Skip to main content
Erschienen in: Molecular Neurodegeneration 1/2015

Open Access 01.12.2015 | Research article

Region-specific dendritic simplification induced by Aβ, mediated by tau via dysregulation of microtubule dynamics: a mechanistic distinct event from other neurodegenerative processes

verfasst von: Nataliya Golovyashkina, Lorène Penazzi, Carlo Ballatore, Amos B. Smith III, Lidia Bakota, Roland Brandt

Erschienen in: Molecular Neurodegeneration | Ausgabe 1/2015

Abstract

Background

Dendritic simplification, a key feature of the neurodegenerative triad of Alzheimer’s disease (AD) in addition to spine changes and neuron loss, occurs in a region-specific manner. However, it is unknown how changes in dendritic complexity are mediated and how they relate to spine changes and neuron loss.

Results

To investigate the mechanisms of dendritic simplification in an authentic CNS environment we employed an ex vivo model, based on targeted expression of enhanced green fluorescent protein (EGFP)-tagged constructs in organotypic hippocampal slices of mice. Algorithm-based 3D reconstruction of whole neuron morphology in different hippocampal regions was performed on slices from APPSDL-transgenic and control animals. We demonstrate that induction of dendritic simplification requires the combined action of amyloid beta (Aβ) and human tau. Simplification is restricted to principal neurons of the CA1 region, recapitulating the region specificity in AD patients, and occurs at sites of Schaffer collateral input. We report that γ-secretase inhibition and treatment with the NMDA-receptor antagonist, CPP, counteract dendritic simplification. The microtubule-stabilizing drug epothilone D (EpoD) induces simplification in control cultures per se. Similar morphological changes were induced by a phosphoblocking tau construct, which also increases microtubule stability. In fact, low nanomolar concentrations of naturally secreted Aβ decreased phosphorylation at S262 in a cellular model, a site which is known to directly modulate tau-microtubule interactions.

Conclusions

The data provide evidence that dendritic simplification is mechanistically distinct from other neurodegenerative events and involves microtubule stabilization by dendritic tau, which becomes dephosphorylated at certain sites. They imply that treatments leading to an overall decrease of tau phosphorylation might have a negative impact on neuronal connectivity.
Hinweise

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

NG carried out slice culture experiments, performed image analysis, performed statistical analysis and helped to draft the manuscript; LP participated in organotypic culture preparations and performed statistical analysis; CB synthesized epothilone D, participated in the design of the study and helped to draft the manuscript; AS participated in the design of the study and helped to draft the manuscript; LB prepared the Sindbis virus, performed image analysis, participated in the design of the study and drafted the manuscript, RB designed and coordinated the study and drafted the manuscript. All authors read and approved the final manuscript.
Abkürzungen
Amyloid beta
ΑΝΟVΑ
Analysis of variance
ΑPP
Amyloid precursor protein
AD
Alzheimer’s disease
DIV
Days in vitro
EGFP
Enhanced green fluorescent protein
EpoD
Epothilone D
LSD
Least significant difference
NMDAR
NMDA receptor

Background

Alzheimer’s disease (AD) is characterized by the neurodegenerative triad of spine changes, dendritic simplification and neuron loss [1]. The pathologic changes are accompanied by the formation of extracellular plaques, composed of Aβ peptides, and intracellular neurofibrillary tangles containing tau in a hyperphosphorylated state [2, 3]. While the development of spine changes and neuron loss has been addressed in various animal and culture models [47], much less is known about how changes in dendritic complexity are mediated. Spires et al. [7] reported dendritic changes in plaque developing mice and Wu et al. [8] provided evidence that activation of the calcium-dependent phosphatase calcineurin is involved in Aβ-induced dendritic simplification, however it remains unclear how these changes are mediated. The lack of knowledge is noteworthy since dendritic simplification, which has long been recognized in AD, is likely to contribute to brain malfunction to a major extent and is considered to be a forerunner to neuronal loss [9].
A number of careful morphological studies of human brain material have established that alterations in dendritic arbor are regionally specific in AD [10]. In the hippocampus, dendritic extent is largely reduced in CA1 pyramidal neurons [11], while no changes were observed in CA3 neurons [12, 13]. The differences appear not to be due to postsynaptic dendritic regression since no excess loss of CA3 pyramidal neurons was observed [14]. Loss of spines is more widespread [15], suggesting that dendritic simplification develops independent from spine changes and neuronal loss. How changes in neuronal morphology develop during disease is however largely unknown.
Neuronal morphology is highly dependent on the organization of the cytoskeleton with microtubules playing an important role in regulating dendritic arborization [16]. The microtubule-associated tau proteins, known to modulate microtubule dynamics in neurons [17], are present and functionally active to a major extent in dendrites [18, 19] and may also be involved in dendritic remodeling during health and disease [20]. However, whether Aβ and tau are involved in dendritic simplification and whether they functionally interact to execute these changes is unknown.
To scrutinize the mechanisms of dendritic simplification in an authentic CNS environment, we employed a previously established ex vivo model based on targeted expression of EGFP-tagged constructs in organotypic hippocampal slices [5]. We demonstrate that dendritic simplification is restricted to principal neurons of the CA1 region, and is induced by Aβ, mediated by tau through NMDA receptor activation. We provide evidence that dendritic simplification involves dysregulation of microtubule dynamics by dendritic tau, which becomes dephosphorylated at certain sites, and is mechanistically distinct from spine changes and neuron loss.

Results

Induction of dendritic simplification requires the action of human tau on an APPSDL transgenic background

We employed an ex vivo model of AD using organotypic hippocampal slice cultures from APPSDL transgenic mice in combination with Sindbis virus-mediated expression of EGFP-tagged constructs. We demonstrated previously that this approach permits analysis of cell death and synaptic changes with respect to the functional interaction between Aβ and tau pathology [5]; here we have extended the approach to determine region-specific changes in dendritic morphology. Virus infections were performed at 12 days in vitro (DIV) and slices were analyzed 3 days later (Fig. 1a). At these conditions efficient neuronal expression of EGFP with low toxicity was obtained [21]. We have also shown previously that the expression levels of different EGFP-tau constructs are similar [5, 21]. Virus titer was adjusted to achieve low infection rate in CA1, CA3 and DG, which permitted 3D imaging of single neurons by high-resolution cLSM with minimal overlap of dendritic arbors (Fig. 1b). Image tile stacks were stitched into single neurons and the morphology of whole neurons was reconstructed in 3D by automated digitalization (Fig. 1c) [22]. The detection gain was adjusted to capture the full arbor, including smaller secondary and tertiary dendrites, irrespective of the EGFP-construct used. Comparing neuronal morphology after virus infection in slices prepared from APPSDL transgenic mice versus B6 controls permitted determination of the effect of the APP transgene on dendritic morphology in the apical (red) and basal (blue) arbor in principal neurons from the different hippocampal regions (Fig. 1d).
For a quantitative assessment of potential changes in dendritic arborization, we determined the total path length and the number of branching points in principal neurons from CA1, CA3 and DG. Expression of EGFP alone on APPSDL transgenic background induced only slight dendritic simplification, which did not reach significance. In contrast, we observed that EGFP-htau expression on APPSDL transgenic background induced pronounced and significant dendritic simplification in CA1 neurons as indicated by a reduction of total path length and number of branching points by 38 % and 30 %, respectively (Fig. 2a, left). No change was seen in CA3 pyramidal neurons and granule cells of the DG (Fig. 2a, middle and right). Human tau expression on non-transgenic background did not induce morphological changes, indicating that dendritic simplification in the CA1 region requires the combination of human tau and APPSDL transgenic background.

Dendritic simplification occurs at sites of Schaffer collateral input in CA1 neurons

CA1 pyramidal neurons receive different inputs in specific subregions of the dendritic tree (Fig. 2b). To determine subregional differences in the induction of dendritic simplification, 3D Sholl analysis [23] was performed on basal and apical branches to quantitate changes in the intersection frequency of dendrites as a function of the distance from the cell body (Fig. 2c, left). On the APP transgenic background we observed a reduction of dendritic complexity in the entire basal part of the tree (Fig. 2c, right). In contrast, in the apical part, dendritic simplification was induced specifically in two segments of the tree. Responsive segments were localized at ~15-25 % and ~60-70 % of total apical length and correlated with regions, where most input from Schaffer collaterals occurs (see Fig. 2b).

