ArticlePDF Available

Deubiquitinase HAUSP Stabilizes REST and Promotes Maintenance of Neural Progenitor Cells

Authors:

Abstract and Figures

The repressor element 1-silencing transcription factor (REST) functions as a master regulator to maintain neural stem/progenitor cells (NPCs). REST undergoes proteasomal degradation through β-TrCP-mediated ubiquitylation during neuronal differentiation. However, reciprocal mechanisms that stabilize REST in NPCs are undefined. Here we show that the deubiquitylase HAUSP counterbalances REST ubiquitylation and prevents NPC differentiation. HAUSP expression declines concordantly with REST on neuronal differentiation and reciprocally with β-TrCP levels. HAUSP knockdown in NPCs decreases REST and induces differentiation. In contrast, HAUSP overexpression upregulates REST by overriding β-TrCP-mediated ubiquitylation. A consensus site (310-PYSS-313) in human REST is required for HAUSP-mediated REST deubiquitylation. Furthermore, REST overexpression in NPCs rescues the differentiation phenotype induced by HAUSP knockdown. These data demonstrate that HAUSP stabilizes REST through deubiquitylation and antagonizes β-TrCP in regulating REST at the post-translational level. Thus, HAUSP-mediated deubiquitylation represents a critical regulatory mechanism involved in the maintenance of NPCs.
HAUSP knockdown promotes neural differentiation and decreases NPC self-renewal, and REST overexpression rescues the differentiation phenotype induced by HAUSP knockdown.(a) Targeting HAUSP with shRNA promotes neuronal differentiation. 15167 NPCs were infected with lentiviruses expressing HAUSP shRNA (B5 clone) or non-targeting (NT) shRNA for 126 h and immunostained for Nestin (an NPC maker, in red) and TUJ1 (a neuronal differentiation marker, in green). (b) Quantified data from a confirmed that HAUSP knockdown increased neuronal lineage specification. The fraction of cells expressing TUJ1 (green) significantly (P<0.001) increased and the fraction of cells expressing Nestin (red) decreased after HAUSP knockdown in the NPCs. Data are means±s.d. (n=3; 200  cells per experiment). (c) Immunofluorescent staining showed that ectopic REST expression rescued the differentiation phenotype induced by HAUSP knockdown. 17231 NPCs were transfected with Flag–REST or vector control, and infected with HAUSP shRNA (B5 clone) or non-targeting shRNA lentiviruses for 126 h and immunostained for Nestin (red) and TUJ1 (green). (d) Quantified data from c indicated that ectopic expression of REST significantly (P<0.001) attenuated the increased fraction of cells expressing TUJ1 induced by HAUSP knockdown. Data are means±s.d. (n=3; 200  cells per experiment). (e) Immunoblotting confirmed that ectopic expression of REST repressed the TUJ1 expression induced by HAUSP knockdown. 17231 NPCs were transfected with Flag–REST or vector, and infected with HAUSP shRNA (B5 clone) or non-targeting shRNA lentiviruses for 96 h, and immunoblotted with specific antibodies against HAUSP, Flag, TUJ1 and α-tubulin. (f) Neurosphere formation assay showed that HAUSP knockdown reduced NPC self-renewal potential. 15167 NPCs were infected with HAUSP shRNA or non-targeting shRNA lentiviruses and allowed to form neurospheres in serum-free suspension culture. HAUSP knockdown reduced the neurosphere size and induced the attachment of neurospheres on the uncoated dishes. (g) Quantified data from f confirmed that HAUSP knockdown with two specific shRNAs (B2 and B5) significantly (P<0.001) decreased the number of neurospheres formed by 15167 NPCs. Data are means±s.d. (n=3). Uncropped images of blots are shown in Supplementary Fig. S8.
