Modulating Protein Phosphatase 2A Rescues ... - ACS Publications

Jun 19, 2018 - ... and in the APP/PS1 model, the levels of Aβ remained unchanged. ... Access to Tau Disease-Associated Post-translational Modificatio...
0 downloads 0 Views 1MB Size
Subscriber access provided by NAGOYA UNIV

Article

Modulating Protein Phosphatase 2A rescues disease phenotype in neurodegenerative tauopathies Simon McKenzie-Nickson, Jacky Chan, Keyla Perez, Lin W. Hung, Lesley Cheng, Amelia Sedjahtera, Lydia Gunawan, Paul A Adlard, David J Hayne, Lachlan E Mcinnes, Paul S. Donnelly, David I. Finkelstein, Professor Andrew Hill, and Kevin J Barnham ACS Chem. Neurosci., Just Accepted Manuscript • DOI: 10.1021/acschemneuro.8b00161 • Publication Date (Web): 19 Jun 2018 Downloaded from http://pubs.acs.org on June 20, 2018

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 31 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

1

Modulating Protein Phosphatase 2A rescues disease phenotype in

2

neurodegenerative tauopathies

3

4

Simon McKenzie-Nickson 1,2a,, Jacky Chan 1, Keyla Perez 1, Lin W. Hung 1, Lesley Cheng 3,

5

Amelia Sedjahtera 1, Lydia Gunawan 1, Paul A. Adlard 1, David J. Hayne

6

McInnes 2b,

Paul S.

2b

, Lachlan E.

Donnelly 2b, David I. Finkelstein 1, Andrew F. Hill 3, Kevin J. Barnham 1,2a

7

8

1

9

Australia

Florey Institute of Neuroscience and Mental Health, Parkville, Melbourne, VIC 3052,

10

2 a

Department of Pharmacology and Therapeutics, b School of Chemistry; Bio21 Molecular

11

and Biotechnology Institute, Parkville, Melbourne, VIC 3052, Australia

12

3

13

Trobe University, Melbourne, VIC 3086, Australia

Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La

14 15

Correspondence

to

K.J.B

([email protected])

16

[email protected]).

17

Author contributions: SMN, KJB, LWH, AFH, DIF designed experiments. SMN, JC, KP, LWH, LC,

18

AS, LG, PAA, DJH, LEM, PSD conducted research. SMN, KP, LWH, LC, PAA analysed research.

19

SMN and KJB wrote the manuscript.

20

The authors declare no competing financial interests.

21

ACS Paragon Plus Environment

and

S.M.N

(simon.mckenzie-

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

22

Abstract

23

Alzheimer’s disease (AD) is the leading cause of dementia worldwide accounting around

24

70% of all cases. There is currently no treatment for AD beyond symptom management and

25

attempts at developing disease-modifying therapies have yielded very little. These strategies

26

have traditionally targeted the peptide Aβ which is thought to drive pathology. However, the

27

lack of clinical translation of these Aβ-centric strategies underscores the need for diverse

28

treatment strategies targeting other aspects of the disease. Metal dyshomeostasis is a common

29

feature of several neurodegenerative diseases such as AD, Parkinson’s disease, and

30

Frontotemporal dementia and manipulation of metal homeostasis has been explored as a

31

potential therapeutic avenue for these diseases. The copper ionophore glyoxalbis-[N4-

32

methylthiosemicarbazonato]Cu(II) (CuII(gtsm)) has previously been shown to improve the

33

cognitive deficits seen in an AD animal model, however the molecular mechanism remained

34

unclear. Here we report that the treatment of two animal tauopathy models (APP/PS1 and

35

rTg4510) with CuII(gtsm) recovers the cognitive deficits seen in both neurodegenerative

36

models. In both models, markers of tau pathology were significantly reduced with CuII(gtsm)

37

treatment and in the APP/PS1 model the levels of Aβ remained unchanged. Analysis of tau

38

kinases (GSK3β and CDK5) revealed no drug induced changes however both models

39

exhibited a significant increase in the levels of the structural subunit of the tau phosphatase,

40

PP2A. These findings suggest that targeting the tau phosphatase PP2A has therapeutic

41

potential for preventing memory impairments and reducing the tau pathology seen in AD and

42

other tauopathies.

