analyses revealed that Dlp1 levels in the
mitochondrial fractions from the hippocampi were not altered by
Fig. 1. Deposition of PK-resistant prion isoforms in ME7-infected mice. Whole ME7 infection (Fig. 3A and B) but were significantly decreased in
brains of control (n = 3) and infected mice (n = 3) in the end stage (160 dpi) were the cytosolic fractions of the infected hippocampi; this finding is
homogenized and blotted with anti-PrP antibody (3F10). A portion of each sample consistent with the decrease in Dlp1 levels in the affected hippo-
was treated with PK (PK 50 lg/ml). b-Actin was used as a loading control. campi that is illustrated in Fig. 2C and D. The concentration of
Opa1, which is primarily localized to the inner membrane of the
increased in the hippocampi of the infected brains, whereas the Dlp1 mitochondria, was not altered in the mitochondria or cytosol iso-
levels were significantly decreased at the end stage (Fig. 2C and D). lated from the hippocampi of the infected brains (Fig. 3C and D).
Additionally, the levels of Mfn1 and Mfn2 were increased in the cere-
bral cortex and striatum, respectively, of infected brains (data not 3.3. Morphological changes in the mitochondria in the hippocampal
shown). In the cerebellum and brain stem, none of the five mitochon- region of infected brain
drial fusion and fission proteins was significantly differentially
expressed between the control and infected mice (data not shown). The modulation of mitochondrial dynamics due to fusion and
These results indicate that the differential regulation of mitochon- fission protein concentration probably contributed to alterations
drial fusion and fission proteins may be involved in the mitochon- in mitochondrial shape [9]. Electron microscopic analyses were
drial dysfunction of the ME7-infected brains, particularly in the performed to assess the relationship between mitochondrial fusion
hippocampus [16]. and fission proteins and mitochondrial shapes in the hippocampal

Fig. 2. Differential expression of mitochondrial fusion and fission proteins in hippocampi of ME7-infected mice. The mitochondrial dynamic proteins of the whole brains and
hippocampal regions of the control (n = 3) and infected mice (n = 3) in the end stage of disease were blotted. b-Actin was used as a loading control for A and C. The intensities
of the bands in panels A and C were measured and quantified (B and D). The values are expressed as the mean ± SD (n = 3). CON: control (white bars); ME7: ME7-infected
(black bars). Statistically significant differences are indicated (⁄p < 0.05 and ⁄⁄p < 0.01).
160 H.-S. Choi et al. / Biochemical and Biophysical Research Communications 448 (2014) 157–162

Fig. 3. Subcellular localization of mitochondrial fusion and fission proteins in hippocampi of ME7-infected mice. Opa1 (fusion) and Dlp1 (fission) from the mitochondria (A)
and cytosolic (C) fractions of the hippocampi of the control (n = 3) and ME7-infected mice (n = 3) at the end stage of the disease were blotted (160 dpi). b-Actin was used as a
loading control. The intensities of the bands in panels A and C were measured and quantified (B and D). The values are expressed as the mean ± SD (n = 3). CON: control (white
bars); ME7: ME7-infected (black bars). Note that both of the Opa1 bands are primarily localized to the mitochondria of hippocampi in both the control and infected
specimens, whereas substantial amounts of Dlp1 were localized to the cytosol. Statistically significant differences are indicated (⁄p < 0.05).

region. As shown in Fig. 4, the mitochondria in the normal hippo- cortex; Mfn1, Dlp1 and Fis1 in the hippocampus; and Mfn2 in
campal neurons were present in a variety of shapes, including rod, the striatum.
round and elliptical shapes. The cristae were normal in appearance. A recent study suggested that most neurodegenerative diseases
In contrast, enlarged mitochondria with damaged cristae were may be associated with the dysregulation of mitochondrial fusion
clearly observed in many of the hippocampal neurons in infected and fission proteins [19]. Mitochondrial fusion and fission are reg-
brains (Fig. 4A). The numbers of total neuronal mitochondria were ulated by large dynamin-related GTPases. Mitochondrial fusion,
significantly decreased in the hippocampi of the infected mice which is regulated by three large GTPases (Mfn1, Mfn2 and
compared to those of controls (Fig. 4B). Additionally, the lengths Opa1), involves the coordinated fusion of both the outer and inner
and widths of the mitochondria in the hippocampal neurons of mitochondrial membranes [20]. The overexpression of mitofusins
infected brains were significantly increased compared to those of causes the normally punctate mitochondria to become elongated,
the controls, which is consistent with the observation of enlarged whereas the repression of mitofusins causes the fragmentation of
hippocampal mitochondria visualized in Fig. 4B. The morphologi- the mitochondrial network in tissue culture cells [21]. Interest-
cal changes seen in mitochondria may be involved in the mito- ingly, we found increased level of Mfn1, which is a transmembrane
chondrial dysfunction in the hippocampi of ME7-infected mice. protein that localizes to the outer membranes of mitochondria, in
the homogenates of whole brain, hippocampus and cerebral cortex
of the infected brain. In addition the expression level of Mfn2 was
4. Discussion increased in the striatum of the infected brain (Data not shown).
Although electron microscopy analyses did not reveal the detailed
In this study, we demonstrated for the first time that the membrane structures of the mitochondria in this study, initial
expression patterns of mitochondrial fusion and fission proteins outer membrane fusion may have occurred in the brains of the
were altered in an experimental mouse model of prion disease; infected mice due to increases in the Mfn1 and Mfn2 proteins.
i.e., ME7 scrapie-infected mice. Of the mitochondrial fusion and fis- Moreover, although there may be distinct pathways or mecha-
sion proteins examined, the levels of Mfn1 were found to be signif- nisms for Mfn1 and Mfn2, a functional interplay between the
icantly increased in whole brains of the ME7-infected mice. Within two proteins may be involved in the control of mitochondrial
the different dissected regions of the ME7-infected brains, we fusion [22].
found differences in the expression levels of various mitochondrial Different strains of mouse-adapted scrapie have been reported
fusion and fission proteins compared to controls. Particularly nota- to preferentially target different specific brain regions; as noted
ble were the alterations in the expression of Mfn1 in the cerebral previously, the ME7 strain is known to be particularly associated
H.-S. Choi et al. / Biochemical and Biophysical Research Communications 448 (2014) 157–162 161

Fig. 4. Electron microscopic analysis of the mitochondria in hippocampal neurons of ME7-infected mice. (A) Control mitochondria were elliptical in shape and exhibited
compact cristae, whereas the infected mitochondria were enlarged and exhibited partially swollen cristae. Scale bar, 1 lm. (B) The total number of mitochondria in the
hippocampal neurons of the control and ME7-infected mice (the total number of mitochondria were counted from 15 neuronal cells from each group). (C) The lengths (white
bar) and widths (black bar) of the mitochondria in the hippocampal neurons of the control and ME7-infected mice (the lengths and widths of 30 mitochondria in each group
were measured). Statistically significant differences are indicated (⁄p < 0.01). CON: control; ME7: ME7-infected.

with hippocampal damage [23]; therefore, we focused on the hip- relationship between expression levels of the mitochondrial fusion
pocampal region of the ME7-infected brains. Our study demon- and fission proteins and the alterations in the shapes, numbers and
strated that ME7 infection led to significant increases in the sizes of the mitochondria in a prion disease model. In the ME7
levels of Mfn1 and Fis1 in the hippocampi of the ME7-infected mouse model, we found that the total number of neuronal mito-
brains, whereas Dlp1 was significantly decreased. Dlp1 localizes chondria was reduced significantly and that a number of enlarged
in several organelles, including the endoplasmic reticulum, micro- and degenerated mitochondria appeared. These mitochondria had
tubules and peroxisomes, but primarily resides in the mitochon- damaged cristae and matrix. The relationship between the mito-
dria and cytosol [24]. We isolated the cytosolic fraction of the chondrial damage and the induction of neurodegeneration and dis-
hippocampal region and investigated the changes in the expression ease is uncertain. Clearly the fact that the number of mitochondria
of mitochondrial fusion and fission proteins in both the purified was reduced combined with degeneration of the remaining mito-
mitochondria fraction and the cytosol. Dlp1 levels were decreased chondria would lead to a loss of neuronal function. It needs to be
in cytosolic fractions but not in mitochondria fractions. These noted that the loss of mitochondria could affect the quantity of
observations imply that imbalances in mitochondrial dynamics proteins observed; this is particularly important in instances
may contribute to the enlargement and the degeneration of mito- where the level of protein decreased (e.g., Dlp1 in the cytosol). Fur-
chondria that occurs in the hippocampi of scrapie-infected mice. thermore, in those instances where there was no change in protein
Changes in the shape of the mitochondria have been observed levels (e.g., Opa1), the reduced number of mitochondria could
in ME7 scrapie-infected brain even in the preclinical stage of infec- mask an increase.
tion [25]. The mitochondrial enlargement demonstrated during the Growing evidence suggests that the delicate balance between
end stage of prion disease in this study was also observed in a pre- mitochondrial fission and fusion is critical for mitochondrial func-
vious study that demonstrated structural abnormalities of the tions, including energy production, Ca2+ signaling, reactive oxygen
mitochondria in the neurons of the hippocampi and cerebral corti- species (ROS) production, apoptosis and senescence [7,27]. Dysreg-
ces of brains from scrapie-infected hamsters [15]. Fragmentation ulation of the mitochondrial fusion and fission proteins in neurons
and clustering of mitochondria in the soma have been observed may be a common pathway leading to the mitochondrial and neu-
in the brains of patients with sporadic AD, and alterations in the ronal dysfunctions that are critical in the pathogenesis of prion dis-
expressions of several fusion and fission proteins have been ease. It remains unclear, however, how the alterations in
observed in these brains [26]. Thus, we sought to determine the mitochondrial fusion and fission proteins that were observed in
162 H.-S. Choi et al. / Biochemical and Biophysical Research Communications 448 (2014) 157–162

