ArticlePDF Available

A brain proteomic investigation of rapamycin effects in the Tsc1 +/− mouse model

Authors:

Abstract and Figures

Background Tuberous sclerosis complex (TSC) is a rare monogenic disorder characterized by benign tumors in multiple organs as well as a high prevalence of epilepsy, intellectual disability and autism. TSC is caused by inactivating mutations in the TSC1 or TSC2 genes. Heterozygocity induces hyperactivation of mTOR which can be inhibited by mTOR inhibitors, such as rapamycin, which have proven efficacy in the treatment of TSC-associated symptoms. The aim of the present study was (1) to identify molecular changes associated with social and cognitive deficits in the brain tissue of Tsc1+/− mice and (2) to investigate the molecular effects of rapamycin treatment, which has been shown to ameliorate genotype-related behavioural deficits. Methods Molecular alterations in the frontal cortex and hippocampus of Tsc1+/− and control mice, with or without rapamycin treatment, were investigated. A quantitative mass spectrometry-based shotgun proteomic approach (LC-MSE) was employed as an unbiased method to detect changes in protein levels. Changes identified in the initial profiling stage were validated using selected reaction monitoring (SRM). Protein Set Enrichment Analysis was employed to identify dysregulated pathways. ResultsLC-MSE analysis of Tsc1+/− mice and controls (n = 30) identified 51 proteins changed in frontal cortex and 108 in the hippocampus. Bioinformatic analysis combined with targeted proteomic validation revealed several dysregulated molecular pathways. Using targeted assays, proteomic alterations in the hippocampus validated the pathways “myelination”, “dendrite,” and “oxidative stress”, an upregulation of ribosomal proteins and the mTOR kinase. LC-MSE analysis was also employed on Tsc1+/− and wildtype mice (n = 34) treated with rapamycin or vehicle. Rapamycin treatment exerted a stronger proteomic effect in Tsc1+/− mice with significant changes (mainly decreased expression) in 231 and 106 proteins, respectively. The cellular pathways “oxidative stress” and “apoptosis” were found to be affected in Tsc1+/− mice and the cellular compartments “myelin sheet” and “neurofilaments” were affected by rapamycin treatment. Thirty-three proteins which were altered in Tsc1+/− mice were normalized following rapamycin treatment, amongst them oxidative stress related proteins, myelin-specific and ribosomal proteins. Conclusions Molecular changes in the Tsc1+/− mouse brain were more prominent in the hippocampus compared to the frontal cortex. Pathways linked to myelination and oxidative stress response were prominently affected and, at least in part, normalized following rapamycin treatment. The results could aid in the identification of novel drug targets for the treatment of cognitive, social and psychiatric symptoms in autism spectrum disorders. Similar pathways have also been implicated in other psychiatric and neurodegenerative disorders and could imply similar disease processes. Thus, the potential efficacy of mTOR inhibitors warrants further investigation not only for autism spectrum disorders but also for other neuropsychiatric and neurodegenerative diseases.
Label-free LC-MSE and Label-based SRM analysis of Tsc1+/− and wildtype mice under rapamycin and vehicle treatment identifies distinct proteomic changes. a) Flow chart of the experimental design. b) Significant protein changes in the hippocampus of rapamycin-treated WT and Tsc1+/− mice identified by label-free LC-MSE. Orange dots refer to proteins that are increased in abundance, and green dots represent downregulated proteins following rapamycin treatment. c2 and 4 (see below) refer to significantly changed proteins following rapamycin treatment in wildtype (“treatment effect in Wt mice”) or Tsc1+/− mice (“treatment effect in Tsc1+/− mice”). Protein enrichment analysis was performed on the identified proteins. Yellow and blue dots represent the TSC genotype effect following rapamycin or vehicle treatments (b1 and 2) and are linked to c1 and 3. Proteins changing due to a combination of TSC genotype and rapamycin treatment are labeled black or purple, respectively. b1 Rapamycin induced changes in wildtype mice as compared to Tsc1+/− mice following vehicle treatment. b2 Rapamycin induced changes in Tsc1+/− mice compared to Tsc1+/− alterations following rapamycin treatment. c) Bar plots of genotype and treatment effects identified through global protein profiling and significantly changed proteins identified in the targeted SRM analysis. Number of significant proteins, percentage of up- and downregulated proteins and enriched pathways linked to the up- and downregulated proteins are displayed
… 
This content is subject to copyright. Terms and conditions apply.
R E S E A R C H Open Access
A brain proteomic investigation of
rapamycin effects in the Tsc1
+/
mouse
model
Hendrik Wesseling
1
, Ype Elgersma
2
and Sabine Bahn
1,2*
Abstract
Background: Tuberous sclerosis complex (TSC) is a rare monogenic disorder characterized by benign tumors in
multiple organs as well as a high prevalence of epilepsy, intellectual disability and autism. TSC is caused by
inactivating mutations in the TSC1 or TSC2 genes. Heterozygocity induces hyperactivation of mTOR which can be
inhibited by mTOR inhibitors, such as rapamycin, which have proven efficacy in the treatment of TSC-associated
symptoms. The aim of the present study was (1) to identify molecular changes associated with social and cognitive
deficits in the brain tissue of Tsc1
+/
mice and (2) to investigate the molecular effects of rapamycin treatment,
which has been shown to ameliorate genotype-related behavioural deficits.
Methods: Molecular alterations in the frontal cortex and hippocampus of Tsc1
+/
and control mice, with or without
rapamycin treatment, were investigated. A quantitative mass spectrometry-based shotgun proteomic approach (LC-
MS
E
) was employed as an unbiased method to detect changes in protein levels. Changes identified in the initial
profiling stage were validated using selected reaction monitoring (SRM). Protein Set Enrichment Analysis was
employed to identify dysregulated pathways.
Results: LC-MS
E
analysis of Tsc1
+/
mice and controls (n= 30) identified 51 proteins changed in frontal cortex and 108
in the hippocampus. Bioinformatic analysis combined with targeted proteomic validation revealed several dysregulated
molecular pathways. Using targeted assays, proteomic alterations in the hippocampus validated the pathways
myelination,dendrite,and oxidative stress, an upregulation of ribosomal proteins and the mTOR kinase. LC-MS
E
analysis was also employed on Tsc1
+/
and wildtype mice (n= 34) treated with rapamycin or vehicle. Rapamycin
treatment exerted a stronger proteomic effect in Tsc1
+/
mice with significant changes (mainly decreased expression)
in 231 and 106 proteins, respectively. The cellular pathways oxidative stressand apoptosiswere found to be affected
in Tsc1
+/
mice and the cellular compartments myelin sheetand neurofilamentswere affected by rapamycin
treatment. Thirty-three proteins which were altered in Tsc1
+/
mice were normalized following rapamycin treatment,
amongst them oxidative stress related proteins, myelin-specific and ribosomal proteins.
Conclusions: Molecular changes in the Tsc1
+/
mouse brain were more prominent in the hippocampus compared to
the frontal cortex. Pathways linked to myelination and oxidative stress response were prominently affected and, at least
in part, normalized following rapamycin treatment. The results could aid in the identification of novel drug targets for
the treatment of cognitive, social and psychiatric symptoms in autism spectrum disorders. Similar pathways have also
been implicated in other psychiatric and neurodegenerative disorders and could imply similar disease processes. Thus,
the potential efficacy of mTOR inhibitors warrants further investigation not only for autism spectrum disorders but also
for other neuropsychiatric and neurodegenerative diseases.
Keywords: Tuberous sclerosis, Rapamycin, Proteomics, SRM, Animal model
* Correspondence: sb209@cam.ac.uk
1
Department of Chemical Engineering and Biotechnology, University of
Cambridge, Tennis Court Road, Cambridge CB2 1QT, UK
2
Department of Neuroscience, Erasmus Medical Center, Rotterdam 3000, CA,
The Netherlands
© The Author(s). 2017 Open Access This 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.
Wesseling et al. Molecular Autism (2017) 8:41
DOI 10.1186/s13229-017-0151-y
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Background
Tuberous sclerosis complex (TSC) is a rare multi-system
monogenic hamartomatous disorder, which is caused by
mutations inactivating the TSC1 (hamartin) or TSC2
(tuberin) genes. TSC is characterized by benign tumors
in multiple organs, including the brain, kidneys, heart
and eyes [1]. Over 90% of TSC patients develop epilepsy,
and around 50% present with neuropsychiatric prob-
lems, such as intellectual disability (50%) [2, 3], autism
spectrum disorder (ASD) (1768%), schizophrenia (10
30%) and anxiety disorders (40%) [4], which account for
most of the mortality and morbidity [5].
At the molecular level, both Tsc1 and Tsc2 protein
products form hetero-dimers which inhibit the GTP-
binding protein RHEB (Ras homolog enriched in the
brain). Consequently, mutations within either Tsc1 or
Tsc2 lead to increased levels of activated RHEB [6],
which causes hyperactivation of mammalian target of
rapamycin (mTOR) signaling, a constitutive phosphoryl-
ation of eukaryotic translation initiation factor 4E-
binding protein 1 (4E-BP1) and activation of ribosomal
protein S6 through S6K1 phosphorylation [7, 8]. The net
effect is enhanced protein translation, cell proliferation
and growth [9]. Notably, increased mTOR signaling and
subsequent changes in global protein synthesis are
shared molecular mechanisms of several rare neurodeve-
lopmental disorders with an increased prevalence of
ASD, such as fragile X syndrome (FXS) [10].
The hyperactivation of mTOR induced by Tsc1 and
Tsc2 heterozygosity can be inhibited by mTOR inhibi-
tors, such as the macrolide rapamycin. Rapamycin is an
immunosuppressant, which is widely prescribed to pre-
vent rejection in organ transplantation and exerts anti-
tumor properties [1113]. Rapamycin binds FK-binding
protein 12 (FKBP12), and as a complex, rapamycin-
FKBP12 directly binds to the mTOR complex 1
(mTORC1), thus reducing phosphorylation of down-
stream mTOR targets [14, 15]. Rapamycin and other
mTOR inhibitors have been shown to be efficacious in
the treatment of several TSC-associated tumors as well
as seizures [1619] and may ameliorate the symptoms of
neurodevelopmental disorders in adults [20, 21]. In TSC
mouse models, rapamycin limits tumor growth [22, 23],
reduces neuropathology and ameliorates epileptic sei-
zures as well as learning deficits [2426]. It was recently
reported that rapamycin normalizes social interaction
deficits relevant to core disabilities associated with ASD
in both Tsc1
+/
and Tsc2
+/
mice [27].
Here, we investigated the Tsc 1
+/
mouse model, which
exhibits haploinsufficiency for the Tsc 1 gene, in an attempt
to identify molecular changes associated with the neuro-
psychiatric phenotype of TSC patients [5]. In this mouse
model, the typical human cerebral pathology of spontan-
eous seizures, cerebral lesions and giant dysmorphic cells
could not been detected using immuno-cytochemistry and
high resolution magnetic resonance imaging, respectively
[28]. Furthermore, spine number and dendritic branching
are normal [28]. However, the Tsc1
+/
mouse shows prom-
inent behavioural deficits which mimic core symptoms of
ASD and other neuropsychiatric disorders [28]. Tsc1
+/
mice show hippocampal learning deficits using the Morris
water maze test and contextual fear conditioning, as well
as social deficits indicated by reduced social interaction
and nest building [28]. Consequently, the Tsc1
+/
mouse is
a suitable model to investigate aspects of the molecular
pathology associated with neuropsychiatric spectrum dis-
orders, especially in relation to ASD and intellectual
disability. In this study, we attempted to identify
changes in molecular pathways in the frontal cortex
and hippocampus of the Tsc1
+/
mouse model using a
mass spectrometry-based proteomics approach. We
also investigated protein changes associated with
rapamycin treatment. Findings from this study could
aid in the identification of novel drug targets for the
treatment of cognitive, social and psychiatric symp-
toms in ASD.
Methods
A more detailed description of the materials and
methods used in this study can be found in the supple-
mentary methods section (Additional file 1).