43

Keywords: Alzheimer’s, tau, phosphatase, copper, neurodegeneration, metals

44

Funding: This work was funded by NHMRC grants APP1031193; APP1002373; 628946

45

Introduction

ACS Paragon Plus Environment

Page 2 of 31

Page 3 of 31 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

46

The growing epidemic of Alzheimer’s disease (AD) is one of the greatest health crises

47

currently facing the global community. As of 2015 1, AD is reported to cost the global

48

economy US$818 billion per year and with an ageing population, both the economic and

49

social cost of this disease are only set to rise. Early onset or familial AD is caused by a

50

mutation in one or more of the key pathological proteins such as amyloid precursor protein,

51

or presenilin 1. However, by far the majority of AD cases are considered to be sporadic or

52

late onset. These cases are not considered to be genetically determined 2; however, a growing

53

number of polymorphisms have been shown to increase risk of the disease 3. The

54

neuropathology of AD manifests as the presence of extracellular deposits, or plaques,

55

consisting primarily of the amyloid-β (Aβ) peptide. This peptide is proteolytically derived

56

from the amyloid precursor protein (APP). Another hallmark pathology of AD are the

57

intracellular ‘tangles’ of the tau protein 4.

58

These features form the basis of the amyloid cascade hypothesis which posits that an

59

accumulation of Aβ (either through overproduction or through lack of clearance from the

60

brain) drives the pathology as the disease progresses

61

development of multiple therapeutics targeting Aβ biology; however, such compounds have

62

had very little success to date 6. Therapies targeting tau have received comparatively little

63

attention, despite a growing body of research demonstrating the crucial role tau plays in the

64

development of disease and its potential as a therapeutic target 7–9.

65

Tau is a cytoskeletal protein with complex regulation and a wide variety of roles

66

function can be regulated through post-translational modifications such as acetylation 11, and,

67

of particular relevance to neurodegenerative diseases, phosphorylation 12. Tau can bind to and

68

stabilise microtubules, however, this process is regulated by phosphorylation at various

69

epitopes

70

brain tissue, tau is frequently seen with a level of tau phosphorylation higher than in non-

13

5555554

. This has resulted in the

10

. Tau

and as such the phosphorylation of tau can impact microtubule stability. .In AD

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

14

Page 4 of 31

diseased controls, both in animal models

72

enters a hyperphosphorylated state there is both a toxic gain-of-function (increased

73

propensity to aggregate

74

Alterations in the biology of the enzymes controlling tau phosphorylation is thought to

75

underlie the increase in tau phosphorylation observed in AD 19,20.

76

The phosphorylation state of tau is the result of a complex interplay between kinase and

77

phosphatase activity. The phosphorylation of tau is mediated primarily by the kinases

78

glycogen synthase kinase 3β (GSK3β) and cyclin dependent kinase 5 (CDK5)

79

ongoing interest in developing specific inhibitors for these enzymes however interest in this

80

area has been dampened with the failure of lithium in clinical trials

81

dephosphorylation occurs through the action of phosphatases, with protein phosphatase 2A

82

(PP2A) responsible for around 70% of tau dephosphorylation

83

consists of three subunits, a structural subunit (designated the A subunit), a regulatory ‘B’

84

subunit, and the ‘C’ subunit which is responsible for the enzymatic activity of PP2A. The B

85

subunit also provides substrate specificity 23. Impairments in PP2A are thought to contribute

86

to the hyperphosphorylation observed in AD

87

decreased in the brain tissue of AD patients

88

Additionally,

89

hyperphosphorylation and cognitive deficits in wildtype rats

90

other pacific islands there is a familial parkinsonism-dementia that has strong tau pathology

91

and decreased PP2A activity

92

and/or activity could be an attractive therapeutic option for AD and other tauopathies.

93

However to date, there has been limited activity in this field as PP2A’s complex biology and

94

regulation has made this problematic 28.

inhibition

and AD patients

15,16

71

. It is thought that when tau

17

) and a loss-of-function (loss of microtubule stability

of

PP2A

27

using

18

).