… 
HAUSP mediates REST deubiquitylation to regulate REST protein levels.(a,b) Immunoprecipitation (IP) showed that HAUSP and REST interact in NPCs. Cell lysates of ENStemA NPCs were immunoprecipitated with anti-REST (monocolonal antibody) or anti-HAUSP antibody or IgG control and then immunoblotted (IB) with anti-HAUSP and anti-REST (rabbit polyclonal) antibodies. (c) Ubiquitylation assays showed that HAUSP knockdown increased REST ubiquitylation in NPCs. 17231 NPCs were infected with lentiviruses expressing HAUSP shRNA or non-targeting shRNA for 48 h and then treated with the proteasome inhibitor MG132 for 6 h before collection for immunoprecipitation. Cell lysates were immunoprecipitated with an anti-REST- or anti-ubiquitin-specific antibody or control IgG, and then immunoblotted with anti-ubiquitin- or anti-REST-specific antibody. Both immunoprecipitation with anti-REST antibody and the reciprocal immunoprecipitation with anti-ubiquitin antibody confirmed that HAUSP knockdown increased REST polyubiquitylation. (d) Ectopic expression of wild-type HAUSP (WT-HAUSP), but not the catalytic dead mutant HAUSPC223S (Mt-HAUSP), reduced REST ubiquitylation. 17231 NPCs were transfected with the Flag-tagged WT-HAUSP (WT), Mt-HAUSP (Mt) or vector (V) control through lentiviral infection for 36 h, then treated with the proteasome inhibitor MG132 for 6 h, and subjected to analysis of REST ubiquitylation. Ectopic expression of WT-HAUSP, but not Mt-HAUSP, reduced REST ubiquitylation in the NPCs. (e) In vitro deubiquitylation assay showed that the β-TrCP-mediated REST ubiquitylation was specifically inhibited by WT-HAUSP (lane 3), but not by the Mt-HAUSP (a catalytic dead mutant, lane 4) or control deubiquitylase USP1 (lane 5). Flag–WT-HAUSP, Flag–Mt-HAUSP, Myc–REST, HA–β-TrCP and Flag–USP1 were individually overexpressed in 293T cells, and then purified with the specific antibody or the corresponding tag antibody for this assay. (f) In vivo deubiquitylation assay confirmed that the β-TrCP-mediated REST ubiquitylation was specifically attenuated by the WT-HAUSP, but not the catalytic dead Mt-HAUSP. 293T cells were transfected with the indicated sets of plasmids for 48 h, treated with the proteasome inhibitor MG132 for 6 h and then subjected to analysis of REST ubiquitylation in the samples expressing different set of proteins as indicated. Uncropped images of blots are shown in Supplementary Fig. S8.
… 
Content may be subject to copyright.
Deubiquitinase HAUSP Stabilizes REST and Promotes
Maintenance of Neural Progenitor Cells
Zhi Huang1, Qiulian Wu1, Olga A. Guryanova1, Lin Cheng1,2, Weinian Shou3, Jeremy N.
Rich1, and Shideng Bao1,*
1Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute,
Cleveland Clinic, Cleveland, Ohio 44195, USA
2Center for Experimental Research, The First People’s Hospital, Shanghai Jiaotong University,
Shanghai 200080, China
3Riley Heart Research Center, Herman B Wells Center for Pediatric Research, Department of
Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
SUMMARY
The repressor element 1-silencing transcription factor (REST) functions as a master regulator to
maintain neural stem/progenitor cells (NPCs). REST undergoes proteasomal degradation through
β-TrCP-mediated ubiquitination during neuronal differentiation. However, reciprocal mechanisms
that stabilize REST in NPCs are undefined. Here we show that deubiquitinase HAUSP
counterbalances REST ubiquitination and prevents NPC differentiation. HAUSP expression
declines concordantly with REST upon neuronal differentiation and reciprocally with β-TrCP
levels. HAUSP knockdown in NPCs decreases REST and induces differentiation. In contrast,
HAUSP overexpression up-regulates REST by overriding β-TrCP-mediated ubiquitination. A
consensus site (310-PYSS-313) of human REST is required for HAUSP-mediated REST
deubiquitination. Furthermore, REST overexpression in NPCs rescues the differentiation
phenotype induced by HAUSP knockdown. These data demonstrate that HAUSP stabilizes REST
through deubiquitination and antagonizes β-TrCP in regulating REST at post-translational level.
Thus, the HAUSP-mediated deubiquitination represents a critical regulatory mechanism involved
in the maintenance of NPCs.