Animals
Tsc1
+/
mice were generated by replacing exons 6
through to 8 of the Tsc1 gene with a selection cassette,
as described previously [29]. This leads to the generation
of Tsc1 null embryos which express Tsc1 transcripts in
which exon 5 and 9 are fused, leading to a premature
TGA stop codon. Consequently, any protein translated
from this allele lacks all of the known functional do-
mains of hamartin including the putative Rho activation
domain. The Tsc1
+/
mutant mouse was crossed six
times into the C57BL/6J OlaHsD background and then
at least three times into the C57BL/6N/HsD back-
ground. The offspring consisted of Tsc1
+/
mice and
wildtype littermates. Mice were genotyped when they
were about 7 days old. They were housed in groups and
kept on a 12-h light/dark cycle, with food and water
available ad libitum. Mice were culled when they were
68 weeks old and genotype groups were sex- and age-
matched for the experiments for consistency with the
published behavioural data [28]. Mouse genotypes were
blinded using codes all the way through to the sample
preparation stage. The codes were un-blinded for the
mass spectrometry analysis since samples had to be dis-
tributed evenly to avoid run time biases. All animal exper-
iments were approved by the Dutch Ethical Committee or
Wesseling et al. Molecular Autism (2017) 8:41 Page 2 of 12
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
in accordance with Institutional Animal Care and Use
Committee guidelines.
Rapamycin treatment
Mice were injected intraperitoneally with 5 mg/kg rapamy-
cin or vehicle for 5 days and culled 24 h after the last injec-
tion [27]. Rapamycin was dissolved in 5% dimethyl sulfoxide
diluted with saline to 5 ml/kg. Mice were 57 weeks old at
thetimeofinjection.
Proteomic sample preparation
Sample preparation was carried out as described previ-
ously [3032]. Based on the lysates, two randomized,
blinded, independent sample preparations were prepared
for liquid chromatography mass spectrometry (in expres-
sion mode; LC-MS
E
) and selected reaction monitoring
mass spectrometry to avoid bias in sample preparations.
Label-free LC-MS
E
proteomic profiling of brain tissue
Brain tissue analysis and data processing were performed
as described previously [31, 33, 34]. The Swiss-Prot hu-
man reference proteome (Uniprot release March 2013,
20,252 entries) was used for protein identification
searches. Protein abundance changes for the compari-
sons between Tsc1
+/
and wildtype were determined by
the MSstats package [35] based on mixed-effect models
using the peptide intensities, following log
2
transform-
ation and exclusion of intensity values deviating by more
than 3 standard deviations from the mean of each
group.
Protein set enrichment analysis
Significantly changed proteins were partitioned into
three bins, according to their ratio: proteins decreased in
abundance (ratio < 1.0), proteins increased in abundance
(ratio > 1.0) and a bin to identify general disturbed path-
ways which included all proteins with increased and de-
creased abundance (ratio > 1 and <1). The Rpackage
database org.mouse.eg.db version 2.8.0 was used for
gene ontology (GO) term annotation based on entrez
gene identifiers and GO-term enrichment analysis was
performed using GOstats.
Label-based SRM mass spectrometry
Abundance alterations of a panel of 43 candidate pro-
teins previously implicated in the Tsc1
+/
mouse path-
ology were measured using a targeted SRM mass
spectrometry approach as described previously [32, 36]
following the guidelines of Lange et al. [37]. SRMstats
was used at default settings [37]. The final transitions,
collision energy and retention time windows used for
each peptide can be requested.
Results
Label-free LC-MS
E
proteomic profiling of Tsc1
+/
mouse
brains
We investigated protein abundance changes in the
frontal cortex and hippocampus of the Tsc1
+/
mouse.
LC-MS
E
analysis resulted in the identification of 522
proteins (7071 peptides) in the frontal cortex and 463
proteins (5149 peptides) in the hippocampus. Of these,
the levels of 51 proteins were altered in the frontal cor-
tex (FDR-adjusted *p< 0.05) and 108 in the hippocam-
pus (FDR-adjusted *p< 0.05). In the frontal cortex, 17 of
the changed proteins were altered by more than 10%, as
were 49 of the 108 changed hippocampal proteins
(Additional file 2). In the case of the frontal cortex, this
included adenylyl cyclase-associated protein 2 (CAP2,
ratio = 0.89, FDR-adjusted *p= 0.013), elongation factor
1α2 (EIF1A2, ratio = 0.97, FDR-adjusted *p = 0.03),
eukaryotic translation initiation factor 3 subunit L (eIF3l,
ratio = 1.17, FDR-adjusted *p = 0.03) and elongation fac-
tor 2 (Eef2, ratio = 0.95, FDR-adjusted *p = 0.05), which
are all regulators of translation. Copine 6 (ratio = 1.1, FDR-
adjusted p= 0.0097) and copine 8 (ratio = 0.8, FDR-
adjusted p=3.9×10
6
), which are associated with synaptic
plasticity, were changed in the hippocampus. Nine proteins
(NCDN⬇⬇, MAP2⬆⬆,SUCB1⬇⬇,MYPR⬆⬇,NDUS7⬇⬇,
DPYL2⬇⬇,AT1A2⬇⬆,CRYM⬇⬆,ARP3⬆⬇)werefoundto
be changed in both frontal cortex (first arrow) and hippo-
campal tissue (second arrow) (Additional file 2).
Gene set enrichment analysis was employed to investi-
gate if the altered 108 and 51 proteins were enriched in
biological pathways and cellular compartments. Based on
GO enrichment analysis, proteins responsible for the bio-
logical pathways reproductive behaviour(p=0.008),
neurological system process(p=0.010)andvisual
learning(p= 0.028) were altered in the frontal cortex of
the Tsc1
+/
mouse. In the hippocampus, the proteins were
related to the biological pathways ribonucleotide energy
metabolism(p= 0.0097), protein polymerisation
(p=0.005)andoxidative stress(p=0.009).Onepath-
way, visual learning, was identified in both the frontal
cortex and hippocampus proteomic analyses. Cellular
compartment GO association enrichment revealed that
the altered proteins were associated with myelination
and dendritein the frontal cortex and myelin sheet
and endoplasmic reticulum-Golgi intermediate compart-
mentin the hippocampus.
Selected reaction monitoring (SRM) validation of Tsc1
+/
brain proteomic alterations
For orthogonal proteomic validation of the proteomics
results, we employed a targeted label-based LC-SRM ap-
proach to specifically quantify the levels of 43 candidate
proteins derived from LC-MS
E
profiling, subsequent path-
way analysis, literature findings and already established in-
Wesseling et al. Molecular Autism (2017) 8:41 Page 3 of 12
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
house assays. Label-based SRM assays are highly specific
and sensitive and outperform immunoblotting analyses for
validation [3841]. This analysis showed that the levels of 5
and 20 of the targeted proteins were significantly changed
in the frontal cortex and hippocampus, respectively
(p< 0.05, Table 1). Specifically, two proteins (MYPR,
PURA) out of five LC-MS
E
candidates were validated in the
frontal cortex and six (NSF, MYPR, MBP, CPNE6, SODC,
MARCS) out of ten LC-MS
E
-derived protein candidates
were validated in the hippocampus (Table 1). Using tar-
geted SRM-assays, we confirmed the more prominent
proteomic alterations in the hippocampus and further vali-
dated GO-terms myelination,dendriteand oxidative
stressthrough confirmation of changes in MYPR, MBP,
TSN2 (all myelin specific proteins), MAP2 (dendritic
marker) and SODC (oxidative stress marker). We further
detected an upregulation in ribosomal proteins and mTOR
kinase in the Tsc 1
+/
mice (Table 1).
Label-free LC-MS
E
proteomic profiling of the Tsc1
+/
hippocampus following rapamycin treatment
To investigate the effects of rapamycin on the brain
proteome, label-free LC-MS
E
analysis was employed on
Tsc1
+/
and wildtype mice treated with rapamycin or ve-
hicle (Fig. 1a). Only the hippocampus was studied in this
case as this brain region was more affected with regard
to significantly changed proteins (Table 1). The hippo-
campus plays not only an important role in cognition,
but hippocampal dysfunction has also been linked to a
wide range of neuropsychiatric symptoms [42, 43]. Defi-
cits in consolidating short- and long-term memory and
spatial navigation have been shown to be impaired in
Tsc1
+/
and Tsc2
+/
mice and were reversed by rapamy-
cin treatment in Tsc2
+/
mice [24]. LC-MS
E
analysis led
to the identification of 8648 total peptides which trans-
lated to 597 proteins, which were detected across all
samples. Interestingly, rapamycin treatment exerted a
stronger proteomic effect in Tsc1
+/
compared to wild-
type mice (Fig. 1c (2 and 4)) with significant changes in
231 and 106 proteins, respectively. An overall decrease
in protein levels was found in both Tsc1
+/
and wildtype
mice.
Next, proteins were tested which were affected in all
four comparisons. This showed that 9 proteins were
changed in common (FRM4A, PEA15, PERQ1, MAP2,
BASP, CLD11, ALBU, TCAL3, CLH) and that the levels
of 54 proteins were affected by rapamycin treatment in
both wildtype and Tsc1
+/
mice; of these proteins, 52
corresponded in their fold change direction (37 of the 52
proteins were decreased in abundance and 15 increased,
respectively). Pathway analysis linked the 52 overlapping
proteins to the biological process of translation
(p= 0.00082), macromolecule biosynthetic process
(p= 0.005) and gene expression(p= 0.014). Using
KEGG (Kyoto Encyclopedia of Genes and Genomes) an-
notation, ribosomewas the most significant pathway
(p= 1.7 × 10
7
) in the enrichment analysis.
We further employed enrichment analysis for the
genotype comparisons and the treatment comparisons
(Fig. 1c (14)). This associated the biological pathways
oxidative stressand apoptosiswith the significantly
changed proteins identified in the Tsc1
+/
vs Wt com-
parison (Fig. 1c (1)). Furthermore, cellular compart-
ments of myelin sheet and neurofilaments were affected
by rapamycin treatment in both Tsc1
+/
and Wt mice
(Fig. 1c (2 and 4)). Proteins with decreased levels due to
rapamycin treatment mostly related to the biological
pathways translation,macromolecular complex as-
semblyand chromosome organization(Fig. 1c (2 and
4)). Downregulation of the pathway chromosome
organizationwas specifically observed in Tsc1
+/
mice
following rapamycin treatment (Fig. 1c (4)).
Importantly, 41 proteins which were altered in vehicle-
treated Tsc1
+/
mice were normalized following rapamy-
cin treatment. These proteins include a set of proteins
where rapamycin treatment normalizes the genotype-
induced protein alterations to wildtype levels (33 pro-
teins) and a set of proteins where rapamycin normalizes
the genotype-effect below or above baseline levels (8
proteins). The former include the Glycine receptor sub-
unit alpha-4 (GLRA4), the Calcium-dependent secretion
activator 1 (CAPS1), Rod cGMP-specific 3,5cyclic
phosphodiesterase beta (PDE6B) and Guanine deami-
nase (GUAD) (Fig. 2 (N)); the latter include Rho-
associated protein kinase 2 (ROCK2) and ribosomal pro-
teins (RS18, RL4, RS9). All ribosomal proteins affected
by rapamycin treatment were decreased in their abun-
dance levels. Furthermore, proteins were identified that
are affected by rapamycin treatment in both wildtype
and mutant mice, although there was no difference in
their abundance levels between vehicle-treated mutant
and wildtype mice (Fig. 2 (R)). This set was comprised of
41 proteins. Amongst them are the anaphase promoting
complex s7 (APC7), calcineurin subunit B type 1
(CANB1) and the GABA aminotransferase (GABT).
Finally, the levels of six proteins were found to be
altered between mutant and wildtype but did not change
following rapamycin treatment. Neuromodulin (NEUM),
the excitatory amino acid transporter 2 (EAAT2) and
SMP25 are examples.