20

. There is

21

. Alternatively, tau

22

. The PP2A holoenzyme

19

24

. PP2A activity has been shown to be

alongside decreased PP2A expression okadaic

acid

treatment

induces

25

.

tau

26

. Furthermore, in Guam and

. These examples may suggest that increasing PP2A levels

ACS Paragon Plus Environment

Page 5 of 31 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

95

Copper(II) complexes of bis(thiosemicarbazone) ligands have shown great promise in

96

treating multiple animal models of neurodegenerative diseases

97

utilising the APP/PS1 mouse model of AD found that treatment with glyoxalbis-[N4-

98

methylthiosemicarbazonato]Cu(II) (CuII(gtsm)) improved the spatial memory deficits seen in

99

the model while a related compound, CuII(atsm), that does not release bioavailable copper did

100

not rescue the phenotype in this model suggesting the liberation of copper is important.

101

Further, biochemical analysis of treated animals revealed a reduction in the levels of

102

phosphorylated tau and a decrease in the levels of an Aβ oligomer

103

treatment with CuII(gtsm) not only improves the behavioural phenotype of APP/PS1 mouse

104

model of AD, but also the rTg4510 mice which overexpresses a mutant form of tau

105

associated with frontotemporal dementia. Furthermore, treatment reduced measures of tau

106

pathology in both models and produced an increase in the levels of the PP2A structural

107

subunit.

108

ACS Paragon Plus Environment

29

. A preliminary study

30

. Here, we show that

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

109

Results and discussion

110

CuII(gtsm) rescues the spatial memory deficit in APP/PS1 and rTg4510 mice

111

In agreement with previous studies, here we showed that the performance of both APP/PS1 31

112

(Figure 1) and rTg4510

113

maze and the Y-maze was impaired. In the probe trial for the Morris water maze (as

114

measured by the length of time spent in the platform quadrant during the probe trial) both the

115

APP/PS1 model (9 months of age (Figure 1, panel B, P = 0.0099) and the rTg4510 model

116

(12 months of age; Figure 2, Panel B, P = 0.017) were impaired as compared to wildtype

117

controls. We found that treatment with CuII(gtsm) significantly improved of the performance

118

of both APP/PS1 (P = 0.0021 compared to vehicle treated transgenic performance) mice and

119

rTg4510 mice (P = 0.018 compared to vehicle treated transgenic performance) such that they

120

performed similarly to wildtype animals when assessing time spent in the platform quadrant

121

(however, drug treatment didn’t influence the learning curve (Figure 1A and 2A

122

respectively). When tested in the Y-maze, vehicle treated APP/PS1 and rTg4510 both

123

performed significantly worse than wildtype mice (Figure 1, panel C, P = 0.0083 and Figure

124

2, panel C, P = 0.0002 respectively). Treatment of APP/PS1 mice with CuII(gtsm) yielded a

125

highly significant improvement in Y-maze performance (P = 0.0005 compared to vehicle

126

treated transgenic performance) while treatment of rTg4510 mice did not significantly alter

127

the performance (P = 0.14). Further analysis utilising a 1-sample T-test against chance

128

reveals that in both APP/PS1 and rTg4510 transgenic mice performance did not differ from

129

chance (P = 0.13 and P = 0.97 respectively). CuII(gtsm) treated mice in both APP/PS1 and

130

rTg4510 models did in fact perform better than chance (P = 0.0062 and P = 0.02 respectively)

131

revealing a preference for the novel arm over and above the chance value of 33.3%.

32

(Figure 2) mouse models spatial memory in the Morris water

132

ACS Paragon Plus Environment

Page 6 of 31

Page 7 of 31 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

133

CuII(gtsm) treatment rescues the severe hyperactivity phenotype of rTg4510 mice

134

In an open field test, rTg4510 mice showed a marked hyperactivity phenotype as measured

135

by total distance moved (P < 0.0001 compared to wildtype; Figure 3, panel A) and by the

136

low total amount of time spent resting (Figure 3, panel B, P < 0.0001 compared to wildtype).

137

Treatment of these mice with CuII(gtsm) significantly reduced the total distance moved (P =

138

0.0003 compared to vehicle treated transgenic mice) and improved the time spent resting (P