Transcriptional regulators of stem cell maintenance and differentiation require exquisite
control to direct cell fate determination. Uncontrolled activation of core stem cell pathways
drives transformation while loss of function in these cellular mechanisms leads to
degenerative conditions. As regenerative medicine advances, understanding the regulation of
self-renewal and lineage commitment becomes imperative. The brain has been the focus of
numerous investigations into molecular mechanisms informing the maintenance and
differentiation of neural stem cells due to the devastating nature of brain cancers and
Users may view, print, copy, download and text and data- mine the content in such documents, for the purposes of academic research,
subject always to the full Conditions of use: http://www.nature.com/authors/editorial_policies/license.html#terms
*Correspondence should be addressed to Shideng Bao [S. B.] Cleveland Clinic, Lerner Research Institute, 9500 Euclid Avenue,
NE30, Cleveland, Ohio 44195, USA; Tel: +1 216 636 1009; Fax: +1 216 636 5454; baos@ccf.org.
AUTHOR CONTRIBUTIONS
Z.H., Q.W., O.G. and L.C. performed and planed all experiments. S.B. developed the hypothesis, coordinated the study, oversaw the
research and results and wrote the manuscript. J.R. helped to write the manuscript and provided input into design and interpretation.
W.S. provided reagents and helpful suggestions. The work was carried out in the laboratory of S.B.
COMPETING FINANCIAL INTERESTS
The authors declare that they have no competing financial interests.
NIH Public Access
Author Manuscript
Nat Cell Biol
. Author manuscript; available in PMC 2011 August 1.
Published in final edited form as:
Nat Cell Biol
. 2011 February ; 13(2): 142–152. doi:10.1038/ncb2153.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
neurodegenerative diseases. REST (also known as neuron restrictive silencer factor, NRSF)
is a critical transcription factor in regulating NPC self-renewal and lineage specification14,
but REST itself is also regulated at both transcriptional and post-transcriptional levels.
During neuronal differentiation, REST protein is targeted for proteosomal degradation by
the E3 ubiquitin ligase SCF-β-TrCP (Skp1-Cul1-F-box protein)5, 6. As REST functions as a
central transcriptional repressor of neuronal differentiation-associated genes to promote
NPC maintenance2, 7, 8, aberrant REST function is associated with neurodegenerative
diseases (e.g. Huntington’s disease) and other pathological states912. Neural tumors,
specifically medulloblastomas and neuroblastomas, express high levels of REST and forced
expression of REST can promote malignant transformation of neural progenitors1315,
although REST has been also reported as a tumor suppressor of colon cancer16. The effects
of inappropriate REST expression would suggest that unidirectional negative regulation of
REST protein by the β-TrCP-mediated ubiquitination during neuronal differentiation are
balanced by a reciprocal mechanism that promotes REST stabilization via deubiquitination
for NPC maintenance. Here, we demonstrated that deubiquitinase HAUSP (Herpesvirus-
Associated Ubiquitin-Specific Protease, also known as Ubiquitin-Specific Protease 7,
USP7)17, 18 prevents REST degradation through deubiquitination and promotes NPC
maintenance.
RESULTS
HAUSP positively regulates REST protein levels in neural progenitor cells
Post-translational modifications are capable of rapidly regulating protein function and
stability in response to cell state or external stimuli creating optimal points of network
control for systems requiring precise regulation, including stem cell pathways. As
irreversible commitment to a neuronal fate is controlled by the loss of REST protein by
ubiquitination-mediated proteosomal degradation, we reasoned that this mechanism requires
counterbalancing deubiquitination to prevent an instability of the control of NPC
maintenance. We therefore screened for deubiquitinases that are nuclear in location and
decline in expression during NPC differentiation. Using these criteria, we identified HAUSP
as a deubiquitinase that gradually decreased in expression coordinated with decreased REST
levels as fetal NPCs were induced to differentiate with all-trans retinoic acid (RA) treatment
(Fig. 1a). HAUSP levels inversely correlated with lineage commitment as measured by
acquisition of the neuronal marker TUJ1 (type III β-tubulin, a REST target gene) and the E3
ubiquitin ligase β-TrCP that targets REST for degradation (Fig. 1a). These results were
confirmed by immunofluorescent staining of NPCs undergoing RA-induced differentiation
(Fig. 1b, c). HAUSP and REST were highly expressed in nuclei of NPCs and their protein
levels declined coordinately during the process of differentiation (Fig. 1b), while the
expression of the neuronal lineage marker TUJ1 significantly increased upon differentiation
(Fig. 1c). Thus, HAUSP deubiquitinase is positively associated with REST protein levels in
NPCs.