SRM validation of rapamycin treatment effects in the Tsc1
+/
hippocampus
The next phase of the study involved a targeted prote-
omic approach to validate the findings of the rapamycin
study (see Fig. 1c (14) [targeted]). This focused on
myelination deficits, alterations in the translational ma-
chinery and proteins found to be altered in the label-free
Wesseling et al. Molecular Autism (2017) 8:41 Page 4 of 12
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Table 1 Significantly changed proteins identified by label-based LC-SRM in the frontal cortex and hippocampus of Tsc1
+/
mice
compared to wildtype mice
The first stage of the analysis consisted of a global profiling approach, followed by validation with a specific and sensitive label-based assay panel. pvalues were
determined using SRMstats (linear model with fixed subject effects) and corrected (p*) to control for multiple hypothesis testing (Benjamini-Hochberg) [90]. For
reasons of clarity, only ratios and significance levels of significantly changing proteins are shown. For full information, see Additional file 3.n.s. not significant, n.d.
not detected. Validated findings are in gray shading
Wesseling et al. Molecular Autism (2017) 8:41 Page 5 of 12
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
LC-MS
E
discovery study of the Tsc 1
+/
mouse (SODC,
NSF, MAP2, PKCG). The rapamycin-target mTOR was
included as a positive control.
This analysis resulted in validation of increased
myelin-associated proteins in Tsc1
+/
compared to wild-
type mice (Fig. 1c (1 and 3)). Furthermore, the decrease
a
b1
b2
c
Fig. 1 Label-free LC-MSE and Label-based SRM analysis of Tsc1+/and wildtype mice under rapamycin and vehicle treatment identifies distinct proteomic
changes. a) Flow chart of the experimental design. b) Significant protein changes in the hippocampus of rapamycin-treated WT and Tsc1
+/
mice identified
by label-free LC-MS
E
.Orange dots refer to proteins that are increased in abundance, and green dots represent downregulated proteins following rapamycin
treatment. c2and 4(see below) refer to significantly changed proteins following rapamycin treatment in wildtype (treatment effect in Wt mice)orTsc1
+/
mice (treatment effect in Tsc1
+/
mice). Protein enrichment analysis was performed on the identified proteins. Yellow and blue dots represent the TSC
genotype effect following rapamycin or vehicle treatments (b1and 2) and are linked to c1and 3. Proteins changing due to a combination of
TSC genotype and rapamycin treatment are labeled black or purple, respectively. b1Rapamycin induced changes in wildtype mice as
compared to Tsc1
+/
mice following vehicle treatment. b2Rapamycin induced changes in Tsc 1
+/
mice compared to Tsc1
+/
alterations
following rapamycin treatment. c) Bar plots of genotype and treatment effects identified through global protein profiling and significantly
changed proteins identified in the targeted SRM analysis. Number of significant proteins, percentage of up- and downregulated proteins
and enriched pathways linked to the up- and downregulated proteins are displayed
Wesseling et al. Molecular Autism (2017) 8:41 Page 6 of 12
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
in oxidative stress-related proteins in the Tsc1
+/
mouse
was validated by confirming decreased levels of SODC
(Fig. 1c (1)). Additionally, altered levels of NSF and
PKCG were also validated (Fig. 1c [targeted] and Fig. 3).
In the case of rapamycin treatment effects, decreased
levels of proteins involved in translation could be vali-
dated. Specifically, decreased levels of all three tested
ribosomal subunits as well as the mTOR-kinase were
identified. Moreover, the analysis showed that rapamycin
normalized the protein levels of SODC, NSF and PKCG
in Tsc1
+/
mice (Fig. 1c [targeted]).
Discussion
The pathogenesis of psychiatric disorders such as ASD re-
mains elusive, and there is accumulating evidence that
several neuronal circuits and pathways are affected. This
is especially true for the social, cognitive and neuropsychi-
atric symptoms associated with these disorders. In an at-
tempt to gain further insight into these pathways, this
study combines unbiased and targeted proteomic ap-
proaches to investigate the hippocampus and frontal cor-
tex of a mouse model of TSC, which is one of the most
frequent causes of syndromic ASD [44]. The investigated
Tsc 1
+/
mouse model exhibits social and cognitive deficits,
which are core behavioural symptoms of ASD in humans
[45] and other relevant rodent models [46], without any
obvious brain pathology (such as tumors or epilepsy). This
makes the Tsc 1
+/
mouse an excellent model of pharma-
cologically treatable ASD. A previous study has demon-
strated the effectiveness of rapamycin to normalize
reciprocal social interactions in this model [27]. The aim
of the present study was to investigate proteins and path-
ways affected by rapamycin treatment, which could sup-
port drug discovery efforts and in turn the development of
improved treatments for TSC, ASD and possibly other
neuropsychiatric disorders.
Proteomic profiling of the frontal cortex and hippo-
campus brain tissue in this study identified and
Fig. 2 Multivariate analysis of LC-MS
E
estimates as shown by condition plots illustrating the differences between the WT and Tsc1
+/
mice with and
without rapamycin treatment. X-axis is condition and y-axis is log ratio of endogenous (Llight) over reference (Hheavy) peptides. Dots represent the
mean of the log
2
ratio for each condition, and error bars indicate the confidence intervals with 0.95 significance. The interval is not related to the model
based analysis. Significant changes as measured by LC-MS
E
are indicated below each protein. Illustrated are examples of proteins which are affected
by rapamycin treatment and which are normalized by rapamycin treatment. Corrected pvalues (p*) were determined by post hoc correction after
Benjamini-Hochberg [91]. CR = Wt +rapamycin,CS=Wt + vehicle, TR = Tsc1
+/
+rapamycin,TS=Tsc1
+/
rapamycin
Wesseling et al. Molecular Autism (2017) 8:41 Page 7 of 12
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
quantified a large number of significantly changed pro-
teins in the Tsc1
+/
mouse model. Differentially
expressed proteins and altered molecular pathways were
identified and selected candidate proteins were validated
using SRM as a highly quantitative method. In a second
stage, proteomic analysis of rapamycin treatment effects
were investigated to identify down-stream effects of
mTOR-pathway inhibition in the hope to gain new in-
sights into the molecular underpinnings of social impair-
ments in ASD and other psychiatric disorders.
We were able to show that myelin proteins and the
translational machinery, specifically several ribosomal
subunits, were significantly altered in Tsc1
+/
mice
treated with rapamycin. Our findings of lower ribosomal
subunit abundances are consistent with a rapamycin-
induced downregulation of ribosomal biogenesis [47].
Regarding the effects on myelination, previous research
has linked the mTOR pathway to oligodendrocyte differ-
entiation and axonogenesis [48]. Oligodendrocytes pro-
duce myelin, and this is specifically regulated at the late
progenitor to immature oligodendrocyte transition stage
(as shown by changes in expression of the myelin
marker proteins MYPR and MBP). We identified an
increase in myelin proteins in the Tsc1
+/
mouse, but
not in Wt mice. A recent study has shown that ablation
of TSC1 is associated with oligodendrocyte-specific
over-activation and subsequent hypomyelination [49].
An increase in myelin proteins in the Tsc1
+/
mouse
brain may be due to the globally enhanced (Table 1) pro-
tein translation and cell proliferation in the context of
mTOR hyperactivation. An increase in cell growth and
proliferation could interfere with oligodendrocyte matur-
ation and thus result in incomplete myelination as seen
in some demyelinating diseases, such as multiple scler-
osis [50].
MYPR and MBP as well as TSN-2 were amongst the
altered myelin proteins. These proteins play an import-
ant role in oligodendrocyte differentiation during devel-
opment. Furthermore, in vitro studies have shown that
MBP mRNA and protein expression are significantly de-
creased by mTOR inhibition [48, 51]. Inhibiting mTOR
in oligodendrocyte precursor cell/dorsal root ganglion
co-cultures potently abrogated oligodendrocyte differen-
tiation and reduced numbers of myelin segments. Disor-
ganized and structurally compromised axons with poor
myelination have already been found in TSC patients,
Fig. 3 Multivariate analysis of SRM estimates as shown by condition plots illustrating the differences between Wt and Tsc1
+/
mice with and
without rapamycin treatment. X-axis represents the condition and the y-axis the log ratio of endogenous (Llight) over reference (Hheavy)
peptides. Dots represent the mean of log
2
ratio for each condition, and error bars indicate the confidence intervals with 0.95 significance. The
interval is not related to the model based analysis. Significant changes as measured by LC-MS
E
are indicated below each protein. SRM was able
to validate protein abundance changes identified by label-free LC-MS
E
. Corrected pvalues (p*) were determined by post hoc correction after
Benjamini-Hochberg [91]
Wesseling et al. Molecular Autism (2017) 8:41 Page 8 of 12
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
and this may at least to some extent explain the behav-
ioural and cognitive deficits associated with the disorder
[52]. Impaired adult myelination has been shown in the
prefrontal cortex of socially isolated mice [53]. Import-
antly, changes in oligodendrocyte function and myelin-
ation abnormalities are amongst the most consistent
hallmarks of psychiatric pathology in post-mortem brain
studies. Changes were reported for schizophrenia, bipo-
lar disorder, depression and ASD [36, 5458]. In wild-
type mice, rapamycin treatment led to a reduction of
myelin and myelin protein expression [59, 60]. This is
consistent with our findings, where rapamycin affects
both wildtype (reduced) and mutant myelin (increased)
protein expression.
Interestingly, several proteins that we found altered in
the Tsc1
+/
mouse model were reversed by rapamycin
treatment. One of these proteins, a glycine receptor sub-
unit, which abundance was decreased in the mutant and
normalized by rapamycin treatment, could be a potential
drug target for novel treatments of ASD and
schizophrenia-spectrum disorders. The glycine receptor
co-localizes with GABA
A
receptors on hippocampal
neurons [61]. A microdeletion at Xq22.2 implicates
GLRA4 to be involved in intellectual disability and be-
havioural problems [62]. In a case report, glycine recep-
tor antibodies could be detected in a patient with
treatment-resistant focal epilepsy, tantrums, clumsiness
and impaired speech [63] and in patients with progres-
sive encephalomyelitis with rigidity and myoclonus stiff
person syndrome [64]. Treatments targeting the glycine
transporter are under investigation as novel treatment
approaches for schizophrenia [65].
Other altered proteins, which are normalized by rapa-
mycin treatment, included the calcium-dependent secre-
tion activator 1 (CAPS1) (decreased in Tsc1
+/
and
normalized by rapamycin), two guanine metabolism as-
sociated proteins (guanine deaminase and PDE6B; both
increased in Tsc1
+/
and normalized by rapamycin) and
the vesicle-fusing ATPase NSF (increased in Tsc1
+/
,
normalized by rapamycin), a molecular component of
the exocytosis machinery [66], which is required for
membrane fusion [67] and regulates the disassembly of
SNARE complexes on early endosomes [68]. The NSF
gene has also been linked with cocaine dependence [69]
and schizophrenia [36, 70]. Direct interactions with cell
surface receptors such as AMPA receptors [71, 72], β2-
adrenergic receptors [73], dopaminergic receptors [74]
and the adrenomedulin receptor [75] have been re-
ported. Interestingly, a coordinated action of NSF and
PKC regulates GABA
B
receptor signaling efficiency [76].
PKCG was found to be strongly downregulated by rapa-
mycin treatment in this study and is known to be in-
volved in the regulation of the neuronal receptors
GLUR4 and NMDAR1 [77]. It binds and phosphorylates
the GLUR4 glutamate receptor and regulates its function
by increasing membrane-associated GRIA4 expression
[78]. Several preclinical and clinical trials have investi-
gated mGLUR antagonists for the treatment of social
deficits in ASD [78, 79] and ASD associated with FXS
[80, 81] and PKCG inhibitors could represent a novel
treatment strategy to ameliorate cognitive and social def-
icits. Notably, Ketamine, which is thought to exert anti-
depressant action through modulation of mTOR
pathway activity [82], potentiates persistent learning and
memory impairment through the PKCG-ERK signaling
pathway [83].