γ-Secretase inhibition and treatment with NMDA receptor inhibitors counteract dendritic simplification

Dendritic simplification could be induced by the presence of mutated APP or might require the generation of Aβ through γ- and β-secretase mediated cleavage of APPSDL. To distinguish between these possibilities, we pretreated the culture with the non-transition state γ-secretase inhibitor DAPT in order to reduce Aβ generation [24]. Treatment with DAPT was started one day before infection with EGFP-htau expressing virus (Fig. 3a, top). DAPT reduced the negative effect of tau and APP on dendritic complexity in CA1 neurons as judged by a significant increase in total path length compared to untreated cultures of the same genotype (Fig. 3a, bottom). Sholl analysis of CA1 neurons revealed that dendritic simplification was completely abolished in basal dendrites and reduced in apical segments (Fig. 3b). The data indicate that dendritic simplification is induced by Aβ rather than mutated APP.
It has previously been shown that Aβ acts via NMDA receptor (NMDAR) activation to induce spine changes and neuron loss [5, 25, 26]. To test whether NMDAR activation is also involved in mediating dendritic simplification, we added the competitive NMDAR antagonist CPP [27] to the cultures one day before infection with EGFP-htau expressing virus (Fig. 4a, top). CPP also abolished the negative effect of tau and Aβ on dendritic complexity in CA1 neurons similar to DAPT (Fig. 4a, bottom, and b), demonstrating that dendritic simplification is mediated by NMDAR activation. It should also be noted that both drugs induced some dendritic simplification in CA3 neurons, which might indicate sensitivity of the CA3 neurons to the drugs themselves.
Table 1
Dendritic simplification in an ex vivo model of Alzheimer’s disease. Summary representation of the effect of the expressed construct, the genotype of the mouse and pharmacological treatments on total path length (A) and number of branching points (B). Statistical evaluation was performed using two-tailed, unpaired Student’s t test for comparison of the two genotypes under control conditions (untreated), and one way ANOVA with post hoc Fisher’s LSD test for multiple comparisons of drug treatments or different tau constructs. P values, which represent statistical significance (p≤0.05) are indicated in bold, except when the alpha level of ANOVA was above 0.05
A. Total path length
Construct
Condition
Region
Genotype
Mice (n)
Cells (n)
Mean ± s.e.m (based on cell number) in mm
Student’s test
  
p values (B6 vs. APP)
EGFP
untreated
CA1
B6
5
9
3.113±0.465
p = 0.775
EGFP
untreated
CA1
APPSDL
5
5
3.364±0.707
EGFP
untreated
CA3
B6
7
8
4.490±0.292
p = 0.401
EGFP
untreated
CA3
APPSDL
5
7
4.187±0.202
EGFP
untreated
DG
B6
6
6
1.223±0.232
p = 0.275
EGFP
untreated
DG
APPSDL
5
7
0.911±0.161
EGFP-htau
untreated
CA1
B6
4
7
3.996±0.237
p = 0.0003
EGFP-htau
untreated
CA1
APPSDL
6
8
2.491±0.163
EGFP-htau
untreated
CA3
B6
5
6
4.344±0.423
p = 0.428
EGFP-htau
untreated
CA3
APPSDL
5
6
3.920±0.283
EGFP-htau
untreated
DG
B6
5
10
0.813±0.071
p = 0.888
EGFP-htau
untreated
DG
APPSDL
7
17
0.826±0.051
Construct
Condition
Region
Genotype
Mice (n)
Cells (n)
Mean ± s.e.m (based on cell number) in mm
One way ANOVA (together with EGFP-htau untreated, both genotypes, same region)
p values (B6 vs. APP)
p values ( vs. untreated EGFP-htau same region and same genotype)
EGFP-htau
DAPT
CA1
B6
4
6
3.691±0.154
F (3,24) = 14.05 ; p < 0.0001
p = 0.0440
p = 0.2657
EGFP-htau
DAPT
CA1
APPSDL
4
7
3.122±0.154
p = 0.0182
EGFP-htau
DAPT
CA3
B6
5
6
3.122±0.258
F (3,22) = 3.634 ; p = 0.0287
p = 0.8981
p = 0.0133
EGFP-htau
DAPT
CA3
APPSDL
4
8
3.177±0.249
p = 0.0940
EGFP-htau
CPP
CA1
B6
5
7
3.671±0.289
F (3,25) = 7.781 ; p = 0.0008
p = 0.2469
p = 0.3517
EGFP-htau
CPP
CA1
APPSDL
6
7
3.265±0.263
p = 0.0279
EGFP-htau
CPP
CA3
B6
6
6
3.714±0.293
F (3,23) = 0.5765 ; p = 0.6363
p = 0.8658
p = 0.2483
EGFP-htau
CPP
CA3
APPSDL
7
9
3.797±0.357
p = 0.8023
EGFP-htau
EpoD
CA1
B6
7
8
3.038±0.288
F (3,27) = 7.040 ; p = 0.0012
p = 0.5142
p = 0.0092
EGFP-htau
EpoD
CA1
APPSDL
8
8
2.820±0.241
p = 0.3273
EGFP-htau
EpoD
CA3
B6
7
8
3.603±0.186
F (3,23) = 5.757 ; p = 0.0043
p = 0.0416
p = 0.0527
EGFP-htau
EpoD
CA3
APPSDL
7
7
2.853±0.126
p = 0.0089
EGFP-Ala htau
untreated
CA1
B6
5
7
2.797±0.268
F (3,24) = 8.839 ; p = 0.0004
p = 0.1005
p = 0.0011
EGFP-Ala htau
untreated
CA1
APPSDL
4
6
3.370±0.249
p = 0.0125
EGFP-Ala htau
untreated
CA3
B6
5
6
2.912±0.224
F (3,20) = 4.276 ; p = 0.0174
p = 0.8829
p = 0.0076
EGFP-Ala htau
untreated
CA3
APPSDL
4
6
2.984±0.396
p = 0.0667
EGFP-PHP htau
untreated
CA1
B6
4
7
3.342±0.298
F (3,24) = 5.250 ; p = 0.0063
p = 0.6532
p = 0.1107
EGFP-PHP htau
untreated
CA1
APPSDL
4
6
3.155±0.429
p = 0.1091
EGFP-PHP htau
untreated
CA3
B6
5
6
3.578±0.450
F (3,20) = 2.724 ; p = 0.0714
p = 0.1941
p = 0.1722
EGFP-PHP htau
untreated
CA3
APPSDL
4
6
2.851±0.352
p = 0.0621
Construct
Condition
Region
Genotype
Mice (n)
Cells (n)
Mean ± s.e.m (based on cell number) in mm
One way ANOVA (together with EGFP untreated, both genotypes, same region)
p values (B6 vs. APP)
p values ( vs. untreated EGFP same region and genotype)
EGFP
CPP
CA1
B6
6
7
3.597±0.207
F (3,23) = 0.4707 ; p = 0.7056
p = 0.7702
p = 0.4144
EGFP
CPP
CA1
APPSDL
6
6
3.787±0.334
p = 0.5514
EGFP
CPP
CA3
B6
5
5
3.603±0.244
F (3,22) = 2.806 ; p = 0.0634
p = 0.7762
p = 0.0473
EGFP
CPP
CA3
APPSDL
5
6
3.474±0.382
p = 0.0974
EGFP
EpoD
CA1
B6
6
6
3.110±0.307
F (3,23) = 0.2951 ; p = 0.8285
p = 0.4532
p = 0.9960
EGFP
EpoD
CA1
APPSDL
7
7
3.591±0.168
p = 0.7353
EGFP
EpoD
CA3
B6
6
6
2.995±0.268
F (3,23) = 5.693 ; p = 0.0046
p = 0.0438
p = 0.0006
EGFP
EpoD
CA3
APPSDL
6
6
3.851±0.287
p = 0.3939
B. Number of branching points
Construct
Condition
Region
Genotype
Mice (n)
Cells (n)
Mean ± s.e.m (based on cell number)
Student’s t test
  