this study contribute to neurodegeneration in prion diseases. [12] T. Milakovic, G.V. Johnson, Mitochondrial respiration and ATP production are
significantly impaired in striatal cells expressing mutant huntingtin, J. Biol.
Taken together, the findings of this study suggest that the disrup-
Chem. 280 (2005) 30773–30782.
tion of the balance of mitochondrial fusion and fission proteins, [13] A.V. Panov, C.A. Gutekunst, B.R. Leavitt, M.R. Hayden, J.R. Burke, W.J.
which may be a common feature of prion diseases, is involved in Strittmatter, J.T. Greenamyre, Early mitochondrial calcium defects in
the mitochondrial and neuronal dysfunction in the brains of Huntington’s disease are a direct effect of polyglutamines, Nat. Neurosci. 5
(2002) 731–736.
ME7-infected mice. [14] F. Squitieri, M. Cannella, G. Sgarbi, V. Maglione, A. Falleni, P. Lenzi, A. Baracca,
G. Cislaghi, C. Saft, G. Ragona, M.A. Russo, L.M. Thompson, G. Solaini, F. Fornai,
Acknowledgments Severe ultrastructural mitochondrial changes in lymphoblasts homozygous for
Huntington disease mutation, Mech. Ageing Dev. 127 (2006) 217–220.
[15] S.I. Choi, W.K. Ju, E.K. Choi, J. Kim, H.Z. Lea, R.I. Carp, H.M. Wisniewski, Y.S. Kim,
This research was supported by Hallym University Research Mitochondrial dysfunction induced by oxidative stress in the brains of
Fund, 2012 (HRF-201210-000), the National Research Foundation hamsters infected with the 263 K scrapie agent, Acta Neuropathol. 96 (1998)
279–286.
of Korea Grant funded by the Korean Government (NRF-2011- [16] J.H. Park, B.H. Kim, S.J. Park, J.K. Jin, Y.C. Jeon, G.Y. Wen, H.Y. Shin, R.I. Carp, Y.S.
619-E0001) and by the framework of international cooperation Kim, Association of endothelial nitric oxide synthase and mitochondrial
program managed by the National Research Foundation of Korea dysfunction in the hippocampus of scrapie-infected mice, Hippocampus 21
(2010) 319–333.
(NRF-2011-616-H00002). [17] J.K. Choi, S.J. Park, Y.C. Jun, J.M. Oh, B.H. Jeong, H.P. Lee, S.N. Park, R.I. Carp, Y.S.
Kim, Generation of monoclonal antibody recognized by the GXXXG motif
References (glycine zipper) of prion protein, Hybridoma (Larchmt) 25 (2006) 271–277.
[18] W.K. Ju, K.J. Park, E.K. Choi, J. Kim, R.I. Carp, H.M. Wisniewski, Y.S. Kim,
Expression of inducible nitric oxide synthase in the brains of scrapie-infected
[1] A. Aguzzi, C. Weissmann, Prion research: the next frontiers, Nature 389 (1997)
mice, J. Neurovirol. 4 (1998) 445–450.
795–798.
[19] S. Frank, Dysregulation of mitochondrial fusion and fission: an emerging
[2] D.C. Bolton, M.P. McKinley, S.B. Prusiner, Identification of a protein that
concept in neurodegeneration, Acta Neuropathol. 111 (2006) 93–100.
purifies with the scrapie prion, Science 218 (1982) 1309–1311.
[20] D.C. Chan, Mitochondrial fusion and fission in mammals, Annu. Rev. Cell Dev.
[3] R.H. Kimberlin, C. Walker, Characteristics of a short incubation model of
Biol. 22 (2006) 79–99.
scrapie in the golden hamster, J. Gen. Virol. 34 (1977) 295–304.
[21] A. Sante, M.T. Fuller, Control of mitochondrial morphology by a human
[4] R.L. Chandler, B.A. Turfrey, Inoculation of voles, Chinese hamsters, gerbils and
mitofusin, J. Cell Sci. 114 (2001) 867–874.
guinea-pigs with scrapie brain material, Res. Vet. Sci. 13 (1972) 219–224.
[22] S.A. Detmer, D.C. Chan, Complementation between mouse Mfn1 and Mfn2
[5] R.I. Carp, X. Ye, R.J. Kascsak, R. Rubenstein, The nature of the scrapie agent.
protects mitochondrial fusion defects caused by CMT2A disease mutations, J.
Biological characteristics of scrapie in different scrapie strain–host
Cell Biol. 176 (2007) 405–414.
combinations, Ann. N. Y. Acad. Sci. 724 (1994) 221–234.
[23] R.M. Deacon, D. Reisel, V.H. Perry, J. Nicholas, P. Rawlins, Hippocampal scrapie
[6] P.J. Lucassen, A. Williams, W.C. Chung, H. Fraser, Detection of apoptosis in
infection impairs operant DRL performance in mice, Behav. Brain Res. 157
murine scrapie, Neurosci. Lett. 198 (1995) 185–188.
(2005) 99–105.
[7] B. Su, X. Wang, L. Zheng, G. Perry, M.A. Smith, X. Zhu, Abnormal mitochondrial
[24] Y. Yoon, E.W. Krueger, B.J. Oswald, M.A. McNiven, The mitochondrial protein
dynamics and neurodegenerative diseases, Biochim. Biophys. Acta 2010 (1802)
hFis1 regulates mitochondrial fission in mammalian cells through an
135–142.
interaction with the dynamin-like protein DLP1, Mol. Cell. Biol. 23 (2003)
[8] M.T. Lin, M.F. Beal, Mitochondrial dysfunction and oxidative stress in
5409–5420.
neurodegenerative diseases, Nature 443 (2006) 787–795.
[25] Z. Sisková, D.J. Mahad, C. Pudney, G. Campbell, M. Cadogan, A. Asuni, V.
[9] H. Chen, D.C. Chan, Mitochondrial dynamics-fusion, fission, movement, and
O’Connor, V.H. Perry, Morphological and functional abnormalities in
mitophagy-in neurodegenerative diseases, Hum. Mol. Genet. 18 (2009) R169–
mitochondria associated with synaptic degeneration in prion disease, Am. J.
R176.
Pathol. 177 (2010) 1411–1421.
[10] X. Wang, B. Su, S.L. Siedlak, P.I. Moreira, H. Fujioka, Y. Wang, G. Casadesus, X.
[26] X. Wang, B. Su, H.G. Lee, X. Li, G. Perry, M.A. Smith, X. Zhu, Impaired balance of
Zhu, Amyloid-beta overproduction causes abnormal mitochondrial dynamics
mitochondrial fission and fusion in Alzheimer’s disease, J. Neurosci. 29 (2009)
via differential modulation of mitochondrial fission/fusion proteins, Proc. Natl.
9090–9103.
Acad. Sci. U.S.A. 105 (2008) 19318–19323.
[27] S. Frank, B. Gaume, E.S. Bergmann-Leitner, W.W. Leitner, E.G. Robert, F. Catez,
[11] H. Deng, M.W. Dodson, H. Huang, M. Guo, The Parkinson’s disease genes pink1
C.L. Smith, R.J. Youle, The role of dynamin-related protein 1, a mediator of
and Parkin promote mitochondrial fission and/or inhibit fusion in Drosophila,
mitochondrial fission, in apoptosis, Dev. Cell 1 (2001) 515–525.
Proc. Natl. Acad. Sci. U.S.A. 105 (2008) 14503–14508. 2.
Journal of Alzheimer’s Disease 49 (2016) 1005–1019 1005
DOI 10.3233/JAD-150283
IOS Press

Upregulation of Connexin 43 Expression


Via C-Jun N-Terminal Kinase Signaling
in Prion Disease
Geon-Hwi Leea,b,1 , Byungki Janga,1 , Hong-Seok Choia,c,1 , Hee-Jun Kima , Jeong-Ho Parka,c ,
Yong-Chul Jeona , Richard I. Carpd , Yong-Sun Kima,c and Eun-Kyoung Choia,b,∗
a IlsongInstitute of Life Science, Hallym University, Anyang, Gyeonggi-do, Republic of Korea
b Department of Biomedical Gerontology, Graduate School of Hallym University, Chuncheon, Gangwon-do,
Republic of Korea
c Department of Microbiology, College of Medicine, Hallym University, Chuncheon, Gangwon-do, Republic of Korea
d New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA

Accepted 24 September 2015

Abstract. Prion infection leads to neuronal cell death, glial cell activation, and the accumulation of misfolded prion proteins.
However, the altered cellular environments in animals with prion diseases are poorly understood. In the central nervous system,
cells connect the cytoplasm of adjacent cells via connexin (Cx)-assembled gap junction channels to allow the direct exchange of
small molecules, including ions, neurotransmitters, and signaling molecules, which regulate the activities of the connected cells.
Here, we investigate the role of Cx43 in the pathogenesis of prion diseases. Upregulated Cx43 expression, which was dependent
on c-Jun N-Terminal Kinase (JNK)/c-Jun signaling cascades, was found in prion-affected brain tissues and hippocampal neuronal
cells. Scrapie infection-induced Cx43 formed aggregated plaques within the cytoplasmic compartments at the cell-cell interfaces.
The ethidium bromide (EtBr) uptake assay and scrape-loading dye transfer assay demonstrated that increased Cx43 has functional
consequences for the activity of Cx43 hemichannels. Interestingly, blockade of PrPSc accumulation reduced Cx43 expression
through the inhibition of JNK signaling, indicating that PrPSc accumulation may be directly involved in JNK activation-mediated
Cx43 upregulation. Overall, our findings describe a scrapie infection-mediated novel regulatory signaling pathway of Cx43
expression and may suggest a role for Cx43 in the pathogenesis of prion diseases.