Another protein strongly downregulated following rapa-
mycin treatment is the anaphase promoting complex S7
(APC7), which is a cell cycle-regulated E3 ubiquitin ligase
controlling progression through mitosis and the G1 phase
of the cell cycle. The control of APC7 through rapamycin
might be a major breakpoint in cell proliferation. Rapamy-
cin has already been shown to also downregulate the ex-
pression of the APC/C inhibitor Emi1 [84].
Copine 6, which we found upregulated in the hippo-
campus of Tsc1
+/
compared to wildtype mice, is a
calcium-dependent regulator of the actin cytoskeleton in
neuronal spines and negatively regulates spine matur-
ation during neuronal development [85]. Changes in
copine 6 expression may be involved in neurodevelop-
mental disorders, as deformed dendritic spines and
changes in spine density are hallmarks of many neurode-
velopmental conditions, such as Downs syndrome [86,
87] and FXS [88]. Interestingly, hippocampi from pa-
tients suffering from uncontrolled epileptic seizures, typ-
ically a problem in tuberous sclerosis patients, exhibit a
decrease in spine density [89]. We also found that
MAP2, a dendritic spine marker, was increased by Tsc1
heterozygocity and decreased by rapamycin treatment.
Interestingly, over twice as many significantly changed
proteins were identified in the Tsc1
+/
hippocampus as
compared to control animals following rapamycin treat-
ment (Fig. 1c; 231 vs. 106 changed proteins, respect-
ively). TSC1 mutations are linked to numerous changes
in biochemical processes, including cell cycle regulation,
translational control and metabolism which are linked to
mTOR pathway hyperactivation. It can be speculated
that rapamycin-related inhibition of the mTORC1 com-
plex results in TSC genotype-dependent adaptations in a
wide range of molecular pathways. These adaptations
could be indirectly involved in the therapeutic effect of
rapamycin. A further explanation for the enhanced rapa-
mycin treatment effect in the Tsc1
+/
mice is selective
vulnerability. Mutant mice might be more susceptible to
the treatment as mTOR hyperactivation modulated simi-
lar downstream molecular pathways during neurodeve-
lopment as are affected by the rapamycin-induced
mTOR hypoactivation.
Wesseling et al. Molecular Autism (2017) 8:41 Page 9 of 12
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Conclusions
Taken together, the results from this comprehensive
study represent the first proteomic characterization of
the Tsc1
+/
mouse model to date. The findings yield
novel insights into the molecular Tsc1
+/
mouse path-
ology as well as the molecular effects of rapamycin treat-
ment, which is an effective treatment for several clinical
symptoms of the tuberous sclerosis complex. Further-
more, the mTOR pathway, which is modulated by rapa-
mycin treatment, is a novel drug target for the treatment
of ASD, schizophrenia and affective disorders. We hope
that the findings from this study will provide evidence
and support for future clinical trials in the field of
neuropsychiatric disorders.
Additional files
Additional file 1: Supplementary methods. Detailed information of the
experimental methods. (DOCX 31 kb)
Additional file 2: Significantly altered proteins identified by label-free
LC-MS
E
analysis in the frontal cortex and hippocampus of the Tsc1
+/
mice compared to Wt mice. Overlapping proteins between hippocampus
and frontal cortex are bold. (XLSX 23 kb)
Additional file 3: Full information for significantly changed proteins
identified by label-based LC-SRM in the frontal cortex and hippocampus
of Tsc1
+/
mice compared to wildtype mice. (DOCX 31 kb)
Abbreviations
ASD: Autism spectrum disorder; LC: Liquid chromatography; MBP: Myelin
basic protein; mTOR: Mammalian target of rapamycin; MYPR: Myelin
proteolipid protein; NSF: Vesicle-fusing ATPase; PKCG: Protein kinase C
gamma; SODC: Superoxide dismutase C; SRM: Selected reaction monitoring;
TSC: Tuberous sclerosis complex; TSC1: Harmartin; TSC2: Tuberin; TSN-
2: Tetraspanin 2; Wt: Wildtype
Acknowledgements
We would like to thank Susan Gooden for the generation and treatment of
the Tsc1
+/-
mouse model and Jadviga Schreiber for proofreading the
manuscript.
Funding
This research was kindly supported by the Stanley Medical Research Institute
(SMRI) and the Dutch Fund for Economic Structure Reinforcement (#0908)
the NeuroBasic PharmaPhenomics project.
Availability of data and materials
The datasets presented in this study can be made available on reasonable
request.
Authorscontributions
HW carried out the label-free LC-MS
E
experiments, designed and carried out
the SRM experiments and performed all statistical and bioinformatic data
analyses. HW prepared the figures and tables and drafted the manuscript. SB
and YE conceived the study and participated in its design and coordination.
SB helped to interpret the results and to write the manuscript. All authors
read, edited and approved the final manuscript.
Ethics approval and consent to participate
All animal experiments were approved by the Dutch Ethical Committee or in
accordance with Institutional Animal Care and Use Committee guidelines.
All animal experiments were approved by the Dutch Animal Experiment
Committee (Dierexperimenten commissie [DEC]) and in accordance with
Dutch animal care and use laws.
Consent for publication
Not applicable.
Competing interests
S.B. is a director of Psynova Neurotech Ltd. and PsyOmics Ltd. The other
authors declare no conflict of interest.
PublishersNote
Springer Nature remains neutral with regard to jurisdictional claims in
published maps and institutional affiliations.
Received: 21 September 2016 Accepted: 14 June 2017
References
1. Islam MP, Roach ES. Tuberous sclerosis complex. Handb Clin Neurol. 2015;
132:97109.
2. de Vries PJ, Prather PA. The tuberous sclerosis complex. N Engl J Med. 2007;
356(1):92. author reply 93-94
3. Joinson C, O'Callaghan FJ, Osborne JP, Martyn C, Harris T, Bolton PF.
Learning disability and epilepsy in an epidemiological sample of individuals
with tuberous sclerosis complex. Psychol Med. 2003;33(2):33544.
4. Roach ES, Smith M, Huttenlocher P, Bhat M, Alcorn D, Hawley L. Diagnostic
criteria: tuberous sclerosis complex. Report of the Diagnostic Criteria
Committee of the National Tuberous Sclerosis Association. J Child Neurol.
1992;7(2):2214.
5. Curatolo P, Moavero R, de Vries PJ. Neurological and neuropsychiatric
aspects of tuberous sclerosis complex. Lancet Neurol. 2015;14(7):73345.
6. Kwiatkowski DJ, Manning BD. Tuberous sclerosis: a GAP at the crossroads of
multiple signaling pathways. Hum Mol Genet. 2005;14 Spec No. 2:R2518.
7. Ehninger D, de Vries PJ, Silva AJ. From mTOR to cognition: molecular and
cellular mechanisms of cognitive impairments in tuberous sclerosis. J
Intellect Disabil Res. 2009;53(10):83851.
8. Klann E, Dever TE. Biochemical mechanisms for translational regulation in
synaptic plasticity. Nat Rev Neurosci. 2004;5(12):93142.
9. Hay N, Sonenberg N. Upstream and downstream of mTOR. Genes Dev.
2004;18(16):192645.
10. Auerbach BD, Osterweil EK, Bear MF. Mutations causing syndromic autism
define an axis of synaptic pathophysiology. Nature. 2011;480(7375):638.
11. Samuels JA. Treatment of renal angiomyolipoma and other hamartomas in
patients with tuberous sclerosis complex. Clin J Am Soc Nephrol. 2017;12(7):
11961202.
12. Hsieh JJ, Purdue MP, Signoretti S, Swanton C, Albiges L, Schmidinger M,
Heng DY, Larkin J, Ficarra V. Renal cell carcinoma. Nat Rev Dis Primers. 2017;
3:17009.
13. Phan AT, Dave B. The pivotal role of mammalian target of rapamycin
inhibition in the treatment of patients with neuroendocrine tumors. Cancer
Med. 2016;5(10):295364.
14. Brown EJ, Albers MW, Shin TB, Ichikawa K, Keith CT, Lane WS, Schreiber SL.
A mammalian protein targeted by G1-arresting rapamycin-receptor
complex. Nature. 1994;369(6483):7568.
15. Chen J, Zheng XF, Brown EJ, Schreiber SL. Identification of an 11-kDa
FKBP12-rapamycin-binding domain within the 289-kDa FKBP12-rapamycin-
associated protein and characterization of a critical serine residue. Proc Natl
Acad Sci U S A. 1995;92(11):494751.
16. Franz DN, Leonard J, Tudor C, Chuck G, Care M, Sethuraman G, Dinopoulos
A, Thomas G, Crone KR. Rapamycin causes regression of astrocytomas in
tuberous sclerosis complex. Ann Neurol. 2006;59(3):4908.
17. Bissler JJ, McCormack FX, Young LR, Elwing JM, Chuck G, Leonard JM,
Schmithorst VJ, Laor T, Brody AS, Bean J, et al. Sirolimus for
angiomyolipoma in tuberous sclerosis complex or
lymphangioleiomyomatosis. N Engl J Med. 2008;358(2):14051.
18. Davies DM, Johnson SR, Tattersfield AE, Kingswood JC, Cox JA, McCartney
DL, Doyle T, Elmslie F, Saggar A, de Vries PJ, et al. Sirolimus therapy in
tuberous sclerosis or sporadic lymphangioleiomyomatosis. N Engl J Med.
2008;358(2):2003.
19. French JA, Lawson JA, Yapici Z, Ikeda H, Polster T, Nabbout R, Curatolo P, de
Vries PJ, Dlugos DJ, Berkowitz N, et al. Adjunctive everolimus therapy for
treatment-resistant focal-onset seizures associated with tuberous sclerosis
(EXIST-3): a phase 3, randomised, double-blind, placebo-controlled study.
Lancet. 2016;388(10056):215363.
Wesseling et al. Molecular Autism (2017) 8:41 Page 10 of 12
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
20. Ehninger D, Li W, Fox K, Stryker MP, Silva AJ. Reversing neurodevelopmental
disorders in adults. Neuron. 2008;60(6):95060.
21. Citraro R, Leo A, Constanti A, Russo E, De Sarro G. mTOR pathway inhibition
as a new therapeutic strategy in epilepsy and epileptogenesis. Pharmacol
Res. 2016;107:33343.
22. Kenerson HL, Aicher LD, True LD, Yeung RS. Activated mammalian target of
rapamycin pathway in the pathogenesis of tuberous sclerosis complex renal
tumors. Cancer Res. 2002;62(20):564550.
23. Kenerson H, Dundon TA, Yeung RS. Effects of rapamycin in the Eker rat
model of tuberous sclerosis complex. Pediatr Res. 2005;57(1):6775.
24. Ehninger D, Han S, Shilyansky C, Zhou Y, Li W, Kwiatkowski DJ, Ramesh V,
Silva AJ. Reversal of learning deficits in a Tsc2+/mouse model of tuberous
sclerosis. Nat Med. 2008;14(8):8438.
25. Zeng LH, Xu L, Gutmann DH, Wong M. Rapamycin prevents epilepsy in a
mouse model of tuberous sclerosis complex. Ann Neurol. 2008;63(4):44453.
26. Meikle L, Pollizzi K, Egnor A, Kramvis I, Lane H, Sahin M, Kwiatkowski DJ.
Response of a neuronal model of tuberous sclerosis to mammalian target
of rapamycin (mTOR) inhibitors: effects on mTORC1 and Akt signaling lead
to improved survival and function. J Neurosci. 2008;28(21):542232.
27. Sato A, Kasai S, Kobayashi T, Takamatsu Y, Hino O, Ikeda K, Mizuguchi M.
Rapamycin reverses impaired social interaction in mouse models of
tuberous sclerosis complex. Nat Commun. 2012;3:1292.
28. Goorden SM, van Woerden GM, van der Weerd L, Cheadle JP, Elgersma Y.
Cognitive deficits in Tsc1+/mice in the absence of cerebral lesions and
seizures. Ann Neurol. 2007;62(6):64855.