To interrogate a mechanistic link between HAUSP activity and REST stability, we
examined the effects of targeting HAUSP with shRNA (short hairpin RNA) on REST
protein levels in NPCs. NPCs were transduced with lentiviruses expressing either a non-
targeting (NT) control shRNA or one of two non-overlapping shRNA sequences (designated
B2 and B5) against human HAUSP resulting in attenuation of HAUSP levels (70–85%
reduction compared to the non-targeting control). HAUSP knockdown reduced REST
protein levels but not the co-repressor CoREST in two separate NPC lines (Fig. 2 a, b),
despite the presence of CoREST in a repressor complex with REST3, 19 supporting a
specificity of HAUSP on REST rather than a general effect on the repressor complex.
Immunofluorescent staining also showed that HAUSP knockdown resulted in reduced REST
protein levels in NPC nuclei (Fig. 2c). This result was confirmed in an additional NPC line
Huang et al. Page 2
Nat Cell Biol
. Author manuscript; available in PMC 2011 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
by puromycin selection of cells expressing the HAUSP-targeting shRNA reflected in nearly
universal reduction of REST (Fig. 2d). In contrast, ectopic expression of HAUSP (HA-
tagged HAUSP) in NPCs increased REST protein levels (Supplementary Information, Fig.
S1a). Immunofluorescent staining validated that single cells overexpressing HA-HAUSP
showed elevated REST protein levels (Supplementary Information, Fig. S1b). Collectively,
these data demonstrate that HAUSP deubiquitinase positively regulates REST protein levels.
HAUSP knockdown in neural progenitor cells induces neuronal differentiation and
disrupts self-renewal
As REST prevents neuronal differentiation13, 5, 7 and we have found that HAUSP
controls REST, we investigated the impact of targeting HAUSP on NPC neuronal
differentiation. To interrogate the effect of HAUSP knockdown on cell fate determination in
NPCs, we assayed Nestin as an NPC marker and TUJ1 as a neuronal marker by
immunofluorescent staining. NPCs were transduced with either HAUSP-targeting shRNA
(shHAUSP) or control NT shRNA. The overwhelming majority of NPCs expressing NT
shRNA were Nestin-positive and rarely TUJ1-positive (Fig. 3a, top panel), indicating
maintenance of an undifferentiated NPC state. In contrast, most cells targeted with
shHAUSP became TUJ1-positive and only rarely remained Nestin-positive (Fig. 3 a, bottom
panel), suggesting that targeting HAUSP induced neuronal differentiation. Indeed,
quantified data indicate that HAUSP knockdown significantly increased the proportion of
differentiated cells (Fig. 3b), suggesting that HAUSP deubiquitinase prevents NPC
differentiation. To further causally link REST regulation to the effect of HAUSP on
preventing NPC differentiation, we attempted to rescue the differentiation phenotype
induced by HAUSP knockdown by overexpressing REST. Immunofluorescent staining
showed that HAUSP knockdown in NPCs transfected with a vector control retained a
differentiation phenotype but this effect was attenuated by overexpression of Flag-REST
(Fig. 3c, d). This result was further validated by immunoblotting analysis demonstrating that
the TUJ1 expression induced by HAUSP knockdown was reduced by the ectopic expression
of Flag-REST in NPCs (Fig. 3e). These data indicate that ectopic expression of REST
largely overrode the cell differentiation induced by HAUSP knockdown. In addition,
knockdown of REST itself to the levels caused by HAUSP knockdown in NPCs induced
similar differentiation (Supplementary Information, Fig. S2a–c), suggesting that REST
knockdown phenocopied HAUSP knockdown in the induction of neuronal differentiation.