Keywords: Connexin 43, gap junction, JNK, prion protein, scrapie

INTRODUCTION Cx43 is the most ubiquitous; it is abundantly expressed


in the brain and highly expressed during embryonic
Gap junctions are a complex of membrane channels development [2, 3]. In the central nervous system
that allow the diffusion of small molecules (<1.5 kDa) (CNS), Cx43 is widely expressed in neurons, astro-
between adjacent cells to form a functional syncytium. cytes, and oligodendrocytes [4–6], and various signal
Gap junctions are composed of two hemichannels molecules, including ATP, glutamate, and Ca2+ ions,
(connexons) formed by hexamers of connexins (Cxs) diffuse through functional Cx43 hemichannels [7–11],
[1]. Of the Cx genes that code for gap junction proteins, thereby modulating crucial CNS processes [12].
Cx43 has contributed to neuronal death in vitro
1 These authors contributed equally to this work.
∗ Correspondence
[11, 13], in a rat cortical ablation model [14], and
to: Eun-Kyoung Choi, Ilsong Institute of Life
in ischemic brain injury [15]. Cx43 plays a protec-
Science and Department of Biomedical Gerontology, Hallym Uni-
versity, 15 Gwanpyeong-ro, 170beon-gil, Dongan-gu, Anyang, tive role against oxidative stress-induced cell death
Gyeonggi-do, 431-815, Republic of Korea. Tel.: +82 31 380 1893; [16, 17]; deletion or blockade of this protein prevents
Fax: +82 31 388 3427; E-mail: ekchoi@hallym.ac.kr. chronic neuropathic pain following spinal cord injury

ISSN 1387-2877/16/$35.00 © 2016 – IOS Press and the authors. All rights reserved
1006 G.-H. Lee et al. / Scrapie Infection-Mediated Cx43 Upregulation

[18, 19] and fetal ischemia [20]. The deletion of Cx43 Animal, Inc. (Seoul, Republic of Korea). The origi-
or expression of a truncated form increases the vulner- nal stocks of the ME7, 22L, 139A, and 263K scrapie
ability to stroke [21, 22]. In addition, Cx43 is involved strains were kindly provided by Dr. Alan Dickinson
in neuronal differentiation [23–25], cellular mortality of the Agriculture and Food Research Council and
[26–29], hippocampal proliferation, and the survival Medical Research Council Institute (Neuropathogene-
of newborn cells [30]. sis Unit, Edinburgh, UK). For scrapie infection, the
Moreover, the role of Cx43 has been implicated in mice were intracerebrally inoculated with 30 ␮l of
various pathological conditions of the brain. Increased 1% w/v brain homogenates of the ME7, 22L, and
Cx43 levels have been identified within amyloid 139A inocula (for mice) or 50 ␮l of 1% w/v ham-
plaques in the brains of Alzheimer’s disease patients ster brain homogenate of the 263K inoculum (for
and in an animal model of the disease [31, 32], as well hamsters) in phosphate-buffered saline (PBS, pH 7.4)
as in the anterior horns of mSOD1-Tg mice, which rep- using a stereotaxic apparatus (Stoelting, Wood Dale,
resent an amyotrophic lateral sclerosis mouse model IL, USA). The control mice received 30 ∼ 50 ␮l of 1%
[33], the MPTP-lesioned striatum of a Parkinson’s w/v normal brain homogenate. The scrapie-infected
disease model [34], and the hippocampal regions of and uninfected mice were sacrificed at 10–150 days
patients with mesial temporal lobe epilepsy [35]. In post-inoculation (dpi) or at the terminal stage (160 dpi
contrast, the loss of Cx43 expression is found in the for ME7, 140 dpi for 22L, and 170 dpi for 139A) when
actively demyelinating lesions of multiple sclerosis the mice displayed typical clinical signs of the dis-
and in the active perivascular lesions of neuromyeli- ease. All experiments were performed in accordance
tis optica [36]. Therefore, Cx43 is associated with the with Korean laws and with the approval of the Hal-
pathophysiology of various tissue conditions. How- lym Medical Center Institutional Animal Care and Use
ever, to the best of our knowledge, there are no data Committee (HMC2011-0-0115-07).
available regarding the role of Cx43 in prion diseases.
Transmissible spongiform encephalopathies or Subcellular fractionation
prion diseases are infectious and fatal neurodegenera-
tive diseases. A mutation and an abnormal structure Mouse brain tissues were lysed in cold hypotonic
of the prion protein (PrPC ) are associated with a buffer (10 mM Tris-HCl, pH 7.4; 1 mM DTT; 5 mM
protease-resistant and infectious form (PrPSc ), which is MgCl2 ; 10 mM KCl; 10 mM NaF; and 1 mM Na3 VO4 )
considered a causative factor for prion diseases. Neu- with a protease inhibitor cocktail tablet (Roche, Indi-
ropathologically, prion diseases are characterized by anapolis, IN, USA) by passing through a 23-gauge
a long incubation period of many months to several syringe needle 10 times. The lysates were centrifuged
decades, neuronal vacuolation (spongiosis), neuronal at 500 g for 10 min to remove the nuclei and unbroken
cell death, and glial cell activation (microgliosis and cells. The post-nuclear supernatants were subsequently
astrocytosis) [37]. These changes lead to the release centrifuged at 100,000 g for 1 h at 4◦ C to separate
of inflammatory molecules, such as proinflammatory the membrane pellet from the cytosolic fraction. The
cytokines, reactive oxygen species, proteases, and membrane pellets were washed with ice-cold PBS
complement proteins that induce neuronal damage and and resuspended in modified RIPA buffer (50 mM
remove the damaged cells [37]. Tris-HCl, pH 7.4; 150 mM NaCl; 2 mM EDTA; 1%
The aim of the present study was to determine the Triton X-100; 1% Nonidet P (NP)-40; 0.25% sodium
specific role of Cx43 in prion pathogenesis using cel- deoxycholate; 1 mM Na3 VO4 ; and 10 mM NaF) with
lular and mouse models of prion disease. In this study, a protease inhibitor cocktail tablet by rocking for 1 h
we report for the first time that Cx43 expression is at 4◦ C, followed by centrifugation at 20,000 g for
increased via the c-Jun N-Terminal Kinase (JNK) sig- 10 min at 4◦ C. The supernatant contained the solubi-
naling pathway in both in vitro and in vivo models of lized membrane proteins.
prion disease.
Western blot analysis
MATERIALS AND METHODS
The mouse brains and cultured cells were lysed
Animals and scrapie strains in modified RIPA buffer with 20 mM Tris-HCl (pH
7.5), 150 mM NaCl, 1% Triton, 0.5% sodium deoxy-
C57BL/6J mice and golden Syrian hamsters (4 to cholate, 0.1% sodium dodecyl sulfate (SDS), 1%
6 weeks of age) were purchased from Central Lab NP-40, 10 mM NaF, 1 mM Na3 VO4 , 1 mM EDTA,
G.-H. Lee et al. / Scrapie Infection-Mediated Cx43 Upregulation 1007