29. Wilson C, Idziaszczyk S, Parry L, Guy C, Griffiths DF, Lazda E, Bayne RA, Smith
AJ, Sampson JR, Cheadle JP. A mouse model of tuberous sclerosis 1
showing background specific early post-natal mortality and metastatic renal
cell carcinoma. Hum Mol Genet. 2005;14(13):183950.
30. Wesseling H, Guest PC, Lee CM, Wong EH, Rahmoune H, Bahn S. Integrative
proteomic analysis of the NMDA NR1 knockdown mouse model reveals
effects on central and peripheral pathways associated with schizophrenia
and autism spectrum disorders. Mol Autism. 2014;5:38.
31. Wesseling H, Want EJ, Guest PC, Rahmoune H, Holmes E, Bahn S.
Hippocampal proteomic and metabonomic abnormalities in
neurotransmission, oxidative stress, and apoptotic pathways in a chronic
phencyclidine rat model. J Proteome Res. 2015;14(8):317487.
32. Wesseling H, Rahmoune H, Tricklebank M, Guest PC, Bahn S. A targeted
multiplexed proteomic investigation identifies ketamine-induced changes in
immune markers in rat serum and expression changes in protein kinases/
phosphatases in rat brain. J Proteome Res. 2015;14(1):41121.
33. Wesseling H, Guest PC, Lee CM, Wong EHF, Rahmoune H, Bahn S.
Integrative proteomic analysis of the NMDA NR1 knockdown mouse model
reveals effects on central and peripheral pathways associated with
schizophrenia and autism spectrum disorders. Mol Autism. 2014;5:38.
34. Gottschalk MG, Wesseling H, Guest PC, Bahn S. Proteomic enrichment
analysis of psychotic and affective disorders reveals common signatures in
presynaptic glutamatergic signaling and energy metabolism. Int J
Neuropsychopharmacol. 2014;18(2).
35. Clough T, Thaminy S, Ragg S, Aebersold R, Vitek O. Statistical protein
quantification and significance analysis in label-free LC-MS experiments with
complex designs. BMC Bioinformatics. 2012;13 Suppl 16:S6.
36. Wesseling H, Gottschalk MG, Bahn S. Targeted multiplexed selected reaction
monitoring analysis evaluates protein expression changes of molecular risk
factors for major psychiatric disorders. Int J Neuropsychopharmacol. 2014;18(1).
37. Lange V, Picotti P, Domon B, Aebersold R. Selected reaction monitoring for
quantitative proteomics: a tutorial. Mol Syst Biol. 2008;4:222.
38. Simicevic J, Schmid AW, Gilardoni PA, Zoller B, Raghav SK, Krier I,
Gubelmann C, Lisacek F, Naef F, Moniatte M, et al. Absolute quantification
of transcription factors during cellular differentiation using multiplexed
targeted proteomics. Nat Methods. 2013;10(6):5706.
39. Picotti P, Aebersold R. Selected reaction monitoring-based proteomics:
workflows, potential, pitfalls and future directions. Nat Methods. 2012;9(6):55566.
40. Method of the Year 2012. Nat Methods. 2013;10(1):1.
41. Aebersold R, Burlingame AL, Bradshaw RA. Western blots versus selected
reaction monitoring assays: time to turn the tables? Mol Cell Proteomics.
2013;12(9):23812.
42. Rubin RD, Watson PD, Duff MC, Cohen NJ. The role of the hippocampus in
flexible cognition and social behavior. Front Hum Neurosci. 2014;8:742.
43. Beadle JN, Tranel D, Cohen NJ, Duff MC. Empathy in hippocampal amnesia.
Front Psychol. 2013;4:69.
44. Abrahams BS, Geschwind DH. Advances in autism genetics: on the
threshold of a new neurobiology. Nat Rev Genet. 2008;9(5):34155.
45. Happe F, Ronald A. The 'fractionable autism triad': a review of evidence
from behavioural, genetic, cognitive and neural research. Neuropsychol Rev.
2008;18(4):287304.
46. Silverman JL, Yang M, Lord C, Crawley JN. Behavioural phenotyping assays
for mouse models of autism. Nat Rev Neurosci. 2010;11(7):490502.
47. Iadevaia V, Huo YL, Zhang Z, Foster LJ, Proud CG. Roles of the mammalian
target of rapamycin, mTOR, in controlling ribosome biogenesis and protein
synthesis. Biochem Soc Trans. 2012;40:16872.
48. Tyler WA, Gangoli N, Gokina P, Kim HA, Covey M, Levison SW,
Wood TL. Activation of the mammalian target of rapamycin (mTOR)
is essential for oligode ndrocyte differentiation. J Neurosci. 2009;29(19):
636778.
49. Lebrun-Julien F, Bachmann L, Norrmen C, Trotzmuller M, Kofeler H, Ruegg
MA, Hall MN, Suter U. Balanced mTORC1 activity in oligodendrocytes is
required for accurate CNS myelination. J Neurosci. 2014;34(25):843248.
50. Lourenco T, Paes de Faria J, Bippes CA, Maia J, Lopes-da-Silva JA, Relvas JB,
Graos M. Modulation of oligodendrocyte differentiation and maturation by
combined biochemical and mechanical cues. Sci Rep. 2016;6:21563.
51. Wood TL, Bercury KK, Cifelli SE, Mursch LE, Min J, Dai J, Macklin WB. mTOR: a
link from the extracellular milieu to transcriptional regulation of
oligodendrocyte development. Asn Neuro. 2013;5(1):6379.
52. Krishnan ML, Commowick O, Jeste SS, Weisenfeld N, Hans A, Gregas MC,
Sahin M, Warfield SK. Diffusion features of white matter in tuberous sclerosis
with tractography. Pediatr Neurol. 2010;42(2):1016.
53. Liu J, Dietz K, DeLoyht JM, Pedre X, Kelkar D, Kaur J, Vialou V, Lobo MK,
Dietz DM, Nestler EJ, et al. Impaired adult myelination in the prefrontal
cortex of socially isolated mice. Nat Neurosci. 2012;15(12):1621-+.
54. Tkachev D, Mimmack ML, Huffaker SJ, Ryan M, Bahn S. Further evidence for
altered myelin biosynthesis and glutamatergic dysfunction in schizophrenia.
Int J Neuropsychopharmacol. 2007;10(4):55763.
55. TkachevD,MimmackML,RyanMM,WaylandM,FreemanT,Jones
PB, Starkey M, Webster MJ, Yolken RH, Bahn S. Oligodendrocyte
dysfunction in schizophrenia and bipolar disorder. Lancet. 2003;
362(9386):798805.
56. Zeidan-Chulia F, de Oliveira BN, Casanova MF, Casanova EL, Noda M,
Salmina AB, Verkhratsky A. Up-regulation of oligodendrocyte lineage
markers in the cerebellum of autistic patients: evidence from network
analysis of gene expression. Mol Neurobiol. 2015.
57. Broek JAC, Lin Z, de Gruiter HM, van 't Spijker H, Haasdijk ED, Cox D, Ozcan S, van
Cappellen GWA, Houtsmuller AB, Willemsen R, de Zeeuw CI, Bahn S. Synaptic
vesicle dynamic changes in a model of fragile X. Mol Autism. 2016;7:17.
58. Bloemen OJ, Deeley Q, Sundram F, Daly EM, Barker GJ, Jones DK, van
Amelsvoort TA, Schmitz N, Robertson D, Murphy KC, et al. White matter
integrity in Asperger syndrome: a preliminary diffusion tensor magnetic
resonance imaging study in adults. Autism Res. 2010;3(5):20313.
59. Flores AI, Narayanan SP, Morse EN, Shick HE, Yin X, Kidd G, Avila RL,
Kirschner DA, Macklin WB. Constitutively active Akt induces enhanced
myelination in the CNS. J Neurosci. 2008;28(28):717483.
60. Narayanan SP, Flores AI, Wang F, Macklin WB. Akt signals through the
mammalian target of rapamycin pathway to regulate CNS myelination. J
Neurosci. 2009;29(21):686070.
61. Levi S, Logan SM, Tovar KR, Craig AM. Gephyrin is critical for glycine
receptor clustering but not for the formation of functional GABAergic
synapses in hippocampal neurons. J Neurosci. 2004;24(1):20717.
62. Labonne JD, Graves TD, Shen Y, Jones JR, Kong IK, Layman LC, Kim HG. A
microdeletion at Xq22.2 implicates a glycine receptor GLRA4 involved in
intellectual disability, behavioral problems and craniofacial anomalies. BMC
Neurol. 2016;16:132.
63. Wuerfel E, Bien CG, Vincent A, Woodhall M, Brockmann K. Glycine receptor
antibodies in a boy with focal epilepsy and episodic behavioral disorder. J
Neurol Sci. 2014;343(12):1802.
64. Turner MR, Irani SR, Leite MI, Nithi K, Vincent A, Ansorge O. Progressive
encephalomyelitis with rigidity and myoclonus Glycine and NMDA receptor
antibodies. Neurology. 2011;77(5):43943.
65. Harvey RJ, Yee BK. Glycine transporters as novel therapeutic targets in
schizophrenia, alcohol dependence and pain. Nat Rev Drug Discov. 2013;
12(11):86685.
66. Jacobsson G, Meister B. Molecular components of the exocytotic machinery
in the rat pituitary gland. Endocrinology. 1996;137(12):534456.
Wesseling et al. Molecular Autism (2017) 8:41 Page 11 of 12
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
67. Whiteheart SW, Rossnagel K, Buhrow SA, Brunner M, Jaenicke R, Rothman
JE. N-ethylmaleimide-sensitive fusion protein: a trimeric ATPase whose
hydrolysis of ATP is required for membrane fusion. J Cell Biol.
1994;126(4):94554.
68. Yan Q, Sun W, McNew JA, Vida TA, Bean AJ. Ca2+ and N-ethylmaleimide-
sensitive factor differentially regulate disassembly of SNARE complexes on
early endosomes. J Biol Chem. 2004;279(18):182706.
69. Fernandez-Castillo N, Cormand B, Roncero C, Sanchez-Mora C, Grau-Lopez L,
Gonzalvo B, Miquel L, Corominas R, Ramos-Quiroga JA, Casas M, et al.
Candidate pathway association study in cocaine dependence: the control of
neurotransmitter release. World J Biol Psychiatry. 2012;13(2):12634.
70. Imai C, Sugai T, Iritani S, Niizato K, Nakamura R, Makifuchi T, Kakita A,
Takahashi H, Nawa H. A quantitative study on the expression of synapsin II
and N-ethylmaleimide-sensitive fusion protein in schizophrenic patients.
Neurosci Lett. 2001;305(3):1858.
71. Nishimune A, Isaac JT, Molnar E, Noel J, Nash SR, Tagaya M, Collingridge GL,
Nakanishi S, Henley JM. NSF binding to GluR2 regulates synaptic
transmission. Neuron. 1998;21(1):8797.
72. Osten P, Srivastava S, Inman GJ, Vilim FS, Khatri L, Lee LM, States BA,
Einheber S, Milner TA, Hanson PI, et al. The AMPA receptor GluR2 C
terminus can mediate a reversible, ATP-dependent interaction with NSF and
alpha- and beta-SNAPs. Neuron. 1998;21(1):99110.
73. Cong M, Perry SJ, Hu LA, Hanson PI, Claing A, Lefkowitz RJ. Binding of the
beta2 adrenergic receptor to N-ethylmaleimide-sensitive factor regulates
receptor recycling. J Biol Chem. 2001;276(48):4514552.
74. Heydorn A, Sondergaard BP, Hadrup N, Holst B, Haft CR, Schwartz TW.
Distinct in vitro interaction pattern of dopamine receptor subtypes with
adaptor proteins involved in post-endocytotic receptor targeting. FEBS Lett.
2004;556(13):27680.
75. Bomberger JM, Parameswaran N, Hall CS, Aiyar N, Spielman WS. Novel
function for receptor activity-modifying proteins (RAMPs) in post-endocytic
receptor trafficking. J Biol Chem. 2005;280(10):9297307.