p values (B6 vs. APP)
EGFP
untreated
CA1
B6
5
9
31±4.11
p = 0.571
EGFP
untreated
CA1
APPSDL
5
5
36±7.22
EGFP
untreated
CA3
B6
7
8
39±3.83
p = 0.203
EGFP
untreated
CA3
APPSDL
5
7
33±1.90
EGFP
untreated
DG
B6
6
6
11±2.68
p = 0.261
EGFP
untreated
DG
APPSDL
5
7
7±0.57
EGFP-htau
untreated
CA1
B6
4
7
36±3.58
p = 0.016
EGFP-htau
untreated
CA1
APPSDL
6
8
23±3.05
EGFP-htau
untreated
CA3
B6
5
6
40±3.24
p = 0.185
EGFP-htau
untreated
CA3
APPSDL
5
6
34±2.24
EGFP-htau
untreated
DG
B6
5
10
8±4.49
p = 0.933
EGFP-htau
untreated
DG
APPSDL
7
17
8±2.73
Construct
Condition
Region
Genotype
Mice (n)
Cells (n)
Mean ± s.e.m (based on cell number)
One way ANOVA (together with EGFP-htau untreated, both genotypes, same region)
p values (B6 vs. APP)
p values ( vs. untreated EGFP-htau same region and genotype)
EGFP-htau
DAPT
CA1
B6
4
6
36±2.11
F (3,24) = 4.920 ; p = 0.0084
p = 0.2443
P > 0.9999
EGFP-htau
DAPT
CA1
APPSDL
4
7
31±2.00
p = 0.0511
EGFP-htau
DAPT
CA3
B6
5
6
30±1.95
F (3,22) = 8.519 ; p = 0.0006
p = 0.1167
p = 0.0058
EGFP-htau
DAPT
CA3
APPSDL
4
8
25±1.51
p = 0.0076
EGFP-htau
CPP
CA1
B6
5
7
32±4.34
F (3,25) = 2.434 ; p = 0.0885
p = 0.4497
p = 0.4497
EGFP-htau
CPP
CA1
APPSDL
6
7
28±3.53
p = 0.3310
EGFP-htau
CPP
CA3
B6
6
6
32±2.77
F (3,23) = 5.381 ; p = 0.0059
p = 0.1041
p = 0.0509
EGFP-htau
CPP
CA3
APPSDL
7
9
26±2.19
p = 0.0339
EGFP-htau
EpoD
CA1
B6
7
8
27±1.83
F (3,27) = 4.824 ; p = 0.0081
p = 0.5753
p = 0.0203
EGFP-htau
EpoD
CA1
APPSDL
8
8
25±1.31
p = 0.5753
EGFP-htau
EpoD
CA3
B6
7
8
29±2.86
F (3,23) = 4.362 ; p = 0.0143
p = 0.4458
p = 0.0120
EGFP-htau
EpoD
CA3
APPSDL
7
7
26±2.93
p = 0.0668
EGFP-Ala htau
untreated
CA1
B6
5
7
26±3.41
F (3,24) = 3.655 ; p = 0.0266
p = 0.1135
p = 0.0430
EGFP-Ala htau
untreated
CA1
APPSDL
4
6
34±3.12
p = 0.0288
EGFP-Ala htau
untreated
CA3
B6
5
6
25±3.06
F (3,20) = 4.572 ; p = 0.0135
p = 0.5032
p = 0.0028
EGFP-Ala htau
untreated
CA3
APPSDL
4
6
28±3.72
p = 0.1878
EGFP-PHP htau
untreated
CA1
B6
4
7
32±3.28
F (3,24) = 3.736 ; p = 0.0246
p = 0.1469
p = 0.3815
EGFP-PHP htau
untreated
CA1
APPSDL
4
6
25±2.45
p = 0.6630
EGFP-PHP htau
untreated
CA3
B6
5
6
38±6.57
F (3,20) = 0.6744 ; p = 0.5778
p = 0.3253
p = 0.7762
EGFP-PHP htau
untreated
CA3
APPSDL
4
6
31±6.14
p = 0.6702
Construct
Condition
Region
Genotype
Mice (n)
Cells (n)
Mean ± s.e.m (based on cell number)
One way ANOVA (together with EGFP untreated, both genotypes, same region)
p values (B6 vs. APP)
p values ( vs. untreated EGFP same region and genotype)
EGFP
CPP
CA1
B6
6
7
34±3.15
F (3,23) = 0.3279 ; p = 0.8052
p = 0.7518
p = 0.6011
EGFP
CPP
CA1
APPSDL
6
6
36±2.74
p > 0.9999
EGFP
CPP
CA3
B6
5
5
33±3.24
F (3,22) = 2.872 ; p = 0.0594
p = 0.1730
p = 0.2132
EGFP
CPP
CA3
APPSDL
5
6
26±3.16
p = 0.1396
EGFP
EpoD
CA1
B6
6
6
25±2.16
F (3,23) = 1.069 ; p = 0.3814
p = 0.3033
p = 0.2778
EGFP
EpoD
CA1
APPSDL
7
7
31±0.57
p = 0.4130
EGFP
EpoD
CA3
B6
6
6
23±3.24
F (3,23) = 4.970 ; p = 0.0084
p = 0.0141
p = 0.0010
EGFP
EpoD
CA3
APPSDL
6
6
35±1.99
p = 0.6501

The microtubule-stabilizing drug EpoD induces dendritic simplification in control cultures

Data from this study could suggest that Aβ induces microtubule destabilization in a tau-dependent manner, which results in a decrease of dendritic complexity. If true, one would expect that microtubule-stabilizing drugs would prevent dendritic simplification. To test this hypothesis we treated the cultures prior to infection with the microtubule-stabilizing drug epothilone D (EpoD), a brain-penetrant small molecule potential therapeutic candidate for AD [28, 29]. Epothilones may have neuroprotective activity; however dose-dependent neurotoxic effects have also been reported [30]. In pilot experiments we observed loss of neurons in control cultures at concentrations of 1 and 5 nM of EpoD indicating some toxicity for the cells. The number of neurons was not affected at 0.2 nM EpoD, a concentration where still induction of microtubule polymerization was observed in model neurons (data not shown). Thus this concentration was subsequently used for the experiments. Surprisingly, EpoD induced dendritic simplification in control cultures as indicated by significantly decreased total path length and branching compared to the respective untreated cultures (Fig. 5a, Table 2). No obvious differences in the dendritic morphology between htau-expressing APP-transgenic and control cultures were observed, which was – except some simplification in basal dendrites – confirmed by Sholl analysis (Fig. 5b, c).
Table 2
Effect of drug treatment on dendritic tau levels. Dendritic tau levels in 2nd or 3rd order of CA1 neurons expressing the respective construct were determined as described in “Methods”. For every neuron, 3–4 measurements were performed and averaged. Statistical evaluation was performed using two-tailed, unpaired Student’s t test for untreated cultures, and one way ANOVA with post hoc Fisher’s LSD test for multiple comparisons of drug treatments or different tau constructs. Note that alpha levels of ANOVA did not reach significance (p≤0.05) for any condition
Construct
Condition
Region
Genotype
Mice (n)
Cells (n)
Mean ± s.e.m (based on cell number) of normalized intensity values
Student’s t test
  
p values (B6 vs. APP)
EGFP-htau
untreated
CA1
B6
3
5
1.061±0.099
p = 0.6210
EGFP-htau
untreated
CA1
APPSDL
5
5
1.111±0.120
Construct
Condition
Region
Genotype
Mice (n)
Cells (n)
Mean ± s.e.m (based on cell number) of normalized intensity values
One way ANOVA (together with EGFP-htau untreated, both genotypes)
p values (B6 vs. APP)
p values (vs. untreated same genotype)
EGFP-htau
DAPT
CA1
B6
3
6
0.827±0.066
F (3,17) = 2.183 ; p = 0.1275
p = 0.0273
p = 0.2007
EGFP-htau
DAPT
CA1
APPSDL
4
5
1.197±0.154
p = 0.5981
EGFP-htau
EpoD
CA1
B6
4
5
1.119±0.138
F (3,16) = 0.2108 ; p = 0.8874
p = 0.8533
p = 0.5763
EGFP-htau
EpoD
CA1
APPSDL
5
5
1.148±0.066
p = 0.8135
EGFP-htau
CPP
CA1
B6
5
5
0.945±0.176
F (3,16) = 1.334 ; p = 0.2984
p = 0.3752
p = 0.6129
EGFP-htau
CPP
CA1
APPSDL
4
5
0.793±0.019
p = 0.0745
EGFP-Ala htau
untreated
CA1
B6
5
5
1.083±0.135
F (3,16) = 0.6240 ; p = 0.6098
p = 0.2952
p = 0.7390
EGFP-Ala htau
untreated
CA1
APPSDL
5
5
0.917±0.068
p = 0.2240
EGFP-PHP htau
untreated
CA1
B6
3
5
1.174±0.064
F (3,16) = 0.5229 ; p = 0.6727
p = 0.9881
p = 0.2937
EGFP-PHP htau
untreated
CA1
APPSDL
3
5
1.172±0.080
p = 0.6495

Expression of phosphoblocking tau induces dendritic simplification per se and increases microtubule stability