1 mM EGTA and 0.1 M phenylmethylsulfonyl fluoride Cell blot assay


(PMSF), as well as a protease inhibitor cocktail. The
homogenates were centrifuged at 15,000 g at 4◦ C for Stable scrapie infection was confirmed after five
30 min, the supernatants were collected, and the protein passages as previously described [40]. Briefly, cells
concentrations were determined using a BCA protein were grown to confluence on Thermanox plastic cover
assay kit (Pierce, Rockford, IL, USA). Equal protein slips (Nalgene Nunc International, Rochester, NY,
amounts were separated by 10 or 12% SDS-PAGE USA) and transferred to nitrocellulose membrane.
and then transferred to PVDF membrane using an After drying for 1 h at 37◦ C, the membrane was
electrotransfer system (Bio-Rad, Hercules, CA, USA). incubated in lysis buffer (50 mM Tris-HCl, pH 8.0;
To detect the target proteins, mouse monoclonal anti- 150 mM NaCl; 0.5% sodium deoxycholate; 0.5% Tri-
PrP (3F10, 1:3,000) [38] rabbit polyclonal anti-Cx43 ton X-100; 2 mM PMSF) containing PK (5 ␮g/ml)
(1:1,000; Cell Signaling Technology, Beverly, MA, at 37◦ C for 10 min. The membrane was placed
USA), mouse monoclonal anti-JNK (1:1,000; Santa into 3 M guanidinium thiocyanate (Sigma-Aldrich),
Cruz Biotechnology, Santa Cruz, CA, USA), mouse 10 mM Tris·HCl (pH 8.0) for 10 min followed
monoclonal anti-phospho-JNK (1:1,000, Santa Cruz by immunostaining with anti-PrP antibody 3F10
Biotechnology), rabbit polyclonal anti-c-Jun (1,000; (1:3,000).
Santa Cruz Biotechnology), rabbit polyclonal anti-
phospho-c-Jun (1:1000; Cell Signaling Technology),
and mouse monoclonal anti-␤-actin (1:10,000; Sigma- Semi-quantitative RT-PCR
Aldrich, Saint Louis, MO, USA) antibodies were used
with the appropriate secondary antibodies conjugated Total RNA was extracted from the brain sam-
to horseradish peroxidase. To detect PrPSc , 50 ␮g/ml ples using TRI reagent (Sigma-Aldrich) according to
or 20 ␮g/ml of proteinase K (PK) were treated for the manufacturer’s protocols. cDNA was synthesized
30 min at 37◦ C in cell lysates or brain homogenates, using the Moloney murine leukemia virus reverse tran-
respectively. The target signals were visualized by dig- scriptase (Promega, Madison, WI, USA). The primer
ital images captured with an ImageQuant™ LAS 4000 sequences were as follows: connexin43 (127 bp),
imager (GE Healthcare Life Sciences, Piscataway, NJ, sense, 5 -CTGAGTGCGGTCTACACCTG-3 , anti-
USA) using an enhanced chemiluminescence western sense, 5 -GAGCGAGAGACACCAAGGAC-3 ; ␤-
blot detection system (Amersham Biosciences, Piscat- actin (196 bp), sense, 5 -TGTGATGGACTCCGGT
away, NJ, USA). GACGG-3 , antisense, 5 -ACAGCTTCTCTTTGATG
TCACGC-3 . The PCR products were separated by
Maintenance and scrapie infection of cultured cell electrophoresis on a 1% agarose gel and visualized
lines under UV light.

Mouse hippocampal neuronal cell lines, including


ZW13-2 (wild-type PrP) and Zpl2-4 (PrP knock- Immunohistochemistry
out) cells, were previously established [39]. The cells
were cultured in Dulbecco’s modified Eagle’s medium Immunohistochemical procedures were performed
(Hyclone, Logan, UT, USA) with 10% fetal bovine as previously described [41]. The sections were
serum (FBS, Hyclone), 100 units/ml penicillin, and blocked with 10% normal donkey serum in Tris-
100 ␮g/ml streptomycin in a 37◦ C incubator with 5% buffered saline (50 mM Tris-HCl and 150 mM NaCl,
CO2 . The ZW13-2 cells were persistently infected pH 7.6) and then incubated overnight at 4◦ C
with the 22L and 139A scrapie strains as previously with rabbit polyclonal anti-Cx43 antibody (1:100).
described [40]. The infected cells were maintained in After washing in PBS, the sections were first
Opti-MEM (Sigma-Aldrich) with 10% fetal calf serum incubated with biotinylated donkey anti-rabbit IgG
(Hyclone) and sub-cultured every 3 days at a 1:2 split antibody (1:500; Vector Laboratories, Burlingame,
for the first 10 passages. The infected cells stably CA, USA) for 1 h and then with avidin-biotin
produced PrPSc for over 50 passages. For immuno- peroxidase complex (ABC Kit, Vector Laborato-
cytochemistry, dye uptake assay, and scrape-loading ries). The sections were mounted in Permount
dye transfer assay, cells were seeded onto coverslips at (Thermo Fisher Scientific, Inc., Waltham, MA, USA).
2 × 104 cells per well in 24-well plates and allowed to The control sections were stained without primary
adhere for 2 days. antibody.
1008 G.-H. Lee et al. / Scrapie Infection-Mediated Cx43 Upregulation

Immunocytochemistry with HBSS and fixed with 100% MeOH for 15 min
at –20◦ C. The cells were observed under a confocal
The cells were fixed with 4% paraformaldehyde in laser scanning microscope. The distances of LY dif-
PBS for 15 min at room temperature and then treated fusion after scrape loading were measured in at least
with 0.1% Triton X-100 in PBS. After a brief wash with twenty random areas from each sample, and the fluo-
PBS, the cells were incubated with blocking buffer rescence intensity was assessed using Image J software
(5% normal goat serum and 0.1% Tween 20 in PBS) and compared between the control and infected cells
for 1 h, followed by incubation with rabbit polyclonal with or without each treatment.
anti-Cx43 antibody (1:200, Cell Signaling Technol-
ogy) and mouse monoclonal anti-PrP (3F10, 1:200) Brilliant blue G (BBG) treatment
[38] overnight at 4◦ C. The cells were subsequently
incubated with either Alexa Fluor 568 goat anti-rabbit Uninfected and 22L scrapie-infected cells (1 × 106
IgG or Alexa Fluor 488 goat anti-mouse IgG antibod- cells/100-mm dish) were treated or not treated with
ies (Invitrogen, Carlsbad, CA, USA) for 1 h. Control BBG (0.6–60 ␮M) for 3 days and then lysed. The
reactions omitting the primary antibodies resulted in expression levels of PrPSc , JNK, phospho-JNK, and
no labeling with the secondary antibodies (data not Cx43 were determined by western blotting as previ-
shown). After rinsing with PBS, the cells were then ously described.
mounted in 4 ,6-diamidino-2-phenylindole (DAPI)-
containing Vectashield Mounting Medium (Vector Statistical analysis
Laboratories) to label nuclei and visualized using a
confocal laser scanning microscope (LSM 700; Carl The data are expressed as the mean ± SEM. Sig-
Zeiss, Oberkochen, Germany). nificant differences between the experimental groups
were evaluated using one-way analysis of variance
Dye uptake assay (ANOVA). Statistical significance was defined as
∗ p < 0.05, ∗∗ p < 0.01, and ∗∗∗ p < 0.001.

To evaluate the functional Cx43 hemichannel,


an uptake assay using the hemichannel-permeable RESULTS
reporter dye ethidium bromide (EtBr) was performed
as previously described [42]. The cells were incu- Upregulation of Cx43 expression in
bated with 5 ␮M EtBr in PBS in the presence or scrapie-infected mouse brains
absence of a connexin hemichannel blocker LaCl3
(500 ␮M) for 5 min at 37◦ C. For JNK inhibition, the We first investigated the expression levels of the
cells were pretreated with the JNK inhibitor SP600125 Cx43 protein in the brain tissues of control mice and
(50 ␮M) for 6 h and then incubated with 5 ␮M EtBr mice infected with one of three scrapie strains (ME7,
for 5 min at 37◦ C. The cells were then washed with 22L, or 139A). These strains possess distinct incuba-
Hank’s balanced salt solution (HBSS), fixed with 4% tion periods and neuropathological features [44]. In the
paraformaldehyde in PBS for 15 min at room tempera- whole brain and dissected brain tissues, including the
ture and washed with HBSS. Dye uptake was examined cerebral cortex, hippocampus, striatum, cerebellum,
with a confocal laser scanning microscope. and brain stem, we demonstrated that Cx43 protein
Fig. 1A, B) and mRNA (Fig. 1C) were highly expressed
Scrape-loading dye transfer assay in animals with scrapie infection compared with the
controls. These results suggest that both the mRNA
To determine the functional Cx43 hemichannels, an and protein levels of Cx43 are upregulated after scrapie
uptake assay using the gap junction permeable reporter infection, and the upregulated protein levels of Cx43
dye Lucifer yellow (LY) (Sigma-Aldrich) was per- in most brain regions are a result of increased gene
formed as previously described [43]. The cells were expression in scrapie-infected mice.
incubated with 0.01% LY in the presence or absence To further determine whether the increased Cx43
of the Cx channel blocker lanthanum chloride (LaCl3 , protein expression is correlated with disease pro-
500 ␮M) for 5 min at 37◦ C. For JNK inhibition, the gression, we performed western blotting using whole
cells were pretreated with a JNK inhibitor SP600125 brains obtained at 50, 100, and 150 dpi for the ME7
(50 ␮M) for 6 h and then incubated with 0.01% LY strain (for mice) and at 10, 30, 60, and 90 dpi for the
in PBS for 5 min at 37◦ C. The cells were washed 263K strain (for hamsters) (Fig. 1D). No difference
G.-H. Lee et al. / Scrapie Infection-Mediated Cx43 Upregulation 1009