76. Pontier SM, Lahaie N, Ginham R, St-Gelais F, Bonin H, Bell DJ, Flynn H, Trudeau
LE, McIlhinney J, White JH, et al. Coordinated action of NSF and PKC regulates
GABAB receptor signaling efficacy. EMBO J. 2006;25(12):2698709.
77. Zheng Z, Keifer J. Protein kinase C-dependent and independent signaling
pathways regulate synaptic GluR1 and GluR4 AMPAR subunits during in
vitro classical conditioning. Neuroscience. 2008;156(4):87284.
78. Gomes AR, Correia SS, Esteban JA, Duarte CB, Carvalho AL. PKC anchoring
to GluR4 AMPA receptor subunit modulates PKC-driven receptor
phosphorylation and surface expression. Traffic. 2007;8(3):25969.
79. Silverman JL, Smith DG, Rizzo SJ, Karras MN, Turner SM, Tolu SS, Bryce DK,
Smith DL, Fonseca K, Ring RH, et al. Negative allosteric modulation of the
mGluR5 receptor reduces repetitive behaviors and rescues social deficits in
mouse models of autism. Sci Transl Med. 2012;4(131):131ra151.
80. Berry-Kravis E, Des Portes V, Hagerman R, Jacquemont S, Charles P, Visootsak
J, Brinkman M, Rerat K, Koumaras B, Zhu L, et al. Mavoglurant in fragile X
syndrome: results of two randomized, double-blind, placebo-controlled
trials. Sci Transl Med. 2016;8(321):321ra325.
81. Jacquemont S, Berry-Kravis E, Hagerman R, von Raison F, Gasparini F, Apostol
G, Ufer M, Des Portes V, Gomez-Mancilla B. The challenges of clinical trials in
fragile X syndrome. Psychopharmacology. 2014;231(6):123750.
82. Li NX, Lee B, Liu RJ, Banasr M, Dwyer JM, Iwata M, Li XY, Aghajanian G, Duman
RS. mTOR-dependent synapse formation underlies the rapid antidepressant
effects of NMDA antagonists. Science. 2010;329(5994):95964.
83. Huang LN, Liu Y, Jin W, Ji XC, Dong ZM. Ketamine potentiates hippocampal
neurodegeneration and persistent learning and memory impairment
through the PKC gamma-ERK signaling pathway in the developing brain.
Brain Res. 2012;1476:16471.
84. Shapira M, Kakiashvili E, Rosenberg T, Hershko DD. The mTOR inhibitor
rapamycin down-regulates the expression of the ubiquitin ligase subunit
Skp2 in breast cancer cells. Breast Cancer Res. 2006;8(4):R46.
85. Reinhard JR, Kriz A, Galic M, Angliker N, Rajalu M, Vogt KE, Ruegg MA. The
calcium sensor Copine-6 regulates spine structural plasticity and learning
and memory. Nat Commun. 2016;7:11613.
86. Suetsugu M, Mehraein P. Spine distribution along the apical dendrites of
the pyramidal neurons in Down's syndrome. A quantitative Golgi study.
Acta Neuropathol. 1980;50(3):20710.
87. Chang KT, Ro H, Wang W, Min KT. Meeting at the crossroads: common
mechanisms in Fragile X and Down syndrome. Trends Neurosci.
2013;36(12):68594.
88. Kaufmann WE, Moser HW. Dendritic anomalies in disorders associated with
mental retardation. Cereb Cortex. 2000;10(10):98191.
89. Swann JW, Al-Noori S, Jiang M, Lee CL. Spine loss and other dendritic
abnormalities in epilepsy. Hippocampus. 2000;10(5):61725.
90. Chang CY, Picotti P, Huttenhain R, Heinzelmann-Schwarz V, Jovanovic M,
Aebersold R, Vitek O. Protein significance analysis in selected reaction monitoring
(SRM) measurements. Mol Cell Proteomics. 2011;11(4):M111 014662.
91. Benjamini Y, Hochberg Y. Controlling the false discovery ratea practical
and powerful approach to multiple testing. J Royal Stat Soc Ser B Methodol.
1995;57(1):289300.
We accept pre-submission inquiries
Our selector tool helps you to find the most relevant journal
We provide round the clock customer support
Convenient online submission
Thorough peer review
Inclusion in PubMed and all major indexing services
Maximum visibility for your research
Submit your manuscript at
www.biomedcentral.com/submit
Submit your next manuscript to BioMed Central
and we will help you at every step:
Wesseling et al. Molecular Autism (2017) 8:41 Page 12 of 12
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... In the last years, proteomic studies have been contributing to elucidate some molecular aspects of CNS lesions and neurological manifestations of TSC. In particular, preliminary proteomic characterizations of some different brain areas of human patients with TSC1 mutations [27] and of Tsc1-knockout mice have been performed [28,29]. Although the promising results evidenced protein alterations and cellular pathway dysregulation linked to TSC clinical manifestations, further studies are needed to define the effects of mTOR hyperactivation and their implications in the disease onset. ...
... GO analysis of differentially represented proteins in Tsc1 ko cells also highlighted that further structural changes probably occur in the "myelin sheath", as already observed in the frontal cortex and hippocampus of Tsc1-deficient mice [28]. Interestingly, Tsc1 ko cells showed a down-representation of fatty acid-binding protein 7 (FABP7), which is one of the factors affecting disorganized axonal myelinogenesis, a wellknown feature of TSC neurological disorders. ...
Article
Full-text available
Tuberous sclerosis complex (TSC) is a rare, multisystem genetic disorder that leads to the development of benign tumors in multiple organs and neurological symptoms. TSC clinical manifestations show a great heterogenicity, with most patients presenting severe neuropsychiatric and neurological disorders. TSC is caused by loss-of-function mutations in either Tsc1 or Tsc2 genes, leading to overexpression of the mechanistic target of rapamycin (mTOR) and, consequently, abnormal cellular growth, proliferation and differentiation as well as to cell migration defects. Beside the growing interest, TSC remains a disorder poorly understood, with limited perspectives in the field of therapeutic strategies. Here we used murine postnatal subventricular zone (SVZ) neural stem progenitor cells (NSPCs) deficient of Tsc1 gene as a TSC model to unravel novel molecular aspects of the pathophysiology of this disease. 2D-DIGE-based proteomic analysis detected 55 differently represented spots in Tsc1-deficient cells, compared to wild-type counterparts, which were associated with 36 protein entries after corresponding trypsinolysis and nanoLC-ESI-Q-Orbitrap-MS/MS analysis. Proteomic results were validated using various experimental approaches. Bioinformatics associated differently represented proteins with oxidative stress and redox pathways, methylglyoxal biosynthesis, myelin sheath, protein S-nitrosylation and carbohydrate metabolism. Because most of these cellular pathways have already been linked to TSC features, these results were useful to clarify some molecular aspects of TSC etiopathogenesis and suggested novel promising therapeutic protein targets. SIGNIFICANCE: Tuberous Sclerosis Complex (TSC) is a multisystemic disorder caused by inactivating mutations of TSC1 or TSC2 genes, which induce overactivation of the mTOR component. The molecular mechanisms underlying the pathogenesis of TSC remain unclear, probably due to complexity of mTOR signaling network. To have a picture of protein abundance changes occurring in TSC disorder, murine postnatal subventricular zone (SVZ) neural stem progenitor cells (NSPCs) deficient of Tsc1 gene were used as a model of disease. Thus, Tsc1-deficient SVZ NSPCs and wild-type cells were comparatively evaluated by proteomics. This analysis evidenced changes in the abundance of proteins involved in oxidative/nitrosative stress, cytoskeleton remodelling, neurotransmission, neurogenesis and carbohydrate metabolism. These proteins might clarify novel molecular aspects of TSC etiopathogenesis and constitute putative molecular targets for novel therapeutic management of TSC-related disorders.
... The most common examples of genetically induced rodent models of autism are BTBR T + tf/J (BTBR), which exhibit behavioral deficits with face validity to the main symptoms observed in individuals with ASD (Zhao et al., 2022); Fmr1 KO (knockout) mouse model of Fragile X Chromosome syndrome (FXS), which presents with of autism and intellectual disability (Kazdoba et al., 2014); mice mutant for neuro-ligin3/4 (Nlgn3 and Nlgn4), which in humans are strongly implicated in development of neuropsychiatric disorders such as autism (Tabuchi et al., 2007); mutant mice for the Shank gene family and Tcs1/Tsc2 genes that show deregulation of proteins linked to synaptic plasticity, which can result in autistic-like traits (Jiang and Ehlers, 2013;Wesseling et al., 2017). ...
Article
Environmental enrichment (EE) is known to produce experience-dependent changes in the brains and behaviors of rodents, and it has therefore been widely used to study neurodevelopmental disorders, including autism. Current studies show significant protocol variation, such as the presence of running wheels, number of cagemates, duration of enrichment, and the age of the animals at the beginning and end of the enrichment interventions. EE has been shown to have prominent positive effects in animal models of idiopathic and syndromic autism, but little is known about the ideal type of EE and the most efficient protocols for reversing autism spectrum disorder (ASD) behaviors modeled in rodents. This review presents evidence that social enrichment is the most effective way to rescue typical behaviors, and that variables such as onset, duration, and type of induction in the ASD model are important for EE success. Understanding which EE protocols are most beneficial for reversing ASD behaviors modeled in rodents opens up possibilities for the potential treatment of neuropsychiatric disorders characterized by behavioral deficits, such as autism.
... [65][66][67][68] There are also some reports of improvement in multiple aspects of social deficit behavior and neurocognition with everolimus treatment, but adequate studies to address these issues are needed. 65,[69][70][71] Lung Lymphangioleiomyomatosis (LAM) is the replacement of alveolar tissue by cysts and proliferation of the smooth muscle, progressively affecting the pulmonary function, and affecting 40% of the female TSC patients. It is more frequently associated with TSC2 pathogenic variants and predominantly affects premenopausal women, in the third to fourth decade of life, 1,2,28,39 leading to the hypothesis that estrogen regulates TSC signaling, and perhaps also the migration of TSC2deficient cells. ...
Article
Full-text available
Inês Gomes,1 Joana Jesus Ribeiro,2 Filipe Palavra3– 5 1Neurology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal; 2Neurology Department, Centro Hospitalar de Leiria, Leiria, Portugal; 3Center for Child Development – Neuropediatrics Unit, Hospital Pediátrico, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal; 4Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal; 5Laboratory of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Coimbra, PortugalCorrespondence: Filipe Palavra, Center for Child Development – Neuropediatrics Unit, Hospital Pediátrico, Avenida Afonso Romão, Coimbra, 3000-602, Portugal, Email fpalavra@fmed.uc.ptAbstract: Tuberous sclerosis complex (TSC) is a rare genetic disease of autosomal dominant transmission that, in most cases, results from the presence of pathogenic variants of the TSC1 or TSC2 genes, encoding hamartin and tuberin, respectively. It is a multisystemic disease, affecting most frequently the brain, skin, kidney, and heart. The wide variety of possible clinical manifestations, given this multisystem dimension, makes the follow-up of patients with TSC an exercise of multidisciplinarity. In fact, these patients may require the intervention of various medical specialties, which thus have to combine their efforts to practice a medicine that is truly holistic. The past few years have witnessed a dramatic leap not only in the diagnosis and management of TSC patients, with standard monitoring recommendations, but also in the therapeutic field, with the use of mTORC1 inhibitors. In this article, we review the clinical manifestations associated with TSC, as well as the treatment and follow-up strategies that should be implemented, from a multidisciplinary perspective.Keywords: tuberous sclerosis complex, multidisciplinary communications, care management, patient
... Therefore deletion, mutation or rearrangement of the Tsc genes leads to a loss of activity of the complex and overactivation of the mTORC1 pathway (Huang and Manning, 2008;Portocarrero et al., 2018). As expected, proteomic analyses show increases of mTOR in both the hippocampus and striatum of Tsc1 +/− mice (Wesseling et al., 2017) and increases of downstream targets such as Ulk1 mRNA and phospho-S6K protein in the Tsc2 +/− mouse brain (Sato et al., 2012). This shows that mTOR overactivation by the deletion of an ASC-associated gene can increase the expression of downstream targets of mTORC1 (Figure 2). ...