Since microtubule stabilization did not have any effect in Aβ-producing cultures, we explored whether tau phosphorylation, which is known to affect microtubule dynamics, is at all involved in mediating dendritic simplification. We employed a phosphoblocking (Ala htau) and a phosphomimicking (PHP htau) construct in which 10 AD-relevant sites were modified to alanine to prevent, or to glutamate to simulate, phosphorylation at these residues (Fig. 6a, top) [31]. Surprisingly, expression of Ala htau induced dendritic simplification per se whereas the phosphomimicking PHP htau did not induce morphological changes (Fig. 6a, bottom, Table 1). This is in sharp contrast to the previous observation that PHP tau is the active species to induce cell death, and importantly indicates that the development of dendritic simplification and tau-dependent cell death are mechanistically distinct. The finding that the effect of Ala htau expression closely resembles the impact of EpoD-treatment could suggest that Ala htau induces dendritic simplification by hyperstabilizing dendritic microtubules. To test this hypothesis, we determined the influence of wt htau, Ala htau and PHP htau expression on microtubule stability by determining the ratio of acetylated to total tubulin, since tubulin acetylation is considered to be a marker for microtubule stability [32]. Expression of Ala htau induced a significantly increased ratio of acetylated to total tubulin compared to wt htau or PHP htau expressing cultures (Fig. 6b). This indicates that non-phosphorylatable htau induces dendritic simplification due to the inherent activity to promote microtubule stabilization. It should however be noted that Ala htau expression prevented dendritic simplification in CA1 neurons from APPSDL transgenic animals (Fig. 6a, bottom), which might indicate that it counteracts Aβ-induced microtubule destabilization in these neurons.
Generally, higher Aβ concentrations are considered to increase rather than decrease tau phosphorylation, but the effect of low concentrations of secreted Aβ on the phosphorylation of human tau is less clear. A previous study using a coculture system has shown that secreted Aβ induces an increased phosphorylation at the AT8 (S202/T205) and the AT180 epitope (T231/S235) [33]. The concentration of Aβ was comparable with that found in cerebrospinal fluid of AD patients, probably in the higher nanomolar range. However, a rough estimate suggests that the amount of Aβ in the brain of APPSDL transgenic mice that have been used for preparation of the organotypic slices is in the low nanomolar range (L. Bakota, unpublished observation), which reflects more a presymptomatic scenario. To test the hypothesis that low nanomolar concentrations of naturally secreted Aβ cause a change in the phosphorylation pattern of tau that might lead to increased microtubule stabilization, we turned to a cellular model, where PAGFP-tagged human tau is stably expressed in rat PC12 cells (Fig. 6c). The cells were seeded in a defined density, neuronally differentiated with NGF and incubated with a supernatant of APPSwe-transfected HEK cells. Western blot analysis showed that 3.5 nM Aβ caused a decreased phosphorylation at S262, while some other sites such as T181 and S214 were not affected. Interestingly, S262 is located in the microtubule-binding region of tau and phosphorylation of S262 largely abolishes its interaction with microtubules [34]. Thus, even a small decrease in S262 phosphorylation could induce tau-dependent microtubule stabilization thereby promoting dendritic simplification.

Discussion

The major findings of our current study comprise: (1) dendritic simplification is induced by Aβ and mediated by tau through NMDAR activation in a spatially highly restricted manner; (2) dendritic simplification is mechanistically distinct from other neurodegenerative events; and (3) dendritic simplification involves microtubule stabilization by dendritic tau, which becomes dephosphorylated at certain sites.
In contrast to spine changes and neuron loss, the mechanisms that contribute to dendritic simplification are much less studied. The pattern of dendritic arborization crucially determines the final integration of inputs and information processing. Thus, it is clear that a reduction of total dendritic path length by about 40 % as we observed it in principal neurons of the CA1 region (see Fig. 2a), has a major impact on synaptic connectivity even if the density of spines would remain unchanged.
It is well established that neuronal activity and activation of NMDARs play a key role in the regulation of dendritic arbor elaboration and dynamics as NMDAR-dependent neurotransmission induces an increased complexity of dendritic branches [35] and hypofunctional NMDARs lead to dendritic simplification [36]. This is in agreement with our observation that Aβ induces dendritic simplification through NMDARs and that morphological changes occur at sites of Schaffer collateral input, i.e. at glutamatergic synapses. Downstream signaling pathways may involve modulation of dendritic Ca2+-dependent plasticity and may finally be mediated through regulation of cytoskeletal dynamics. Previous studies have shown that members of the Rho family of small GTPases such as RhoA and Cdc42 influence dendritic development, possibly by functionally linking surface receptor activation to the organization of the actin cytoskeleton [37]. Very recently it also has been shown that a RhoA signaling pathway regulates the formation of Golgi outposts that contribute to dendritic growth and branch dynamics [38]. Interestingly, the activity level of RhoA appears also to modify the phosphorylation state of tau [39]. On the other side, tau belongs to the class of intrinsically disordered proteins, which are known to serve as hubs in cellular protein-protein interaction networks and are involved in signaling [40]. Thus, it is conceivable that tau phosphorylation influences various signal transduction mechanisms that are involved in the complex process of dendritic arborization.
We have provided evidence that Aβ induces changes in htau by acting on NMDARs, which then lead to local modulation of microtubule dynamics (Fig. 7). This provokes the question, how the mechanisms of dendritic simplification relate to the other facets of the neurodegenerative triad that occur during AD, i.e., to spine changes and neuron loss. We and others have previously reported that soluble Aβ causes loss of spines in an NMDAR-dependent manner, but independent of tau [5, 26]. This contrasts with the requirement of tau for the induction of dendritic simplification and indicates that different mechanisms operate to mediate spine alterations on the one hand, and changes of dendritic arborization on the other. Neuron loss requires a combined action of Aβ and tau [5, 41, 42], indicating that dendritic simplification and loss of neurons share the engagement of tau. However, tau-dependent cell loss occurred in the CA3 region, but not in CA1 neurons [21], whereas dendritic simplification was mostly evident in CA1 and not in CA3. The inverse regional relationship argues against the hypothesis that dendritic simplification is a forerunner to neuronal loss [9]. Furthermore, neuron loss was blocked in the presence of phosphoblocking tau [5], whereas the same construct induced dendritic simplification per se as shown in the present study. Thus the data indicate that dendritic simplification is mechanistically distinct from the other two features of the neurodegenerative triad.
Importantly, we demonstrated that dendritic simplification was induced in a highly localized manner in the hippocampus. In AD patients, major dendritic regression of CA1 neurons was found [11], whereas no reduction of dendritic extent in CA3 pyramidal neurons occurred [12, 13]. Remarkably, the region-specificity was recapitulated in our model suggesting that this model provides a useful system to analyze the mechanism of region-specific dendritic simplification. Since soluble Aβ is globally present, dendritic tau appears to play a decisive role in mediating region specific effects, which might differ between cells and subcellular microdomains. It has previously been shown that tau is present to a major extent in dendrites where it generally has a higher phosphorylation state than in the axon [19, 43]. In this study we have demonstrated that a phosphoblocking tau construct leads to increased microtubule stabilization that causes dendritic simplification suggesting that conditions, which decrease the phosphorylation state of dendritic tau at certain sites can be harmful. None of the tau constructs or treatment conditions changed the dendritic tau level suggesting that a redistribution of tau does not play a role in mediating dendritic simplification (Table 2). Using a cellular model system we showed in this study that low nanomolar concentrations of secreted Aβ cause a decreased phosphorylation at S262, a site that, when phosphorylated, abolishes tau’s interaction with microtubules [34].
Interestingly, phosphorylated S262 (but not T181 or S214) is a preferential target for the major brain phosphatase calcineurin (PP2B) [44, 45], which has previously been shown to be activated by Aβ in a NMDAR-dependent pathway [5, 26] and was reported to be involved in dendritic simplification [8]. Our data could suggest that phosphorylation at S262 might be a target for a selective intervention in the mechanisms that lead to dendritic simplification. Furthermore we have shown that NMDAR activation is involved in inducing dendritic simplification and that dendritic simplification takes place at sites of Schaffer collateral inputs where postsynaptic LTP occurs [46].

Conclusions

Using an ex vivo model we showed that (1) dendritic simplification is induced by Aβ and mediated by tau through NMDAR activation in a spatially highly restricted manner; (2) dendritic simplification is mechanistically distinct from other neurodegenerative events; and (3) dendritic simplification involves microtubule stabilization by dendritic tau, which is dephosphorylated at specific sites. The proposed pathway that mediates dendritic simplification is shown in Fig. 7.
The finding that the mechanisms inducing and mediating dendritic simplification are distinct from other AD related abnormalities could be relevant for the development of treatments to slow down the neurodegenerative triad and should be taken into account when testing new approaches. In particular, treatments leading to an overall decrease of tau phosphorylation [47] might have a negative impact on neuronal connectivity since they could induce dendritic simplification.

Methods

Animals

Heterozygous APPSDL transgenic mice [48] expressing human APP695 with three familial AD mutations were used. APPSDL mice express equimolar concentrations of Aβ40 and Aβ42 already at embryonal age [49]. Genotyping was performed as described previously [5]. APPSDL transgenic and C57BL/6 J (B6) control animals were maintained and sacrificed according to National Institutes of Health Guidelines and German Animal Care Regulations.

Materials

Chemicals were purchased from Sigma (Steinheim, Germany), culture medium and supplements from Sigma and Invitrogen (Darmstadt, Germany), culture dishes and plates from Nunc (Langenselbold, Germany), and membrane culture inserts from Millipore (Billerica, MA, USA). γ-secretase inhibitor N-[N-(3,5-difluorophenacetyl)-1-alanyl]-S-phenylglycine t-butyl ester (DAPT) was purchased from Merck (Darmstadt, Germany). Epothilone D (EpoD) was prepared as previously described [50, 51]. The spectroscopic properties of the compound were identical to those reported in the literature. Compound purity was >95 % as determined by LC-MS and NMR analyses.