Fig. 1. Increased expression of Cx43 protein and mRNA in the brains of scrapie-infected mice. A) Cx43 protein expression in whole brain
and in dissected brains of the control and ME7 (160 dpi), 22 L (140 dpi), and 139A (170 dpi) scrapie-infected mice was evaluated by western
blot with anti-Cx43 antibody. ␤-actin was used as a loading control. B) The intensities of the Cx43 bands in panel A were measured and
quantified. The values are expressed as the mean ± SEM (n = 3). C) In whole brains, the Cx43 mRNA levels were analyzed by RT-PCR with
three individuals per group. The error bars represent the SEM. D) Cx43 protein expression was analyzed in the whole brains of the control and
scrapie-infected mice by western blot at the indicated time points after inoculation. ␤-actin was used as a loading control, and PrPSc was detected
using anti-PrP antibody after PK (50 ␮g/ml) treatment. Each experiment was repeated at least three times, and similar results were obtained.
∗ p < 0.05, ∗∗ p < 0.01, ∗∗∗ p < 0.001.

between the control and scrapie-infected brains was increase relative to the stage of scrapie development
identified at 50 dpi in ME7 mice or in 263K hamsters and was significantly increased at 90 dpi (for ham-
at 30 dpi. However, Cx43 protein expression tended to sters) and 150 dpi (for mice). These time points were
1010 G.-H. Lee et al. / Scrapie Infection-Mediated Cx43 Upregulation

Fig. 2. Immunohistochemical staining of Cx43 in brain sections from control and scrapie-infected mice. Cx43 protein was detected in various
brain sections obtained from the control and ME7, 22 L and 139A scrapie-infected mice at 150 days post-inoculation. Arrows, Cx43-positive
cell-cell cotact or blood vessels. Scale bar, 25 ␮m.

correlated with marked accumulations of PrPSc , indi- of cell-cell contact or blood vessels (arrows in Fig. 2).
cating that Cx43 protein increased at the end stage of Overall, these findings suggest that scrapie infection
scrapie infection. increases Cx43 expression in the brain.
To confirm the increase in Cx43 after scrapie
infection, we subsequently performed immunohisto- Increased Cx43 in the membrane fraction from the
chemical staining of Cx43 in various brain regions brains of scrapie-infected mice
of the control and scrapie-infected mice. Cx43 was
prominent in most brain regions, including the cere- Because Cx43 is an integral plasma membrane pro-
bral cortex, hippocampus, striatum, cerebellum, and tein, we subsequently examined whether the increased
brain stem, at the end stage of scrapie infection com- Cx43 is related to membrane localization using cytoso-
pared with levels in the controls (Fig. 2). A marked lic and membrane fractions prepared from control
increase in Cx43 immunoreactivity was seen in the brains and from ME7, 22L, and 139A scrapie-infected
hippocampus and the Purkinje layer of the cerebellum brains as described in the Materials and Methods.
in the scrapie-infected mice. In addition, Cx43, which Calnexin and enolase were used as markers of the
showed a diffuse staining pattern, localized to regions membrane and cytosolic fractions, respectively. As
G.-H. Lee et al. / Scrapie Infection-Mediated Cx43 Upregulation 1011

Association of JNK activation and Cx43


upregulation in scrapie-infected cells and mice

JNK is a stress-activated kinase reported to be


important in intracellular signaling pathways that reg-
ulate Cx43 expression [45–47]. To determine whether
JNK signaling is involved in the induction of Cx43, we
examined the activation of JNK and its downstream
Fig. 3. Increased Cx43 protein in membrane fractions of scrapie- molecule c-Jun in scrapie-infected neuronal cells and
infected brains. Whole brains were lysed, separated into cytosol
and membrane fractions, and subjected to western blot analysis
mice. Scrapie infection was confirmed by the detec-
as described in the Materials and Methods. Each fraction of the tion of PK-resistant PrPSc (Fig. 5A, second panels).
brain homogenates was evaluated using anti-calnexin as a membrane Interestingly, Cx43 was upregulated and associated
marker and anti-enolase as a cytosolic marker. with JNK activation, which was demonstrated by
increased phospho-JNK (P-JNK) in scrapie-infected
expected, the Cx43 protein was preferentially detected neuronal cells and in infected brains (Fig. 5A). The
in the membrane fraction of all scrapie-infected brains levels of P-c-Jun were also significantly increased in
compared with levels in the controls (Fig. 3), which is association with JNK activation; however, the total
consistent with the immunoblot analysis data (Fig. 1). levels of JNK and c-Jun were not altered. To further
These data suggest that increases in membrane- elucidate the functional roles of JNK in the induction
associated Cx43 protein result from scrapie-induced of Cx43 by scrapie infection, control and infected cell
pathological conditions. lines were treated with various concentrations of an
ATP-competitive inhibitor of JNK, SP600125
Increased Cx43 expression in scrapie-infected (0–100 ␮M), for 6 h. As shown in Fig. 5B and C,
hippocampal cell lines increased Cx43 and P-JNK/P-c-Jun by scrapie infec-
tion were effectively downregulated by SP600125
Gap junctions are complexes of intercellular chan- treatment in a dose- and time-dependent manner.
nels between adjacent cells [1]. We subsequently These findings suggest that JNK-c-Jun signaling is
examined whether a scrapie infection-induced increase essential for the Cx43 overexpression induced by
in Cx43 expression is associated with gap junction scrapie infection.
plaque formation in hippocampal neuronal cell lines,
as previously established [39]. Although Cx43 is a Increased dye uptake by scrapie-induced Cx43
primary gap junction protein in astrocytes, we demon- expression
strated that the ZW13-2 and Zpl2-4 hippocampal
neuronal cell lines endogenously express Cx43. Thus, The uptake of the fluorescent dye EtBr has been
these cell lines were used as an in vitro model of prion used as an indicator of hemichannel opening [42, 48].
replication after infection with either of two mouse- To determine whether Cx43 upregulation by scrapie
derived scrapie strains (22L or 139A). As shown in infection affects the opening of Cx43 hemichannels,
Fig. 4, Cx43 (red) was detected in both ZW13-2 and we performed EtBr uptake assays in scrapie-infected
Zpl2-4 cells, and intense expression of Cx43 was and un-infected ZW13-2 neuronal cells. As shown in
mainly localized within plaques at the cell-cell contact Fig. 6 enhanced EtBr uptake was observed in both the
areas. In the presence of PrP (ZW13-2), Cx43 staining 22L and 139A scrapie-infected cells compared with
consisted of sparse but large puncta distinctly sepa- the control cells. The scrapie infection-enhanced EtBr
rated from the nucleus (blue). Interestingly, Cx43 was uptake was inhibited in the presence of LaCl3 (500 ␮M;
clearly detected with more aggregates between neigh- a known connexin hemichannel blocker) and the JNK
boring scrapie-infected ZW13-2 cells but not between inhibitor SP600125, indicating that scrapie infection-
Zpl2-4 cells or uninfected cells. Uptake of infection mediated upregulation of Cx43 controls the functional
was confirmed by the PK-resistant PrPSc levels in state of hemichannels.
cultures after multiple passages (Supplementary Fig- The scrape-loading dye transfer technique is used to
ure 1). These results demonstrated that pathogenic determine the intrinsic gap junction intercellular com-
PrP or scrapie infection induce the upregulation of munication (GJIC) [43, 49]. Next, scrapie-infected and
Cx43 expression and the formation of gap junction-like un-infected ZW13-2 neuronal cells were subjected to
plaques in neuronal cells. a scrape-loading dye transfer assay in the absence or
1012 G.-H. Lee et al. / Scrapie Infection-Mediated Cx43 Upregulation

Fig. 4. Subcellular distribution of Cx43 and PrP in hippocampal cell lines with or without scrapie infection. Double immunofluorescence
staining was carried using anti-Cx43 (red) and anti-PrP (green) in the PrP knockout (Zpl2-4) and wild-type (ZW13-2) hippocampal neuronal
cell lines with or without the 22 L or 139A scrapie infection. DAPI was used for nuclear staining. Note that Cx43 was mainly localized in plaque
formations at the cell-cell contact areas (merged and bright-field images). Arrows indicate parts of the gap junctions. Scale bars, 20 ␮m.

presence of LaCl3 and a JNK inhibitor as described in Suppression of Cx43 expression with decreased
the Materials and Methods. As shown in Fig. 7A (top JNK activity by BBG-mediated blockade of prion
panels), gap junction-mediated intercellular transfer of conversion
LY was increased in the 22L and 139A scrapie-infected
cells compared with the control cells. This increase was To further investigate whether Cx43 induction is
abolished by either hemichannel inhibition (LaCl3 ) regulated by PrPSc accumulation, we conducted a
or JNK inhibition (SP600125) (Fig. 7A, middle and blocking experiment of PrPSc accumulation using
bottom panels, respectively). Hemichannel inhibition BBG, which has anti-prion activity through the
almost blocked GJIC in either control or infected cells inhibition of PrPSc accumulation [50]. Un-infected
(Fig. 7A middle panels), whereas JNK inhibition main- and 22L scrapie-infected ZW13-2 neuronal cells
tained basal levels of GJIC in control and infected cells were incubated with various concentrations of BBG
(Fig. 7A ottom panels) without a significant differ- (0–60 ␮M) for 3 days, and the expression lev-
ence between control and infected cells. The distances els of Cx43, PrPSc , and p-JNK were subsequently
of LY diffusion were measured and compared with measured. As shown in Fig. 8 BBG treatment
those in the controls and infected cells with or with- efficiently inhibited PrPSc accumulation in a dose-
out treatments (Fig. 7B). These results indicate that dependent manner. In correlation with the decrease
the upregulation of Cx43 expression by scrapie infec- in PrPSc , the expression levels of Cx43 and p-JNK
tion represents the efficient communication capacity were gradually reduced following BBG treatment of
of Cx43 hemichannels and GJIC and likely occurs via 22L scrapie-infected cells. In contrast, no changes
JNK activation. were observed in the uninfected cells treated with
G.-H. Lee et al. / Scrapie Infection-Mediated Cx43 Upregulation 1013