Article
Full-text available
Autism Spectrum Conditions (ASC) are a group of neurodevelopmental disorders characterized by deficits in social communication and interaction as well as repetitive behaviors and restricted range of interests. ASC are complex genetic disorders with moderate to high heritability, and associated with atypical patterns of neural connectivity. Many of the genes implicated in ASC are involved in dendritic spine pruning and spine development, both of which can be mediated by the mammalian target of rapamycin (mTOR) signaling pathway. Consistent with this idea, human postmortem studies have shown increased spine density in ASC compared to controls suggesting that the balance between autophagy and spinogenesis is altered in ASC. However, murine models of ASC have shown inconsistent results for spine morphology, which may underlie functional connectivity. This review seeks to establish the relevance of changes in dendritic spines in ASC using data gathered from rodent models. Using a literature survey, we identify 20 genes that are linked to dendritic spine pruning or development in rodents that are also strongly implicated in ASC in humans. Furthermore, we show that all 20 genes are linked to the mTOR pathway and propose that the mTOR pathway regulating spine dynamics is a potential mechanism underlying the ASC signaling pathway in ASC. We show here that the direction of change in spine density was mostly correlated to the upstream positive or negative regulation of the mTOR pathway and most rodent models of mutant mTOR regulators show increases in immature spines, based on morphological analyses. We further explore the idea that these mutations in these genes result in aberrant social behavior in rodent models that is due to these altered spine dynamics. This review should therefore pave the way for further research on the specific genes outlined, their effect on spine morphology or density with an emphasis on understanding the functional role of these changes in ASC.
... Examination of common downstream mTORC1 effector pathways noted to be altered in previous transcriptomic and/or proteomic profiling studies in surgically resected human cortical tubers or cortical samples taken from TSC animal models has identified a few prominent pathways impacted by mTORC1 hyperactivation, including metabolic dysregulation compromising amino acid metabolism, lipid biosynthesis, neuronal morphogenesis, the actin cytoskeleton and growth-related signals (6)(7)(8)(9). Interestingly, several of these pathways are known to be major upstream regulators of the evolutionarily conserved Hippo-Yap signaling pathway, including metabolic cues and cytoskeletal changes (due to mechanical tension), actin dynamics and extracellular signaling (6,7,9). In line with this, recent evidence suggests that Yes-associated protein (Yap) signaling is an mTORC1 target whose activity is promoted through an inhibition of Yap autophagy (10)(11)(12). ...
Article
The multi-systemic genetic disorder tuberous sclerosis complex (TSC) impacts multiple neurodevelopmental processes including neuronal morphogenesis, neuronal migration, myelination, and gliogenesis. These alterations contribute to the development of cerebral cortex abnormalities and malformations. Although TSC is caused by mTORC1 hyperactivation, cognitive and behavioral impairments are not improved through mTORC1 targeting, making the study of the downstream effectors of this complex important for understanding the mechanisms underlying TSC. As mTORC1 has been shown to promote the activity of the transcriptional co-activator Yap, we hypothesized that altered Yap/Taz signaling contributes to the pathogenesis of TSC. We first observed that the level of Yap/Taz are increased in a human cortical tuber sample and in embryonic cortices of Tsc2 conditional knockout (cKO) mice. Next, to determine how abnormal upregulation of Yap/Taz impacts the neuropathology of TSC, we deleted Yap/Taz in Tsc2 cKO mice. Importantly, Yap/Taz/Tsc2 tcKO animals show reduced cortical thickness and cortical neuron cell size, despite the persistence of high mTORC1 activity, suggesting that Yap/Taz play a downstream role in cytomegaly. Furthermore, Yap/Taz/Tsc2 tcKO significantly restored cortical and hippocampal lamination defects and reduced hippocampal heterotopia formation. Finally, the loss of Yap/Taz increased the distribution of myelin basic protein in Tsc2 cKO animals, consistent with an improvement in myelination. Overall, our results indicate that targeting Yap/Taz lessens the severity of neuropathology in a TSC animal model. This study is the first to implicate Yap/Taz as contributors to cortical pathogenesis in TSC and therefore as potential novel targets in the treatment of this disorder.
... Consistent with our functional cluster analysis in the mTOR activationsensitive genes in C1483Y cells, mTOR activation-sensitive genes in FMCD were also enriched in mitochondrial function and energy production ( Figure 3C and 4I). Consistent with our findings, other studies have provided evidence suggesting that activation of the mTOR pathway leads to increased translation of a subset of mitochondrial and energy production-related mRNAs (62,63) and that 5′ TOP genes are insensitive to the activation of mTOR (64,65). Taken together, our data indicated that translational control and functions mediated by somatic activating mutations in MTOR are distinct from those mediated by pharmacological inhibition of mTOR. ...
Thesis
Full-text available
Brain somatic mutations confer genomic diversity in the human brain and cause neurodevelopmental disorders. Dysregulation of eukaryotic initiation factor 4E (eIF4E), which is a core translational regulator, is a common molecular hallmark in various neurological disorders. Individuals carrying inherited mutations in the mechanistic target of rapamycin (mTOR) or Ras/mitogen-activated protein kinase (MAPK) pathway, upstream activators of eIF4E, have abnormalities in translation and are prone to developing epilepsy, megalencephaly, autism spectrum disorder (ASD), brain tumor, and neuropshychiatric disorder. It has been hypothesized that brain somatic mutation that activates eIF4E is a potential culprit of neurological disorders. Here we report for the first time a mouse model with behavioral abnormalities including anxiety, defective sociability, deficits in repetitive behavior, and anhedonia caused by brain somatic mutation in MTOR. Recently, brain somatic activating mutations in MTOR have been identified as a major etiology of intractable epilepsy in patients with cortical malformations. However, the molecular genetic mechanism of how brain somatic mutations in MTOR cause intractable epilepsy has remained elusive. In this study, translational profiling of intractable epilepsy mouse models with brain somatic mutations and genome-edited cells revealed a novel translational dysregulation mechanism and mTOR activation–sensitive targets mediated by human MTOR mutations that lead to intractable epilepsy with cortical malformation. These mTOR targets were found to be regulated by novel mTOR-responsive 5′-UTR motifs, distinct from known mTOR inhibition–sensitive targets regulated by 5′ terminal oligopyrimidine motifs. Novel mTOR target genes were validated in patient brain tissues, and the mTOR downstream effector eIF4E was identified as a new therapeutic target in intractable epilepsy via pharmacological or genetic inhibition. We show that metformin, an FDA-approved eIF4E inhibitor, suppresses intractable epilepsy. However, there is no clinically relevant drug which specifically inhibits eIF4E in the brain. We found that antisense oligonucleotides (ASO) directed at eIF4E in the brain downregulated eIF4E and reversed the behavioral, neurophysiological, and bio-chemical abnormalities in mouse model with brain somatic mutation in MTOR. Altogether, the present study describes behavioral, neurophysiological, and biochemical abnormalities resulting from eIF4E dysregulation by mTOR pathway activation, and its therapeutic reduction could represent a promising and broadly effective translational therapy for neurological disorders where eIF4E is dysregulated. Altogether, the present study describes translational dysregulation resulting from brain somatic mutations in MTOR, as well as the pathogenesis and potential therapeutic targets of intractable epilepsy.
... As both mouse mutations are homozygously lethal, heterozygously mutating Rheb or Tsc1 has been shown to lower protein expression, and respectively suppress or enhance mTORC1 activity by one third (Goorden et al., 2011;Wesseling et al., 2017). Both these mouse models, which were studied previously in light of neuronal development but never life span, were crossed with Ercc1 ∆/− mice in the same F1 C57BL6J/FVB hybrid background. ...
Article
Full-text available
Dietary restriction (DR) and rapamycin extend healthspan and life span across multiple species. We have recently shown that DR in progeroid DNA repair‐deficient mice dramatically extended healthspan and trippled life span. Here, we show that rapamycin, while significantly lowering mTOR signaling, failed to improve life span nor healthspan of DNA repair‐deficient Ercc1∆/− mice, contrary to DR tested in parallel. Rapamycin interventions focusing on dosage, gender, and timing all were unable to alter life span. Even genetically modifying mTOR signaling failed to increase life span of DNA repair‐deficient mice. The absence of effects by rapamycin on P53 in brain and transcription stress in liver is in sharp contrast with results obtained by DR, and appoints reducing DNA damage and transcription stress as an important mode of action of DR, lacking by rapamycin. Together, this indicates that mTOR inhibition does not mediate the beneficial effects of DR in progeroid mice, revealing that DR and rapamycin strongly differ in their modes of action. In this study, we have subjected Ercc1Δ/− progeroid DNA repair‐deficient mice to rapamycin and dietary restriction (DR), two well‐known interventions for extending life span across multiple species. DR again resulted in extreme benefits for these mice while rapamycin interventions, focusing on dosage, gender, and timing as well as modulation of mTor via genetic means, all failed to improve life span, healthspan, or the DNA damage indicator transcription stress.
Article
Autism spectrum disorder (ASD) has become a common neurodevelopmental disorder. The heterogeneity of ASD poses great challenges for its research and clinical translation. On the basis of reviewing the heterogeneity of ASD, this review systematically summarized the current status and progress of pathogenesis, diagnostic markers, and interventions for ASD. We provided an overview of the ASD molecular mechanisms identified by multi‐omics studies and convergent mechanism in different genetic backgrounds. The comorbidities, mechanisms associated with important physiological and metabolic abnormalities (i.e., inflammation, immunity, oxidative stress, and mitochondrial dysfunction), and gut microbial disorder in ASD were reviewed. The non‐targeted omics and targeting studies of diagnostic markers for ASD were also reviewed. Moreover, we summarized the progress and methods of behavioral and educational interventions, intervention methods related to technological devices, and research on medical interventions and potential drug targets. This review highlighted the application of high‐throughput omics methods in ASD research and emphasized the importance of seeking homogeneity from heterogeneity and exploring the convergence of disease mechanisms, biomarkers, and intervention approaches, and proposes that taking into account individuality and commonality may be the key to achieve accurate diagnosis and treatment of ASD.
Article
Caloric Restriction (CR) is an innovative therapy used in tumor tissue and tumor model studies to promote cell death and decrease cell viability. Caloric restriction mimetics (CRM) are a class of drugs that induce CR and starvation conditions within a cell. When used simultaneously with other chemotherapy agents, the effects are synergistic and effective at promoting tumor cell death. In this review, we discuss CRMs and their potential as cancer therapeutics. Firstly, we establish an overview of CR and its impacts on healthy and tumor cells. CR and CRM drugs have shown to decrease age‐related diseases and can act as an anti‐cancer agent. As it can be challenging for an individual to diligently stick to a diet that would induce CR, CRMs are even more desirable. Then we discuss the drug class by highlighting three CRMs; resveratrol, (‐)‐hydroxycitric acid and rapamycin. These CRMs are commonly known for their dietary effects, but the underlying mechanisms that drive cellular metabolic and proteomic changes show promise as a cancer therapeutic. Lastly, we highlight the use of mass spectrometry and proteomic techniques on experiments utilizing CRM drugs to understand the cellular pathways impacted by this drug class, leading to a better understanding of the anti‐cancer properties and potentials of caloric restriction mimetics. This article is protected by copyright. All rights reserved
Article
Full-text available
Renal cell carcinoma (RCC) denotes cancer originated from the renal epithelium and accounts for >90% of cancers in the kidney. The disease encompasses >10 histological and molecular subtypes, of which clear cell RCC (ccRCC) is most common and accounts for most cancer-related deaths. Although somatic VHL mutations have been described for some time, more-recent cancer genomic studies have identified mutations in epigenetic regulatory genes and demonstrated marked intra-tumour heterogeneity, which could have prognostic, predictive and therapeutic relevance. Localized RCC can be successfully managed with surgery, whereas metastatic RCC is refractory to conventional chemotherapy. However, over the past decade, marked advances in the treatment of metastatic RCC have been made, with targeted agents including sorafenib, sunitinib, bevacizumab, pazopanib and axitinib, which inhibit vascular endothelial growth factor (VEGF) and its receptor (VEGFR), and everolimus and temsirolimus, which inhibit mechanistic target of rapamycin complex 1 (mTORC1), being approved. Since 2015, agents with additional targets aside from VEGFR have been approved, such as cabozantinib and lenvatinib; immunotherapies, such as nivolumab, have also been added to the armamentarium for metastatic RCC. Here, we provide an overview of the biology of RCC, with a focus on ccRCC, as well as updates to complement the current clinical guidelines and an outline of potential future directions for RCC research and therapy.