Sindbis virus constructs

Construction of virus was performed as described previously [21]. The following constructs were used for the experiments: pSinRep5-EGFP, pSinRep5-EGFP-352wt htau, pSinRep5-EGFP-352 Ala htau, pSinRep5-EGFP-352 PHP htau. For Ala and PHP htau, respectively, 10 sites (Ser198, Ser199, Ser202, Thr231, Ser235, Ser396, Ser404, Ser409, Ser413, and Ser422) were mutated to alanine to block or to glutamate to mimic phosphorylation at the respective residues [31].

Organotypic hippocampal slice cultures and Sindbis virus infection

Hippocampal slice cultures were prepared from mice of either sex and processed as described previously [5, 52]. On day 12 in vitro slice cultures were infected with Sindbis virus. For some experiments, slices were treated with 0.5 μM DAPT, 20 μM 3-(2-carboxypiperazin-4-yl)propyl-1-phosphonic acid (CPP), or 0.2 nM EpoD starting on day 11 in vitro. Cultures were fixed at day 3 post infection. For preparation of lysates, cultured hippocampal slices were visually inspected on day 15 in vitro. Slices with similar infection rates were lysed in RIPA buffer as described previously [5].

Confocal microscopy, image processing and semi-automated analysis of neuronal morphology

Confocal high-resolution microscopy of complete principal hippocampal neurons was performed as described previously [22]. Each single neuron was imaged in 8 to 12 overlapping image stacks with voxel size of 0.14 × 0.14 × 0.44 μm in x-y-z directions. 3D reconstruction of whole neurons was performed using Neuromantic software (University of Reading, Reading, UK) in semi-automated mode.
For the determination of dendritic tau levels, non overlapping basal dendritic segments of 2nd or 3rd order of CA1 neurons expressing EGFP-tagged tau constructs were selected. Image stacks between first and last appearance of the respective dendritic segment were projected onto a single layer and mean intensity values in the middle of the process were determined and corrected for the detector gain.

Culture of PC12 and HEK cells

PC12 cells stably transfected to express PAGFP-tagged wt 352 htau were cultured in serum-DMEM in the presence of 250 μg/ml Geneticin as described previously [53]. Cells were plated onto 10-cm collagen-coated TC-plates at 2 × 104 cells/cm2. On the next day, medium was replaced with low serum DMEM (DMEM with 1 % (vol/vol) serum) containing 100 ng/ml 7S mouse NGF (Alomone Laboratories). After 4 days, medium was replaced with serum-free medium (NB/B-27, Thermo Fisher Scientific) containing 100 ng/ml 7S mouse NGF and supernatant of Aβ-producing HEK cells (HEK-SW) to yield 0.7 or 3.5 nM of Aβ. Same amounts of supernatant of untransfected HEK cells (HEK-con) were added as control. Cell lysates were prepared 24 h. later as described by [53].
HEK-SW cells (HEK 293-APPswe; [54]), which were stably transfected with APP carrying the Swedish familial AD mutation resulting in increased Aβ secretion, were obtained from C. Haass (University of Munich) and cultured in DMEM containing 10 % fetal calf serum. As a control, HEK 293FT cells (Life Technologies) were used. Cells were cultured until 60–70 % confluency, medium was exchanged and the supernatant collected 24 h. later. The supernatant was filtered (0.22 μm pore size) and shock frozen in aliquots in liquid nitrogen. The concentration of Aβ1–40 and Aβ1–42 was determined by ELISAs according to the manufacturer’s description (EZHS40, EZHS42; Millipore). The ratio of Aβ1–42 to Aβ1–40 in the supernatant of HEK-SW cells was 1:4.

Immunoblot analysis

Same volumes of lysates were subjected to SDS-PAGE, transferred to Immobilon-P (Millipore) and stained. Immunodetection used the following antibodies: Phosphorylation-independent tau antibodies: HT7 (human tau, mouse; Pierce); phosphorylation-dependent tau antibodies: AT270 (pThr181, mouse; Pierce), p214 (Ser-214, rabbit; Biosource), pS262 (pSer262, rabbit; Biosource). Also used were anti-GFP (rabbit; Invitrogen), anti-tubulin (mouse, DM1A), anti-acetylated tubulin (mouse; 6-11B-1), and anti-actin (mouse, JLA20; Amersham) antibodies. As secondary antibodies, peroxidase–conjugated goat anti–mouse and anti–rabbit antibodies (Jackson ImmunoResearch Laboratories, Inc.) were used. Detection using enhanced chemiluminiscence and quantitation were performed as described previously [17].

Statistical analysis

Statistical evaluation was performed using one way analysis of variance (ANOVA) with post hoc Fisher's least significant difference (LSD) test for multiple comparisons and two-tailed, unpaired Student’s t test for comparison of the two genotypes at control conditions. For the tau phosphorylation experiments, one-sample Student’s t test was performed. Data are shown as mean and s.e.m. Sholl analysis [23] was performed separately for basal and apical parts of dendritic trees.

Acknowledgements

Funds have been provided by the Deutsche Forschungsgemeinschaft (DFG BR1192/11-2), the Alzheimer Foschungsinitiative (AFI) (to L.B.) and the National Institute of Health (U01 AG029213-01A2). We thank Christian Haass (University of Munich) for providing HEK-SW cells. We appreciate the help of Fredrik Sündermann with setting up the morphological analysis software and Anna Beck and Vanessa Herkenhoff with performing Western blot analysis.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