Fig. 5. JNK activation-mediated Cx43 upregulation in scrapie-infected cells and mice. A) Increased Cx43 expression and JNK activation in
control and scrapie-infected ZW13-2 cells (left panels) and mouse brains (right panels). To detect PrPSc , cell lysates and brain homogenates
were treated for 30 min at 37◦ C with PK at 50 ␮g/ml or 20 ␮g/ml, respectively. B, C) JNK inhibition attenuates Cx43 expression in a dose-
(B) and time-dependent (C) manner. Control, 22L-, and 139A-infected ZW13-2 cells were treated with 0, 25, 50, and 100 ␮M of the JNK
inhibitor SP600125 for 6 h (B) or with 50 ␮M SP600125 for the time indicated (C). Each protein was analyzed by western blot with the indicated
antibodies.

BBG. These results suggest that PrPSc accumulation Under various pathological conditions, astrocytes
may be responsible for JNK signaling-mediated Cx43 become reactive, triggering a long-lasting process
upregulation. that comprises complex phenotypic changes [56].
Astrocyte activation, in which there is both hypertro-
DISCUSSION phy and proliferation, involves changes in the cellular
phenotype and in the expression of transporters, recep-
In the present study, we demonstrated that the upreg- tors and ion channels, which can be deleterious for
ulation of Cx43 expression, which controls functional neurons and thus lead to neuronal cell death [57]. These
properties of hemichannels, is mediated by JNK acti- changes can explain how connexins can undergo both
vation and correlated with PrPSc accumulation in both up- and downregulation depending on the type of brain
in vivo and in vitro systems of prion diseases. These pathology, nature of the injury, time scale and distance
findings represent the first demonstration of the link from the lesioned area [58, 59].
between the Cx43 protein and prion pathogenesis. Continuous astrocyte-neuron interactions are
Cx43 hemichannels contribute to cell death and are required to maintain Cx30 expression and increased
responsible for tissue damage [11, 13, 15, 18, 20, 33, Cx43 levels in astrocytes [60]. The expression level of
35, 51]. Several signaling molecules are linked to brain Cx43 and its hemichannel activity were significantly
inflammation and unwarranted cell death. It is conceiv- increased in scrapie-infected brains and in cultured
able that the excessive diffusion of neurotransmitters hippocampal cells, even though the neuronal loss
or ions, such as glutamate, ATP, and Ca2+ , from dam- associated with prion diseases reduces the level of
aged cells to adjacent normal cells via Cx43-promoted Cx43 in astrocytes associated with the lost neurons.
gap junctions may accelerate this pathophysiological In our study, it is difficult to identify which cell
process [8–10]. Astrocytes and microglia release gluta- type(s) is mainly stained for Cx43. However, it is
mate via gap junctions [8, 52–54], which then induces possible that astroglial Cx43 upregulation may be
neuronal cell death [54, 55]. caused by scrapie infection-mediated pathological
1014 G.-H. Lee et al. / Scrapie Infection-Mediated Cx43 Upregulation

Fig. 6. Increased EtBr uptake in scrapie-infected cells. An EtBr uptake assay was performed using control or scrapie-infected ZW13-2 cells.
Cells were pretreated with 500 ␮M LaCl3 for 5 min (middle panels) or 50 ␮M of the JNK inhibitor SP600125 for 6 h (bottom panels) and then
incubated with 5 ␮M EtBr for 5 min at 37◦ C; the fluorescent signal was then analyzed using a confocal microscope.

changes since reactive astrocytes are one of the major the limited involvement of functional Cx43 expression
pathological changes associated with prion diseases. in the glial cells from this study.
Using previously established hippocampal neuronal According to previous findings, the increased
cell lines [39], increased Cx43 was observed following production of reactive oxygen species and pro-
inoculation with two independent scrapie strains. inflammatory cytokines, including IL-1␣, IL-1␤, and
Although Cx43 is primarily expressed in astrocytes TNF-␣, in the brains of scrapie-infected mice can
[4, 60, 61] and astrocyte gap junctions coupled with lead astrocytes to become reactive [62]. These astro-
CNS cells [59], these neuronal cell lines are a useful cytes can be well-coupled via gap junctions, leading
in vitro system to study the functional role of Cx43 to increases in hemichannel activity because of the
in prion diseases because they express endogenous increased Cx43 mRNA and protein levels in scrapie-
Cx43. In addition, our data suggest that scrapie infected brains. Consistent with this phenomenon,
infection-enhanced Cx43 proteins may consistently Cx43 was associated with gap junctional plaques via
exacerbate the pathogenesis of prion diseases despite distribution to neighboring cells after scrapie infec-
G.-H. Lee et al. / Scrapie Infection-Mediated Cx43 Upregulation 1015

Fig. 7. Increased scrape-loading dye transfer in scrapie-infected cells. Control or scrapie-infected ZW13-2 cells were incubated with 0.1%
Lucifer yellow (LY) for 5 min at 37◦ C (top panels) or with 500 ␮M LaCl3 treatment (middle panels). For JNK inhibition, the cells were
pretreated with the JNK inhibitor SP600125 (50 ␮M) for 6 h prior to LY treatment (bottom panels). LY fluorescence was determined using a
confocal microscope (A) and mean length of LY-filled area (the distance from the center to the edge, n = 20) was represented by bars (B). The
arrow indicates scrape; lines drawn in upper panels indicate the distance for dye transfer. ∗∗∗ p < 0.001. Scale bar, 50 ␮m.
1016 G.-H. Lee et al. / Scrapie Infection-Mediated Cx43 Upregulation

astrocytes [65]. Thus, the functional role of Cx43 and


pannexin in the pathogenesis of prion diseases remains
unresolved.
In addition, previous reports have demonstrated that
JNK signaling activation is involved in the pathogen-
esis of scrapie-infected hamster brains [66] and JNK
signaling cascades are necessary for Cx43 expression
[45–47]. JNK regulates downstream gene expression
through the activation or inactivation of various tran-
scriptional factors, such as c-Jun, c-fos, and SP1 [67].
We also demonstrated that the activated JNK/c-Jun cas-
cade is responsible for Cx43 expression in both in vitro
Fig. 8. Attenuation of JNK signaling-mediated Cx43 upregula- and in vivo models of prion disease. Surprisingly, the
tion by inhibiting the accumulation of PrPSc . Control and 22 L accumulation of pathogenic PrPSc appears to occur
scrapie-infected ZW13-2 cells were treated with various concen-
trations of Brilliant blue G (BBG; 0-60 ␮M) for 3 days, and upstream of JNK activation, which is then followed
the expression levels of Cx43, phosphorylated JNK, and PrPSc by increased Cx43 expression. Although PrP did not
were analyzed by western blot with the indicated antibodies. co-localize in Cx43 plaque formation, scrapie infec-
To detect PrPSc , the total cell lysates were treated with 50 ␮g/ml
tion increased Cx43 aggregates at the cell-cell contact
of PK for 30 min at 37◦ C. ␤-actin was used as a loading
control. areas. Therefore, PrPSc accumulation may be respon-
sible for the upregulation of Cx43 expression and gap
junction formation.
tion (Fig. 4). Although increases in Cx43 and gap Overall, for the first time, these results demon-
junctional communication and their effects on neu- strated that scrapie infection activates JNK signaling
ronal death and inflammatory responses have been cascades to enhance Cx43 and gap junctions in neu-
linked to chronic and progressive neurodegenerative ronal cells and in the brain. Pathogenic PrPSc (or
diseases such as stroke, Alzheimer’s disease, Parkin- scrapie)-mediated upregulation of Cx43 through JNK
son’s disease, and Huntington’s disease [57], it remains activation may exacerbate prion pathology, includ-
a matter of debate whether Cx43 is neuroprotective or ing neuronal death. Although blocking gap junctions,
deleterious. which inhibit the diffusion of neurotoxic molecules,
Accumulating evidence has indicated that connexins has been proposed as a candidate therapy for var-
act as phosphoproteins via a shift in their elec- ious neurodegenerative diseases, it can also limit
trophoretic mobility or direct incorporation of 32 P [63]. essential physiological signals. Thus, this strategy has
The JNK signaling cascade is an important intracellu- both neurotoxic and neuroprotective effects on dis-
lar signaling pathway that regulates Cx43 [47]. Our ease progression. A previous report demonstrated that
data demonstrated that upregulated Cx43 was a result a JNK inhibitor reduced PrP106-126 peptide-induced
of JNK activation and was blocked by SP600125, a neuronal apoptosis [68]. Therefore, we suggest that
potent inhibitor of JNK. In this study, the assays used blocking the gap junctions (functional Cx hemichan-
to measure Cx43 predominately identified a single nels) expressed by neurons, astrocytes, and microglia
band, and we were unable to distinguish the phos- in the region of prion propagation might represent a
phorylation status of Cx43. Because Cx43 can be novel strategy to reduce disease phenotypes in prion
phosphorylated by various kinases, the activation of a diseases.
specific kinase may not correlate with Cx43 phospho-
rylation in our models. A recent study has suggested
that astroglial Cx43 plays a protective role in oxidative ACKNOWLEDGMENTS
stress-induced cell death, which depends on the phos-
phorylation state of Cx43 [17]. In addition, the opening We thank Dr. Joy J. Goto (California State Uni-
of hemichannels formed by pannexin 1, the mam- versity, Fresno, USA) for critical reading of this
malian ortholog of the invertebrate gap junction protein paper and helpful discussions. This work was sup-
innexin, has also been implicated in neuronal death ported by the National Research Foundation of Korea
after ischemia [64]. Moreover, during the astroglio- (NRF) grant funded by the Korean Government (NRF-
sis response observed 24 h after reperfusion, de novo 2013R1A1A2007071) and by the Hallym University
synthesis of pannexin 2 occurs in hippocampal rat Specialization Fund (HRF-S-41).
G.-H. Lee et al. / Scrapie Infection-Mediated Cx43 Upregulation 1017