Article
Full-text available
Background: Everolimus, a mammalian target of rapamycin (mTOR) inhibitor, has been used for various benign tumours associated with tuberous sclerosis complex. We assessed the efficacy and safety of two trough exposure concentrations of everolimus, 3-7 ng/mL (low exposure) and 9-15 ng/mL (high exposure), compared with placebo as adjunctive therapy for treatment-resistant focal-onset seizures in tuberous sclerosis complex. Methods: In this phase 3, randomised, double-blind, placebo-controlled study, eligible patients aged 2-65 years with tuberous sclerosis complex and treatment-resistant seizures (≥16 in an 8-week baseline phase) receiving one to three concomitant antiepileptic drugs were recruited from 99 centres across 25 countries. Participants were randomly assigned (1:1:1), via permuted-block randomisation (block size of six) implemented by Interactive Response Technology software, to receive placebo, low-exposure everolimus, or high-exposure everolimus. Randomisation was stratified by age subgroup (<6 years, 6 to <12 years, 12 to <18 years, and ≥18 years). Patients, investigators, site personnel, and the sponsor's study team were masked to treatment allocation. The starting dose of everolimus depended on age, body-surface area, and concomitant use of cytochrome 3A4/P-glycoprotein inducers. Dose adjustments were done to attain target trough ranges during a 6-week titration period, and as needed during a 12-week maintenance period of core phase. Patients or their caregivers recorded events in a seizure diary throughout the study. The primary endpoint was change from baseline in the frequency of seizures during the maintenance period, defined as response rate (the proportion of patients achieving ≥50% reduction in seizure frequency) and median percentage reduction in seizure frequency, in all randomised patients. This study is registered with ClinicalTrials.gov, number NCT01713946. Findings: Between July 3, 2013, and May 29, 2015, 366 patients were enrolled and randomly assigned to placebo (n=119), low-exposure everolimus, (n=117), or high-exposure everolimus (n=130). The response rate was 15·1% with placebo (95% CI 9·2-22·8; 18 patients) compared with 28·2% for low-exposure everolimus (95% CI 20·3-37·3; 33 patients; p=0·0077) and 40·0% for high-exposure everolimus (95% CI 31·5-49·0; 52 patients; p<0·0001). The median percentage reduction in seizure frequency was 14·9% (95% CI 0·1-21·7) with placebo versus 29·3% with low-exposure everolimus (95% CI 18·8-41·9; p=0·0028) and 39·6% with high-exposure everolimus (95% CI 35·0-48·7; p<0·0001). Grade 3 or 4 adverse events occurred in 13 (11%) patients in the placebo group, 21 (18%) in the low-exposure group, and 31 (24%) in the high-exposure group. Serious adverse events were reported in three (3%) patients who received placebo, 16 (14%) who received low-exposure everolimus, and 18 (14%) who received high-exposure everolimus. Adverse events led to treatment discontinuation in two (2%) patients in the placebo group versus six (5%) in the low-exposure group and four (3%) in the high-exposure group. Interpretation: Adjunctive everolimus treatment significantly reduced seizure frequency with a tolerable safety profile compared with placebo in patients with tuberous sclerosis complex and treatment-resistant seizures. Funding: Novartis Pharmaceuticals Corporation.
Article
Full-text available
Significant advances have been made toward understanding the biology of neuroendocrine tumors (NET) in terms of defining prognosis and improving clinical management; however, many unmet needs remain. The treatment landscape for NET has evolved, with the approval of the targeted agents everolimus and sunitinib for the treatment of advanced pancreatic NET in 2011 followed by the approval of everolimus for the treatment of advanced nonfunctional gastrointestinal and lung NET in 2016. Mammalian target of rapamycin (mTOR) and components of the mTOR pathway play pivotal roles in NET pathogenesis. Effects of the mTOR inhibitor everolimus have been well documented in preclinical and clinical studies, both as monotherapy and combination therapy. mTOR inhibition as backbone therapy within the NET treatment landscape is a focus of continuing research, which includes evaluation of the growing armamentarium of approved and investigational agents as potential combination partners. Data evaluating the clinical benefits of agents targeting mTOR and related pathways (alone and in combination) in the treatment of patients with NET continue to increase. Many of the findings to date are encouraging.
Article
Full-text available
Background Among the 21 annotated genes at Xq22.2, PLP1 is the only known gene involved in Xq22.2 microdeletion and microduplication syndromes with intellectual disability. Using an atypical microdeletion, which does not encompass PLP1, we implicate a novel gene GLRA4 involved in intellectual disability, behavioral problems and craniofacial anomalies. Case presentationWe report a female patient (DGDP084) with a de novo Xq22.2 microdeletion of at least 110 kb presenting with intellectual disability, motor delay, behavioral problems and craniofacial anomalies. While her phenotypic features such as cognitive impairment and motor delay show overlap with Pelizaeus-Merzbacher disease (PMD) caused by PLP1 mutations at Xq22.2, this gene is not included in our patient’s microdeletion and is not dysregulated by a position effect. Because the microdeletion encompasses only three genes, GLRA4, MORF4L2 and TCEAL1, we investigated their expression levels in various tissues by RT-qPCR and found that all three genes were highly expressed in whole human brain, fetal brain, cerebellum and hippocampus. When we examined the transcript levels of GLRA4, MORF4L2 as well as TCEAL1 in DGDP084′s family, however, only GLRA4 transcripts were reduced in the female patient compared to her healthy mother. This suggests that GLRA4 is the plausible candidate gene for cognitive impairment, behavioral problems and craniofacial anomalies observed in DGDP084. Importantly, glycine receptors mediate inhibitory synaptic transmission in the brain stem as well as the spinal cord, and are known to be involved in syndromic intellectual disability. Conclusion We hypothesize that GLRA4 is involved in intellectual disability, behavioral problems and craniofacial anomalies as the second gene identified for X-linked syndromic intellectual disability at Xq22.2. Additional point mutations or intragenic deletions of GLRA4 as well as functional studies are needed to further validate our hypothesis.
Article
Full-text available
Hippocampal long-term potentiation (LTP) represents the cellular response of excitatory synapses to specific patterns of high neuronal activity and is required for learning and memory. Here we identify a mechanism that requires the calcium-binding protein Copine-6 to translate the initial calcium signals into changes in spine structure. We show that Copine-6 is recruited from the cytosol of dendrites to postsynaptic spine membranes by calcium transients that precede LTP. Cpne6 knockout mice are deficient in hippocampal LTP, learning and memory. Hippocampal neurons from Cpne6 knockouts lack spine structural plasticity as do wild-type neurons that express a Copine-6 calcium mutant. The function of Copine-6 is based on its binding, activating and recruiting the Rho GTPase Rac1 to cell membranes. Consistent with this function, the LTP deficit of Cpne6 knockout mice is rescued by the actin stabilizer jasplakinolide. These data show that Copine-6 links activity-triggered calcium signals to spine structural plasticity necessary for learning and memory.
Article
Full-text available
Background: Fragile X syndrome (FXS) is a single-gene disorder that is the most common heritable cause of intellectual disability and the most frequent monogenic cause of autism spectrum disorders (ASD). FXS is caused by an expansion of trinucleotide repeats in the promoter region of the fragile X mental retardation gene (Fmr1). This leads to a lack of fragile X mental retardation protein (FMRP), which regulates translation of a wide range of messenger RNAs (mRNAs). The extent of expression level alterations of synaptic proteins affected by FMRP loss and their consequences on synaptic dynamics in FXS has not been fully investigated. Methods: Here, we used an Fmr1 knockout (KO) mouse model to investigate the molecular mechanisms underlying FXS by monitoring protein expression changes using shotgun label-free liquid-chromatography mass spectrometry (LC-MS(E)) in brain tissue and synaptosome fractions. FXS-associated candidate proteins were validated using selected reaction monitoring (SRM) in synaptosome fractions for targeted protein quantification. Furthermore, functional alterations in synaptic release and dynamics were evaluated using live-cell imaging, and interpretation of synaptic dynamics differences was investigated using electron microscopy. Results: Key findings relate to altered levels of proteins involved in GABA-signalling, especially in the cerebellum. Further exploration using microscopy studies found reduced synaptic vesicle unloading of hippocampal neurons and increased vesicle unloading in cerebellar neurons, which suggests a general decrease of synaptic transmission. Conclusions: Our findings suggest that FMRP is a regulator of synaptic vesicle dynamics, which supports the role of FMRP in presynaptic functions. Taken together, these studies provide novel insights into the molecular changes associated with FXS.
Article
Tuberous sclerosis complex (TSC) is a neurocutaneous syndrome that can affect the brain, skin, eyes, kidneys, heart, and lungs. TSC alters cellular proliferation and differentiation, resulting in hamartomas of various organs, tumor formation, and altered neuronal migration. The phenotype is highly variable. Most individuals have seizures, commonly including infantile spasms, and there is variable intellectual disability and autism. Neonates can present with cardiac failure due to intracardiac rhabdomyomas. The likelihood of renal angiomyolipomas increases with age, and renal disease is the most common cause of death in adults with TSC. Pulmonary involvement occurs predominantly in women and carries a high morbidity and mortality. TSC is inherited as an autosomal dominant trait, but spontaneous mutations are common. A mutation of either TSC1 on chromosome 9 or TSC2 on chromosome 16 leads to dysfunction of hamartin or tuberin, respectively. These two proteins form a functional complex that modulates the mammalian target of rapamycin (mTOR) pathway. Medications that inhibit mTOR are being used to treat TSC-related tumors, and current studies are investigating whether these agents could alleviate other TSC complications. Consensus statements guide identification and optimal management of many of the TSC-related complications at diagnosis and throughout the lifespan. A multidisciplinary approach is necessary for optimal management of individuals with TSC.
Article
Tuberous sclerosis complex is an autosomal dominant genetic disease characterized by growth of benign tumors (hamartomas) in multiple organs, especially the kidneys, brain, heart, lungs, and skin. Tuberous sclerosis complex is usually caused by a mutation in either the tuberous sclerosis complex 1 or tuberous sclerosis complex 2 gene, resulting in constitutive activation of mammalian target of rapamycin signaling. Currently, mammalian target of rapamycin inhibitors are recommended in adult patients with tuberous sclerosis complex for the treatment of asymptomatic, growing renal angiomyolipoma that are >3 cm in diameter and pediatric or adult patients with brain lesions (subependymal giant cell astrocytoma) that either are growing or are not amenable to surgical resection. Clinical evidence suggests that systemic administration of a mammalian target of rapamycin inhibitor may provide concurrent improvements in multiple lesions and symptoms of tuberous sclerosis complex. With the major paradigm shift in consensus guidelines toward screening at diagnosis and ongoing monitoring and with the recent availability of an effective oral treatment, it is important that nephrologists have a thorough understanding of our role in the management of patients with tuberous sclerosis complex. Because the various manifestations of tuberous sclerosis complex typically emerge at different periods during patients' lifetimes, patients will need to be followed throughout their lives. Unlike brain and cardiac lesions, renal lesions are more likely to emerge as patients age and can grow at any time. Considerations regarding long-term medication administration for the potential control of multiple tuberous sclerosis complex manifestations will need to be addressed; these include the most appropriate starting dose, appropriate doses for tumor shrinkage versus prevention of regrowth, and management of adverse events. Best practices and potential obstacles for nephrologists treating patients with tuberous sclerosis complex who have multiple manifestations are considered.