NG carried out slice culture experiments, performed image analysis, performed statistical analysis and helped to draft the manuscript; LP participated in organotypic culture preparations and performed statistical analysis; CB synthesized epothilone D, participated in the design of the study and helped to draft the manuscript; AS participated in the design of the study and helped to draft the manuscript; LB prepared the Sindbis virus, performed image analysis, participated in the design of the study and drafted the manuscript, RB designed and coordinated the study and drafted the manuscript. All authors read and approved the final manuscript.
Literatur
1.
4.
Zurück zum Zitat Gotz J, Ittner LM. Animal models of Alzheimer's disease and frontotemporal dementia. Nat Rev Neurosci. 2008;9:532–44.CrossRefPubMed Gotz J, Ittner LM. Animal models of Alzheimer's disease and frontotemporal dementia. Nat Rev Neurosci. 2008;9:532–44.CrossRefPubMed
5.
Zurück zum Zitat Tackenberg C, Brandt R. Divergent pathways mediate spine alterations and cell death induced by amyloid-beta, wild-type tau, and R406W tau. J Neurosci. 2009;29:14439–50.CrossRefPubMed Tackenberg C, Brandt R. Divergent pathways mediate spine alterations and cell death induced by amyloid-beta, wild-type tau, and R406W tau. J Neurosci. 2009;29:14439–50.CrossRefPubMed
6.
Zurück zum Zitat Zempel H, Thies E, Mandelkow E, Mandelkow EM. Abeta oligomers cause localized Ca(2+) elevation, missorting of endogenous Tau into dendrites, Tau phosphorylation, and destruction of microtubules and spines. J Neurosci. 2010;30:11938–50.CrossRefPubMed Zempel H, Thies E, Mandelkow E, Mandelkow EM. Abeta oligomers cause localized Ca(2+) elevation, missorting of endogenous Tau into dendrites, Tau phosphorylation, and destruction of microtubules and spines. J Neurosci. 2010;30:11938–50.CrossRefPubMed
7.
Zurück zum Zitat Spires TL, Meyer-Luehmann M, Stern EA, McLean PJ, Skoch J, Nguyen PT, et al. Dendritic spine abnormalities in amyloid precursor protein transgenic mice demonstrated by gene transfer and intravital multiphoton microscopy. J Neurosci. 2005;25:7278–87.PubMedCentralCrossRefPubMed Spires TL, Meyer-Luehmann M, Stern EA, McLean PJ, Skoch J, Nguyen PT, et al. Dendritic spine abnormalities in amyloid precursor protein transgenic mice demonstrated by gene transfer and intravital multiphoton microscopy. J Neurosci. 2005;25:7278–87.PubMedCentralCrossRefPubMed
8.
Zurück zum Zitat Wu HY, Hudry E, Hashimoto T, Kuchibhotla K, Rozkalne A, Fan Z, et al. Amyloid beta induces the morphological neurodegenerative triad of spine loss, dendritic simplification, and neuritic dystrophies through calcineurin activation. J Neurosci. 2010;30:2636–49.PubMedCentralCrossRefPubMed Wu HY, Hudry E, Hashimoto T, Kuchibhotla K, Rozkalne A, Fan Z, et al. Amyloid beta induces the morphological neurodegenerative triad of spine loss, dendritic simplification, and neuritic dystrophies through calcineurin activation. J Neurosci. 2010;30:2636–49.PubMedCentralCrossRefPubMed
9.
Zurück zum Zitat Anderton BH, Callahan L, Coleman P, Davies P, Flood D, Jicha GA, et al. Dendritic changes in Alzheimer's disease and factors that may underlie these changes. Prog Neurobiol. 1998;55:595–609.CrossRefPubMed Anderton BH, Callahan L, Coleman P, Davies P, Flood D, Jicha GA, et al. Dendritic changes in Alzheimer's disease and factors that may underlie these changes. Prog Neurobiol. 1998;55:595–609.CrossRefPubMed
10.
Zurück zum Zitat Coleman PD, Flood DG. Neuron numbers and dendritic extent in normal aging and Alzheimer's disease. Neurobiol Aging. 1987;8:521–45.CrossRefPubMed Coleman PD, Flood DG. Neuron numbers and dendritic extent in normal aging and Alzheimer's disease. Neurobiol Aging. 1987;8:521–45.CrossRefPubMed
11.
Zurück zum Zitat Flood DG. Region-specific stability of dendritic extent in normal human aging and regression in Alzheimer's disease. II. Subiculum. Brain Res. 1991;540:83–95.CrossRefPubMed Flood DG. Region-specific stability of dendritic extent in normal human aging and regression in Alzheimer's disease. II. Subiculum. Brain Res. 1991;540:83–95.CrossRefPubMed
12.
Zurück zum Zitat Flood DG, Buell SJ, Horwitz GJ, Coleman PD. Dendritic extent in human dentate gyrus granule cells in normal aging and senile dementia. Brain Res. 1987;402:205–16.CrossRefPubMed Flood DG, Buell SJ, Horwitz GJ, Coleman PD. Dendritic extent in human dentate gyrus granule cells in normal aging and senile dementia. Brain Res. 1987;402:205–16.CrossRefPubMed
13.
Zurück zum Zitat Flood DG, Guarnaccia M, Coleman PD. Dendritic extent in human CA2-3 hippocampal pyramidal neurons in normal aging and senile dementia. Brain Res. 1987;409:88–96.CrossRefPubMed Flood DG, Guarnaccia M, Coleman PD. Dendritic extent in human CA2-3 hippocampal pyramidal neurons in normal aging and senile dementia. Brain Res. 1987;409:88–96.CrossRefPubMed
14.
Zurück zum Zitat West MJ, Coleman PD, Flood DG, Troncoso JC. Differences in the pattern of hippocampal neuronal loss in normal ageing and Alzheimer's disease. Lancet. 1994;344:769–72.CrossRefPubMed West MJ, Coleman PD, Flood DG, Troncoso JC. Differences in the pattern of hippocampal neuronal loss in normal ageing and Alzheimer's disease. Lancet. 1994;344:769–72.CrossRefPubMed
15.
Zurück zum Zitat Catala I, Ferrer I, Galofre E, Fabregues I. Decreased numbers of dendritic spines on cortical pyramidal neurons in dementia. A quantitative Golgi study on biopsy samples. Hum Neurobiol. 1988;6:255–9.PubMed Catala I, Ferrer I, Galofre E, Fabregues I. Decreased numbers of dendritic spines on cortical pyramidal neurons in dementia. A quantitative Golgi study on biopsy samples. Hum Neurobiol. 1988;6:255–9.PubMed
16.
Zurück zum Zitat Conde C, Caceres A. Microtubule assembly, organization and dynamics in axons and dendrites. Nat Rev Neurosci. 2009;10:319–32.CrossRefPubMed Conde C, Caceres A. Microtubule assembly, organization and dynamics in axons and dendrites. Nat Rev Neurosci. 2009;10:319–32.CrossRefPubMed
17.
Zurück zum Zitat Janning D, Igaev M, Sundermann F, Bruhmann J, Beutel O, Heinisch JJ, et al. Single-molecule tracking of tau reveals fast kiss-and-hop interaction with microtubules in living neurons. Mol Biol Cell. 2014;25:3541–51.PubMedCentralCrossRefPubMed Janning D, Igaev M, Sundermann F, Bruhmann J, Beutel O, Heinisch JJ, et al. Single-molecule tracking of tau reveals fast kiss-and-hop interaction with microtubules in living neurons. Mol Biol Cell. 2014;25:3541–51.PubMedCentralCrossRefPubMed
18.
Zurück zum Zitat Ittner LM, Ke YD, Delerue F, Bi M, Gladbach A, van Eersel J, et al. Dendritic function of tau mediates amyloid-beta toxicity in Alzheimer's disease mouse models. Cell. 2010;142:387–97.CrossRefPubMed Ittner LM, Ke YD, Delerue F, Bi M, Gladbach A, van Eersel J, et al. Dendritic function of tau mediates amyloid-beta toxicity in Alzheimer's disease mouse models. Cell. 2010;142:387–97.CrossRefPubMed
19.
Zurück zum Zitat Papasozomenos SC, Binder LI. Phosphorylation determines two distinct species of Tau in the central nervous system. Cell Motil Cytoskeleton. 1987;8:210–26.CrossRefPubMed Papasozomenos SC, Binder LI. Phosphorylation determines two distinct species of Tau in the central nervous system. Cell Motil Cytoskeleton. 1987;8:210–26.CrossRefPubMed
20.
Zurück zum Zitat Dickstein DL, Brautigam H, Stockton Jr SD, Schmeidler J, Hof PR. Changes in dendritic complexity and spine morphology in transgenic mice expressing human wild-type tau. Brain Struct Funct. 2010;214:161–79.PubMedCentralCrossRefPubMed Dickstein DL, Brautigam H, Stockton Jr SD, Schmeidler J, Hof PR. Changes in dendritic complexity and spine morphology in transgenic mice expressing human wild-type tau. Brain Struct Funct. 2010;214:161–79.PubMedCentralCrossRefPubMed
21.
Zurück zum Zitat Shahani N, Subramaniam S, Wolf T, Tackenberg C, Brandt R. Tau aggregation and progressive neuronal degeneration in the absence of changes in spine density and morphology after targeted expression of Alzheimer's disease-relevant tau constructs in organotypic hippocampal slices. J Neurosci. 2006;26:6103–14.CrossRefPubMed Shahani N, Subramaniam S, Wolf T, Tackenberg C, Brandt R. Tau aggregation and progressive neuronal degeneration in the absence of changes in spine density and morphology after targeted expression of Alzheimer's disease-relevant tau constructs in organotypic hippocampal slices. J Neurosci. 2006;26:6103–14.CrossRefPubMed
22.
Zurück zum Zitat Golovyashkina N, Sündermann F, Brandt R, Bakota L. Reconstruction and morphometric analysis of hippocampal neurons from mice expressing fluorescent proteins. NeuroMethods. 2014;87:251–62.CrossRef Golovyashkina N, Sündermann F, Brandt R, Bakota L. Reconstruction and morphometric analysis of hippocampal neurons from mice expressing fluorescent proteins. NeuroMethods. 2014;87:251–62.CrossRef
23.
Zurück zum Zitat Sholl DA. Dendritic organization in the neurons of the visual and motor cortices of the cat. J Anat. 1953;87:387–406.PubMedCentralPubMed Sholl DA. Dendritic organization in the neurons of the visual and motor cortices of the cat. J Anat. 1953;87:387–406.PubMedCentralPubMed
24.
Zurück zum Zitat Dovey HF, John V, Anderson JP, Chen LZ, de Saint Andrieu P, Fang LY, et al. Functional gamma-secretase inhibitors reduce beta-amyloid peptide levels in brain. J Neurochem. 2001;76:173–81.CrossRefPubMed Dovey HF, John V, Anderson JP, Chen LZ, de Saint Andrieu P, Fang LY, et al. Functional gamma-secretase inhibitors reduce beta-amyloid peptide levels in brain. J Neurochem. 2001;76:173–81.CrossRefPubMed
25.
Zurück zum Zitat Tackenberg C, Grinschgl S, Trutzel A, Santuccione AC, Frey MC, Konietzko U, et al. NMDA receptor subunit composition determines beta-amyloid-induced neurodegeneration and synaptic loss. Cell Death Dis. 2013;4:e608.PubMedCentralCrossRefPubMed Tackenberg C, Grinschgl S, Trutzel A, Santuccione AC, Frey MC, Konietzko U, et al. NMDA receptor subunit composition determines beta-amyloid-induced neurodegeneration and synaptic loss. Cell Death Dis. 2013;4:e608.PubMedCentralCrossRefPubMed
26.
Zurück zum Zitat Shankar GM, Bloodgood BL, Townsend M, Walsh DM, Selkoe DJ, Sabatini BL. Natural oligomers of the Alzheimer amyloid-beta protein induce reversible synapse loss by modulating an NMDA-type glutamate receptor-dependent signaling pathway. J Neurosci. 2007;27:2866–75.CrossRefPubMed Shankar GM, Bloodgood BL, Townsend M, Walsh DM, Selkoe DJ, Sabatini BL. Natural oligomers of the Alzheimer amyloid-beta protein induce reversible synapse loss by modulating an NMDA-type glutamate receptor-dependent signaling pathway. J Neurosci. 2007;27:2866–75.CrossRefPubMed
27.
Zurück zum Zitat Harris EW, Ganong AH, Monaghan DT, Watkins JC, Cotman CW. Action of 3-((+/−)-2-carboxypiperazin-4-yl)-propyl-1-phosphonic acid (CPP): a new and highly potent antagonist of N-methyl-D-aspartate receptors in the hippocampus. Brain Res. 1986;382:174–7.CrossRefPubMed Harris EW, Ganong AH, Monaghan DT, Watkins JC, Cotman CW. Action of 3-((+/−)-2-carboxypiperazin-4-yl)-propyl-1-phosphonic acid (CPP): a new and highly potent antagonist of N-methyl-D-aspartate receptors in the hippocampus. Brain Res. 1986;382:174–7.CrossRefPubMed
28.
Zurück zum Zitat Brunden KR, Zhang B, Carroll J, Yao Y, Potuzak JS, Hogan AM, et al. Epothilone D improves microtubule density, axonal integrity, and cognition in a transgenic mouse model of tauopathy. J Neurosci. 2010;30:13861–6.PubMedCentralCrossRefPubMed Brunden KR, Zhang B, Carroll J, Yao Y, Potuzak JS, Hogan AM, et al. Epothilone D improves microtubule density, axonal integrity, and cognition in a transgenic mouse model of tauopathy. J Neurosci. 2010;30:13861–6.PubMedCentralCrossRefPubMed
29.