Authors’ disclosures available online (http://j-alz. [16] Giardina SF, Mikami M, Goubaeva F, Yang J (2007) Con-
com/manuscript-disclosures/15-0283r1). nexin 43 confers resistance to hydrogen peroxide-mediated
apoptosis. Biochem Biophys Res Commun 362, 747-752.
[17] Le HT, Sin WC, Lozinsky S, Bechberger J, Vega JL, Guo XQ,
SUPPLEMENTARY MATERIAL Saez JC, Naus CC (2014) Gap junction intercellular commu-
nication mediated by connexin43 in astrocytes is essential
for their resistance to oxidative stress. J Biol Chem 289,
The supplementary material is available in the elec-
1345-1354.
tronic version of this article: http://dx.doi.org/10.3233/ [18] Cronin M, Anderson PN, Cook JE, Green CR, Becker DL
JAD-150283. (2008) Blocking connexin43 expression reduces inflamma-
tion and improves functional recovery after spinal cord injury.
Mol Cell Neurosci 39, 152-160.
REFERENCES [19] Chen MJ, Kress B, Han X, Moll K, Peng W, Ji RR, Nedergaard
M (2012) Astrocytic CX43 hemichannels and gap junctions
[1] Goodenough DA, Paul DL (2009) Gap junctions. Cold Spring play a crucial role in development of chronic neuropathic pain
Harb Perspect Biol 1, a002576. following spinal cord injury. Glia 60, 1660-1670.
[2] Bruzzone R, Dermietzel R (2006) Structure and function of [20] Davidson JO, Green CR, Nicholson LF, O’Carroll SJ, Fraser
gap junctions in the developing brain. Cell Tissue Res 326, M, Bennet L, Gunn AJ (2012) Connexin hemichannel block-
239-248. ade improves outcomes in a model of fetal ischemia. Ann
[3] Elias LA, Kriegstein AR (2008) Gap junctions: Multifaceted Neurol 71, 121-132.
regulators of embryonic cortical development. Trends Neu- [21] Siushansian R, Bechberger JF, Cechetto DF, Hachinski VC,
rosci 31, 243-250. Naus CC (2001) Connexin43 null mutation increases infarct
[4] Dermietzel R, Hertberg EL, Kessler JA, Spray DC (1991) Gap size after stroke. J Comp Neurol 440, 387-394.
junctions between cultured astrocytes: Immunocytochemical, [22] Kozoriz MG, Bechberger JF, Bechberger GR, Suen MW,
molecular, and electrophysiological analysis. J Neurosci 11, Moreno AP, Maass K, Willecke K, Naus CC (2010) The con-
1421-1432. nexin43 C-terminal region mediates neuroprotection during
[5] Nagy JI, Dudek FE, Rash JE (2004) Update on connexins and stroke. J Neuropathol Exp Neurol 69, 196-206.
gap junctions in neurons and glia in the mammalian nervous [23] Santiago MF, Alcami P, Striedinger KM, Spray DC, Scemes
system. Brain Res Brain Res Rev 47, 191-215. E (2010) The carboxyl-terminal domain of connexin43 is a
[6] Nualart-Marti A, Solsona C, Fields RD (2013) Gap junction negative modulator of neuronal differentiation. J Biol Chem
communication in myelinating glia. Biochim Biophys Acta 285, 11836-11845.
1828, 69-78. [24] Lemcke H, Kuznetsov SA (2013) Involvement of connexin43
[7] Stout CE, Costantin JL, Naus CC, Charles AC (2002) in the EGF/EGFR signalling during self-renewal and differen-
Intercellular calcium signaling in astrocytes via ATP tiation of neural progenitor cells. Cell Signal 25, 2676-2684.
release through connexin hemichannels. J Biol Chem 277, [25] Rinaldi F, Hartfield EM, Crompton LA, Badger JL, Glover
10482-10488. CP, Kelly CM, Rosser AE, Uney JB, Caldwell MA (2014)
[8] Ye ZC, Wyeth MS, Baltan-Tekkok S, Ransom BR (2003) Cross-regulation of Connexin43 and ␤-catenin influences dif-
Functional hemichannels in astrocytes: A novel mechanism ferentiation of human neural progenitor cells. Cell Death Dis
of glutamate release. J Neurosci 23, 3588-3596. 5, e1017.
[9] Lurtz MM, Louis CF (2007) Intracellular calcium regulation [26] Huang GY, Cooper ES, Waldo K, Kirby ML, Gilula NB, Lo
of connexin43. Am J Physiol Cell Physiol 293, C1806-C1813. CW (1998) Gap junction-mediated cell-cell communication
[10] Kang J, Kang N, Lovatt D, Torres A, Zhao Z, Lin J, Neder- modulates mouse neural crest migration. J Cell Biol 143,
gaard M (2008) Connexin 43 hemichannels are permeable to 1725-1734.
ATP. J Neurosci 28, 4702-4711. [27] Cina C, Maass K, Theis M, Willecke K, Bechberger JF,
[11] Orellana JA, Froger N, Ezan P, Jiang JX, Bennett MV, Naus Naus CC (2009) Involvement of the cytoplasmic C-terminal
CC, Giaume C, Saez JC (2011a) ATP and glutamate released domain of connexin43 in neuronal migration. J Neurosci 29,
via astroglial connexin 43 hemichannels mediate neuronal 2009-2021.
death through activation of pannexin 1 hemichannels. J Neu- [28] Behrens J, Kameritsch P, Wallner S, Pohl U, Pogoda K (2010)
rochem 118, 826-840. The carboxyl tail of Cx43 augments p38 mediated cell migra-
[12] Orellana JA, Martinez AD, Retamal MA (2013) Gap junc- tion in a gap junction-independent manner. Eur J Cell Biol
tion channels and hemichannels in the CNS: Regulation by 89, 828-838.
signaling molecules. Neuropharmacology 75, 567-582. [29] Liu X, Sun L, Torii M, Rakic P (2012) Connexin 43 controls
[13] Orellana JA, Shoji KF, Abudara V, Ezan P, Amigou E, the multipolar phase of neuronal migration to the cerebral
Sáez PJ, Jiang JX, Naus CC, Sáez JC, Giaume C (2011b) cortex. Proc Natl Acad Sci U S A 109, 8280-8285.
Amyloid ␤-induced death in neurons involves glial and neu- [30] Liebmann M, Stahr A, Guenther M, Witte OW, Frahm C
ronal hemichannels. J Neurosci 31, 4962-4977. (2013) Astrocytic Cx43 and Cx30 differentially modulate
[14] Ishii Y, Shintani-Ishida K, Yoshida K (2013) Connexin-43 adult neurogenesis in mice. Neurosci Lett 17, 40-45.
hemichannels contribute to the propagation of ␮-calpain- [31] Nagy JI, Li W, Hertzberg EL, Marotta CA (1996) Elevated
mediated neuronal death in a cortical ablation injury model. connexin43 immunoreactivity at sites of amyloid plaques in
Biochem Biophys Res Commun 441, 457-462. Alzheimer’s disease. Brain Res 717, 178.
[15] Frantseva MV, Kokarovtseva L, Perez Velazquez JL (2002) [32] Mei X, Ezan P, Giaume C, Koulakoff A (2010) Astroglial con-
Ischemia-induced brain damage depends on specific gap- nexin immunoreactivity is specifically altered at ␤-amyloid
junctional coupling. J Cereb Blood Flow Metab 22, plaques in ␤-amyloid precursor protein/presenilin1 mice.
453-462. Neuroscience 171, 92-105.
1018 G.-H. Lee et al. / Scrapie Infection-Mediated Cx43 Upregulation