Zurück zum Zitat Lou K, Yao Y, Hoye AT, James MJ, Cornec AS, Hyde E, et al. Brain-penetrant, orally bioavailable microtubule-stabilizing small molecules are potential candidate therapeutics for Alzheimer's disease and related tauopathies. J Med Chem. 2014;57:6116–27.PubMedCentralCrossRefPubMed Lou K, Yao Y, Hoye AT, James MJ, Cornec AS, Hyde E, et al. Brain-penetrant, orally bioavailable microtubule-stabilizing small molecules are potential candidate therapeutics for Alzheimer's disease and related tauopathies. J Med Chem. 2014;57:6116–27.PubMedCentralCrossRefPubMed
30.
Zurück zum Zitat Chiorazzi A, Nicolini G, Canta A, Oggioni N, Rigolio R, Cossa G, et al. Experimental epothilone B neurotoxicity: results of in vitro and in vivo studies. Neurobiol Dis. 2009;35:270–7.CrossRefPubMed Chiorazzi A, Nicolini G, Canta A, Oggioni N, Rigolio R, Cossa G, et al. Experimental epothilone B neurotoxicity: results of in vitro and in vivo studies. Neurobiol Dis. 2009;35:270–7.CrossRefPubMed
31.
Zurück zum Zitat Eidenmuller J, Fath T, Hellwig A, Reed J, Sontag E, Brandt R. Structural and functional implications of tau hyperphosphorylation: information from phosphorylation-mimicking mutated tau proteins. Biochemistry. 2000;39:13166–75.CrossRefPubMed Eidenmuller J, Fath T, Hellwig A, Reed J, Sontag E, Brandt R. Structural and functional implications of tau hyperphosphorylation: information from phosphorylation-mimicking mutated tau proteins. Biochemistry. 2000;39:13166–75.CrossRefPubMed
33.
Zurück zum Zitat Tokutake T, Kasuga K, Yajima R, Sekine Y, Tezuka T, Nishizawa M, et al. Hyperphosphorylation of Tau induced by naturally secreted amyloid-beta at nanomolar concentrations is modulated by insulin-dependent Akt-GSK3beta signaling pathway. J Biol Chem. 2012;287:35222–33.PubMedCentralCrossRefPubMed Tokutake T, Kasuga K, Yajima R, Sekine Y, Tezuka T, Nishizawa M, et al. Hyperphosphorylation of Tau induced by naturally secreted amyloid-beta at nanomolar concentrations is modulated by insulin-dependent Akt-GSK3beta signaling pathway. J Biol Chem. 2012;287:35222–33.PubMedCentralCrossRefPubMed
34.
Zurück zum Zitat Drewes G, Trinczek B, Illenberger S, Biernat J, Schmitt-Ulms G, Meyer HE, et al. Microtubule-associated protein/microtubule affinity-regulating kinase (p110mark). A novel protein kinase that regulates tau-microtubule interactions and dynamic instability by phosphorylation at the Alzheimer-specific site serine 262. J Biol Chem. 1995;270:7679–88.CrossRefPubMed Drewes G, Trinczek B, Illenberger S, Biernat J, Schmitt-Ulms G, Meyer HE, et al. Microtubule-associated protein/microtubule affinity-regulating kinase (p110mark). A novel protein kinase that regulates tau-microtubule interactions and dynamic instability by phosphorylation at the Alzheimer-specific site serine 262. J Biol Chem. 1995;270:7679–88.CrossRefPubMed
35.
Zurück zum Zitat Wayman GA, Impey S, Marks D, Saneyoshi T, Grant WF, Derkach V, et al. Activity-dependent dendritic arborization mediated by CaM-kinase I activation and enhanced CREB-dependent transcription of Wnt-2. Neuron. 2006;50:897–909.CrossRefPubMed Wayman GA, Impey S, Marks D, Saneyoshi T, Grant WF, Derkach V, et al. Activity-dependent dendritic arborization mediated by CaM-kinase I activation and enhanced CREB-dependent transcription of Wnt-2. Neuron. 2006;50:897–909.CrossRefPubMed
36.
Zurück zum Zitat Balu DT, Basu AC, Corradi JP, Cacace AM, Coyle JT. The NMDA receptor co-agonists, D-serine and glycine, regulate neuronal dendritic architecture in the somatosensory cortex. Neurobiol Dis. 2012;45:671–82.PubMedCentralCrossRefPubMed Balu DT, Basu AC, Corradi JP, Cacace AM, Coyle JT. The NMDA receptor co-agonists, D-serine and glycine, regulate neuronal dendritic architecture in the somatosensory cortex. Neurobiol Dis. 2012;45:671–82.PubMedCentralCrossRefPubMed
37.
Zurück zum Zitat Gonzalez-Billault C, Munoz-Llancao P, Henriquez DR, Wojnacki J, Conde C, Caceres A. The role of small GTPases in neuronal morphogenesis and polarity. Cytoskeleton (Hoboken). 2012;69:464–85.CrossRef Gonzalez-Billault C, Munoz-Llancao P, Henriquez DR, Wojnacki J, Conde C, Caceres A. The role of small GTPases in neuronal morphogenesis and polarity. Cytoskeleton (Hoboken). 2012;69:464–85.CrossRef
38.
Zurück zum Zitat Quassollo G, Wojnacki J, Salas DA, Gastaldi L, Marzolo MP, Conde C, et al. A RhoA signaling pathway regulates dendritic Golgi outpost formation. Curr Biol. 2015;25:971–82.CrossRefPubMed Quassollo G, Wojnacki J, Salas DA, Gastaldi L, Marzolo MP, Conde C, et al. A RhoA signaling pathway regulates dendritic Golgi outpost formation. Curr Biol. 2015;25:971–82.CrossRefPubMed
39.
Zurück zum Zitat Hamano T, Yen SH, Gendron T, Ko LW, Kuriyama M. Pitavastatin decreases tau levels via the inactivation of Rho/ROCK. Neurobiol Aging. 2012;33:2306–20.CrossRefPubMed Hamano T, Yen SH, Gendron T, Ko LW, Kuriyama M. Pitavastatin decreases tau levels via the inactivation of Rho/ROCK. Neurobiol Aging. 2012;33:2306–20.CrossRefPubMed
40.
41.
Zurück zum Zitat Jin M, Shepardson N, Yang T, Chen G, Walsh D, Selkoe DJ. Soluble amyloid beta-protein dimers isolated from Alzheimer cortex directly induce Tau hyperphosphorylation and neuritic degeneration. Proc Natl Acad Sci U S A. 2011;108:5819–24.PubMedCentralCrossRefPubMed Jin M, Shepardson N, Yang T, Chen G, Walsh D, Selkoe DJ. Soluble amyloid beta-protein dimers isolated from Alzheimer cortex directly induce Tau hyperphosphorylation and neuritic degeneration. Proc Natl Acad Sci U S A. 2011;108:5819–24.PubMedCentralCrossRefPubMed
42.
Zurück zum Zitat Rapoport M, Dawson HN, Binder LI, Vitek MP, Ferreira A. Tau is essential to beta -amyloid-induced neurotoxicity. Proc Natl Acad Sci U S A. 2002;99:6364–9.PubMedCentralCrossRefPubMed Rapoport M, Dawson HN, Binder LI, Vitek MP, Ferreira A. Tau is essential to beta -amyloid-induced neurotoxicity. Proc Natl Acad Sci U S A. 2002;99:6364–9.PubMedCentralCrossRefPubMed
43.
Zurück zum Zitat Maas T, Eidenmuller J, Brandt R. Interaction of tau with the neural membrane cortex is regulated by phosphorylation at sites that are modified in paired helical filaments. J Biol Chem. 2000;275:15733–40.CrossRefPubMed Maas T, Eidenmuller J, Brandt R. Interaction of tau with the neural membrane cortex is regulated by phosphorylation at sites that are modified in paired helical filaments. J Biol Chem. 2000;275:15733–40.CrossRefPubMed
44.
Zurück zum Zitat Wei Q, Holzer M, Brueckner MK, Liu Y, Arendt T. Dephosphorylation of tau protein by calcineurin triturated into neural living cells. Cell Mol Neurobiol. 2002;22:13–24.CrossRefPubMed Wei Q, Holzer M, Brueckner MK, Liu Y, Arendt T. Dephosphorylation of tau protein by calcineurin triturated into neural living cells. Cell Mol Neurobiol. 2002;22:13–24.CrossRefPubMed
45.
Zurück zum Zitat Rahman A, Grundke-Iqbal I, Iqbal K. PP2B isolated from human brain preferentially dephosphorylates Ser-262 and Ser-396 of the Alzheimer disease abnormally hyperphosphorylated tau. J Neural Transm. 2006;113:219–30.CrossRefPubMed Rahman A, Grundke-Iqbal I, Iqbal K. PP2B isolated from human brain preferentially dephosphorylates Ser-262 and Ser-396 of the Alzheimer disease abnormally hyperphosphorylated tau. J Neural Transm. 2006;113:219–30.CrossRefPubMed
46.
Zurück zum Zitat Shipton OA, Paulsen O. GluN2A and GluN2B subunit-containing NMDA receptors in hippocampal plasticity. Philos Trans R Soc Lond B Biol Sci. 2014;369:20130163.PubMedCentralCrossRefPubMed Shipton OA, Paulsen O. GluN2A and GluN2B subunit-containing NMDA receptors in hippocampal plasticity. Philos Trans R Soc Lond B Biol Sci. 2014;369:20130163.PubMedCentralCrossRefPubMed
47.
Zurück zum Zitat Iqbal K, Gong CX, Liu F. Microtubule-associated protein tau as a therapeutic target in Alzheimer's disease. Expert Opin Ther Targets. 2014;18:307–18.CrossRefPubMed Iqbal K, Gong CX, Liu F. Microtubule-associated protein tau as a therapeutic target in Alzheimer's disease. Expert Opin Ther Targets. 2014;18:307–18.CrossRefPubMed
48.
Zurück zum Zitat Blanchard V, Moussaoui S, Czech C, Touchet N, Bonici B, Planche M, et al. Time sequence of maturation of dystrophic neurites associated with Abeta deposits in APP/PS1 transgenic mice. Exp Neurol. 2003;184:247–63.CrossRefPubMed Blanchard V, Moussaoui S, Czech C, Touchet N, Bonici B, Planche M, et al. Time sequence of maturation of dystrophic neurites associated with Abeta deposits in APP/PS1 transgenic mice. Exp Neurol. 2003;184:247–63.CrossRefPubMed
49.
Zurück zum Zitat Leschik J, Welzel A, Weissmann C, Eckert A, Brandt R. Inverse and distinct modulation of tau-dependent neurodegeneration by presenilin 1 and amyloid-beta in cultured cortical neurons: evidence that tau phosphorylation is the limiting factor in amyloid-beta-induced cell death. J Neurochem. 2007;101:1303–15.CrossRefPubMed Leschik J, Welzel A, Weissmann C, Eckert A, Brandt R. Inverse and distinct modulation of tau-dependent neurodegeneration by presenilin 1 and amyloid-beta in cultured cortical neurons: evidence that tau phosphorylation is the limiting factor in amyloid-beta-induced cell death. J Neurochem. 2007;101:1303–15.CrossRefPubMed
50.
Zurück zum Zitat Lee CB, Wu Z, Zhang F, Chappell MD, Stachel SJ, Chou TC, et al. Insights into long-range structural effects on the stereochemistry of aldol condensations: a practical total synthesis of desoxyepothilone F. J Am Chem Soc. 2001;123:5249–59.CrossRefPubMed Lee CB, Wu Z, Zhang F, Chappell MD, Stachel SJ, Chou TC, et al. Insights into long-range structural effects on the stereochemistry of aldol condensations: a practical total synthesis of desoxyepothilone F. J Am Chem Soc. 2001;123:5249–59.CrossRefPubMed
51.
Zurück zum Zitat Rivkin A, Yoshimura F, Gabarda AE, Cho YS, Chou TC, Dong H, et al. Discovery of (E)-9,10-dehydroepothilones through chemical synthesis: on the emergence of 26-trifluoro-(E)-9,10-dehydro-12,13-desoxyepothilone B as a promising anticancer drug candidate. J Am Chem Soc. 2004;126:10913–22.CrossRefPubMed Rivkin A, Yoshimura F, Gabarda AE, Cho YS, Chou TC, Dong H, et al. Discovery of (E)-9,10-dehydroepothilones through chemical synthesis: on the emergence of 26-trifluoro-(E)-9,10-dehydro-12,13-desoxyepothilone B as a promising anticancer drug candidate. J Am Chem Soc. 2004;126:10913–22.CrossRefPubMed
52.
Zurück zum Zitat Sundermann F, Golovyashkina N, Tackenberg C, Brandt R, Bakota L. High-resolution imaging and evaluation of spines in organotypic hippocampal slice cultures. Methods Mol Biol. 2012;846:277–93.CrossRefPubMed Sundermann F, Golovyashkina N, Tackenberg C, Brandt R, Bakota L. High-resolution imaging and evaluation of spines in organotypic hippocampal slice cultures. Methods Mol Biol. 2012;846:277–93.CrossRefPubMed
53.
Zurück zum Zitat Gauthier-Kemper A, Weissmann C, Golovyashkina N, Sebo-Lemke Z, Drewes G, Gerke V, et al. The frontotemporal dementia mutation R406W blocks tau's interaction with the membrane in an annexin A2-dependent manner. J Cell Biol. 2011;192:647–61.PubMedCentralCrossRefPubMed Gauthier-Kemper A, Weissmann C, Golovyashkina N, Sebo-Lemke Z, Drewes G, Gerke V, et al. The frontotemporal dementia mutation R406W blocks tau's interaction with the membrane in an annexin A2-dependent manner. J Cell Biol. 2011;192:647–61.PubMedCentralCrossRefPubMed
54.
Zurück zum Zitat Citron M, Oltersdorf T, Haass C, McConlogue L, Hung AY, Seubert P, et al. Mutation of the beta-amyloid precursor protein in familial Alzheimer's disease increases beta-protein production. Nature. 1992;360:672–4.CrossRefPubMed Citron M, Oltersdorf T, Haass C, McConlogue L, Hung AY, Seubert P, et al. Mutation of the beta-amyloid precursor protein in familial Alzheimer's disease increases beta-protein production. Nature. 1992;360:672–4.CrossRefPubMed
Metadaten
Titel
Region-specific dendritic simplification induced by Aβ, mediated by tau via dysregulation of microtubule dynamics: a mechanistic distinct event from other neurodegenerative processes
verfasst von
Nataliya Golovyashkina
Lorène Penazzi
Carlo Ballatore
Amos B. Smith III
Lidia Bakota
Roland Brandt
Publikationsdatum
01.12.2015
Verlag
BioMed Central
Erschienen in
Molecular Neurodegeneration / Ausgabe 1/2015
Elektronische ISSN: 1750-1326
DOI
https://doi.org/10.1186/s13024-015-0049-0