[33] Cui Y, Masaki K, Yamasaki R, Imamura S, Suzuki SO, [48] Contreras JE, Sáez JC, Bukauskas FF, Bennett MV (2003)
Hayashi S, Sato S, Nagara Y, Kawamura MF, Kira J (2014) Gating and regulation of connexin 43 (Cx43) hemichannels.
Extensive dysregulations of oligodendrocytic and astrocytic Proc Natl Acad Sci U S A 100, 11388-11393.
connexins are associated with disease progression in an amy- [49] McKarns SC, Doolittle DJ (1992) Limitations of the scrape-
otrophic lateral sclerosis mouse model. J Neuroinflammation loading/dye transfer technique to quantify inhibition of gap
11, 42. junctional intercellular communication. Cell Biol Toxicol 8,
[34] Rufer M, Wirth SB, Hofer A, Dermietzel R, Pastor A, 89-103.
Kettenmann H, Unsicker K (1996) Regulation of connexin- [50] Iwamaru Y, Takenouchi T, Murayama Y, Okada H, Imamura
43, GFAP, and FGF-2 is not accompanied by changes in M, Shimizu Y, Hashimoto M, Mohri S, Yokoyama T, Kitani
astroglial coupling in MPTP-lesioned, FGF-2-treated parkin- H (2012) Anti-prion activity of Brilliant Blue G. PLoS One
sonian mice. J Neurosci Res 46, 606-617. 7, e37896.
[35] Fonseca CG, Green CR, Nicholson LF (2002) Upregulation [51] Frantseva MV, Kokarovtseva L, Naus CG, Carlen PL, Mac-
in astrocytic connexin 43 gap junction levels may exacerbate Fabe D, Perez Velazquez JL (2002b) Specific gap junctions
generalized seizures in mesial temporal lobe epilepsy. Brain enhance the neuronal vulnerability to brain traumatic injury.
Res 929, 105-116. J Neurosci 22, 644-653.
[36] Masaki K, Suzuki SO, Matsushita T, Matsuoka T, Imamura [52] Parpura V, Scemes E, Spray DC (2004) Mechanisms of gluta-
S, Yamasaki R, Suzuki M, Suenaga T, Iwaki T, Kira J (2013) mate release from astrocytes: Gap junction “hemichannels”,
Connexin 43 astrocytopathy linked to rapidly progressive purinergic receptors and exocytotic release. Neurochem Int
multiple sclerosis and neuromyelitis optica. PLoS One 8, 45, 259-264.
e72919. [53] Takeuchi H, Jin S, Wang J, Zhang G, Kawanokuchi J, Kuno
[37] Prusiner SB (1998) Prions. Proc Natl Acad Sci U S A 95, R, Sonobe Y, Mizuno T, Suzumura A (2006) Tumor necrosis
13363-13383. factor-alpha induces neurotoxicity via glutamate release from
[38] Choi JK, Park SJ, Jun YC, Oh JM, Jeong BH, Lee HP, Park SN, hemichannels of activated microglia in an autocrine manner.
Carp RI, Kim YS (2006) Generation of monoclonal antibody J Biol Chem 281, 21362-21368.
recognized by the GXXXG motif (glycine zipper) of prion [54] Takeuchi H, Jin S, Suzuki H, Doi Y, Liang J, Kawanokuchi
protein. Hybridoma (Larchmt) 25, 271-277. J, Mizuno T, Sawada M, Suzumura A (2008) Blockade of
[39] Kim BH, Kim JI, Choi EK, Carp RI, Kim YS (2005) A neu- microglial glutamate release protects against ischemic brain
ronal cell line that does not express either prion or doppel injury. Exp Neurol 214, 144-146.
proteins. Neuroreport 16, 465-429. [55] Ridet JL, Malhotra SK, Privat A, Gage FH (1997) Reactive
[40] Harrison CF, Lawson VA, Coleman BM, Kim YS, Masters astrocytes: Cellular and molecular cues to biological function.
CL, Cappai R, Barnham KJ, Hill AF (2010) Conservation of Trends Neurosci 20, 570-577.
a glycine-rich region in the prion protein is required for uptake [56] Yawata I, Takeuchi H, Doi Y, Liang J, Mizuno T, Suzumura
of prion infectivity. J Biol Chem 285, 20213-20223. A (2008) Macrophage-induced neurotoxicity is mediated by
[41] Jang B, Jin JK, Jeon YC, Cho HJ, Ishigami A, Choi KC, Carp glutamate and attenuated by glutaminase inhibitors and gap
RI, Maruyama N, Kim YS, Choi EK (2010) Involvement of junction inhibitors. Life Sci 82, 1111-1116.
peptidylarginine deiminase-mediated post-translational cit- [57] Orellana JA, Sáez PJ, Shoji KF, Schalper KA, Palacios-Prado
rullination in pathogenesis of sporadic Creutzfeldt-Jakob N, Velarde V, Giaume C, Bennett MV, Sáez JC (2009) Mod-
disease. Acta Neuropathol 119, 199-210. ulation of brain hemichannels and gap junction channels by
[42] Orellana JA, Hernández DE, Ezan P, Velarde V, Bennett MV, pro-inflammatory agents and their possible role in neurode-
Giaume C, Sáez JC (2010) Hypoxia in high glucose followed generation. Antioxid Redox Signal 11, 369-399.
by reoxygenation in normal glucose reduces the viability of [58] Rouach N, Avignone E, Même W, Koulakoff A, Venance L,
cortical astrocytes through increased permeability of con- Blomstrand F, Giaume C (2002) Gap junctions and connexin
nexin 43 hemichannels. Glia 58, 329-343. expression in the normal and pathological central nervous
[43] Krutovskikh VA, Troyanovsky SM, Piccoli C, Tsuda H, system. Biol Cell 94, 457-475.
Asamoto M, Yamasaki H (2000) Differential effect of subcel- [59] Giaume C, Theis M (2010) Pharmacological and genetic
lular localization of communication impairing gap junction approaches to study connexin-mediated channels in glial cells
protein connexin43 on tumor cell growth in vivo. Oncogene of the central nervous system. Brain Res Rev 63, 160-176.
19, 505-513. [60] Koulakoff A, Ezan P, Giaume C (2008) Neurons control the
[44] Bruce ME, McConnell I, Fraser H, Dickinson AG (1991) The expression of connexin 30 and connexin 43 in mouse cortical
disease characteristics of different strains of scrapie in Sinc astrocytes. Glia 56, 1299-1311.
congenic mouse lines: Implications for the nature of the agent [61] Dermietzel R, Traub O, Hwang TK, Beyer E, Bennett MV,
and host control of pathogenesis. J Gen Virol 72, 595-603. Spray DC, Willecke K (1989) Differential expression of three
[45] Shyu KG, Wang BW, Yang YH, Tsai SC, Lin S, Lee CC gap junction proteins in developing and mature brain tissues.
(2004) Amphetamine activates connexin43 gene expression in Proc Natl Acad Sci U S A 86, 10148-10152.
cultured neonatal rat cardiomyocytes through JNK and AP-1 [62] Kim JI, Ju WK, Choi JH, Choi E, Carp RI, Wisniewski HM,
pathway. Cardiovasc Res 63, 98-108. Kim YS (1999) Expression of cytokine genes and increased
[46] Gao FH, Wang Q, Wu YL, Li X, Zhao KW, Chen GQ (2007) c- nuclear factor-kappa B activity in the brains of scrapie-
Jun N-terminal kinase mediates AML1-ETO protein-induced infected mice. Brain Res Mol Brain Res 73, 17-27.
connexin-43 expression. Biochem Biophys Res Commun 356, [63] Lampe PD, Lau AF (2004) The effects of connexin phospho-
505-511. rylation on gap junctional communication. Int J Biochem Cell
[47] Li K, Chi Y, Gao K, Yan Q, Matsue H, Takeda M, Kitamura M, Biol 36, 1171-1186.
Yao J (2013) Connexin43 hemichannel-mediated regulation [64] Thompson RJ, Zhou N, MacVicar BA (2006) Ischemia opens
of connexin43. PLoS One 8, e58057. neuronal gap junction hemichannels. Science 312, 924-927.
G.-H. Lee et al. / Scrapie Infection-Mediated Cx43 Upregulation 1019

[65] Zappalà A, Li Volti G, Serapide MF, Pellitteri R, Falchi M, connexin 43 proximal promoter in neonatal cardiomyocytes.
La Delia F, Cicirata V, Cicirata F (2007) Expression of pan- Gene 322, 123-136.
nexin2 protein in healthy and ischemized brain of adult rats. [68] Carimalo J, Cronier S, Petit G, Peyrin JM, Boukhtouche F,
Neuroscience 148, 653-667. Arbez N, Lemaigre-Dubreuil Y, Brugg B, Miquel MC (2005)
[66] Lee HP, Jun YC, Choi JK, Kim JI, Carp RI, Kim YS (2005) Activation of the JNK-c-Jun pathway during the early phase
Activation of mitogen-activated protein kinases in hamster of neuronal apoptosis induced by PrP106-126 and prion infec-
brains infected with 263K scrapie agent. J Neurochem 95, tion. Eur J Neurosci 21, 2311-2319.
584-593.
[67] Teunissen BE, Jansen AT, van Amersfoorth SC, O’Brien
TX, Jongsma HJ, Bierhuizen MF (2003) Analysis of the rat

You might also like