Weitere Artikel der Ausgabe 1/2015

Molecular Neurodegeneration 1/2015 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Akuter Schwindel: Wann lohnt sich eine MRT?

28.04.2024 Schwindel Nachrichten

Akuter Schwindel stellt oft eine diagnostische Herausforderung dar. Wie nützlich dabei eine MRT ist, hat eine Studie aus Finnland untersucht. Immerhin einer von sechs Patienten wurde mit akutem ischämischem Schlaganfall diagnostiziert.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Frühe Alzheimertherapie lohnt sich

25.04.2024 AAN-Jahrestagung 2024 Nachrichten

Ist die Tau-Last noch gering, scheint der Vorteil von Lecanemab besonders groß zu sein. Und beginnen Erkrankte verzögert mit der Behandlung, erreichen sie nicht mehr die kognitive Leistung wie bei einem früheren Start. Darauf deuten neue Analysen der Phase-3-Studie Clarity AD.

Viel Bewegung in der Parkinsonforschung

25.04.2024 Parkinson-Krankheit Nachrichten

Neue arznei- und zellbasierte Ansätze, Frühdiagnose mit Bewegungssensoren, Rückenmarkstimulation gegen Gehblockaden – in der Parkinsonforschung tut sich einiges. Auf dem Deutschen Parkinsonkongress ging es auch viel um technische Innovationen.

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.