Collectively, these data demonstrate that HAUSP functions largely through REST to prevent
NPC differentiation.
Self-renewal is a defining characteristic of stem cells so we examined the role of HAUSP in
regulating NPC self-renewal potential. Targeting HAUSP via shRNA reduced the self-
renewal of two different NPC preparations as demonstrated by the neurosphere formation
assay (Fig. 3f, g; Supplementary Information, Fig. S3a, b). shHAUSP not only attenuated
neurosphere formation frequency (a measure of self-renewal) and the neurosphere size (a
measure of proliferation) but also induced cell attachment under neurosphere culture
conditions (Fig. 3f; Supplementary Information, Fig. S3a). Moreover, the reduced
neurosphere formation induced by HAUSP knockdown was significantly rescued by ectopic
expression of Flag-REST (Supplementary Information, Fig. S3c, d). Taken together, these
data demonstrate that HAUSP deubiquitinase maintains NPCs mainly by controlling REST
levels.
HAUSP stabilizes REST protein through deubiquitination
To determine the mechanism through which HAUSP regulates REST expression, we
interrogated effects of HAUSP on REST at different regulating levels. Theoretically,
HAUSP might indirectly modulate REST through transcriptional control but we confirmed
Huang et al. Page 3
Nat Cell Biol
. Author manuscript; available in PMC 2011 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
that HAUSP knockdown did not alter REST mRNA expression on RT-PCR analysis (Fig.
4). HAUSP functions as a deubiquitinase making post-translational regulation, most likely
deubiquitination, as a potential link to REST control. We determined that HAUSP
physically interacts with REST as demonstrated in co-immunoprecipitation assays (Fig. 5a,
b; Supplementary Information, Fig. S4a, b), suggesting that HAUSP may mediate REST
deubiquitination to prevent REST proteosomal degradation. We therefore examined effects
of HAUSP knockdown on REST ubiquitination in NPCs. NPCs were transduced with NT
shRNA or shHAUSP and treated with the MG132 proteasome inhibitor to assay the
ubiquitinated REST. HAUSP knockdown increased REST poly-ubiquitination as
demonstrated by both anti-REST immunoprecipitation (IP) and anti-Ubiquitin reciprocal IP
(Fig. 5c; Supplementary Information, Fig. S4c). To confirm this result, we examined the
effect of HAUSP overexpression on REST ubiquitination. NPCs were transduced with
lentiviruses expressing Flag-tagged wild-type (Wt) HAUSP or catalytically dead mutant
(Mt) HAUSP (C223S)20 and then assessed for REST ubiquitination. Ectopic expression of
Wt-HAUSP but not the Mt-HAUSP decreased REST ubiquitination and increased REST
protein levels (Fig. 5d; Supplementary Information, Fig. S4d). Collectively, these data
demonstrate that HAUSP negatively regulates REST ubiquitination and promotes REST
stabilization.
To further address whether REST deubiquitination is directly and specifically mediated by
HAUSP, we performed an
in vitro
deubiquitination assay20 with purified wild-type HAUSP
(Flag-Wt-HAUSP), catalytic dead mutant HAUSP (Flag-Mt-HAUSP), or a control
deubiquitinase USP1 (Flag-USP1). Wt-HAUSP specifically counteracted β-TrCP-mediated
REST ubiquitination, while the Mt-HAUSP or the control deubiquitinase USP1 did not alter
the REST ubiquitination (Fig. 5e), suggesting that HAUSP directly and specifically
deubiquitinated REST and suppressed REST ubiquitination. This result was validated by the
in vivo
deubiquitination assay in 293T cells expressing Wt-HAUSP or the catalytic dead Mt-
HAUSP in combination with REST, Ubiquitin and β-TrCP (Fig. 5f). Consistently,
expression of β-TrCP increased REST poly-ubiquitination (Fig. 5f, lane 2, 3), which was
attenuated by expression of Wt-HAUSP (Fig. 5f, lane 2–4). In contrast, expression of the
catalytic dead Mt-HAUSP did not modulate the REST poly-ubiquitination induced by β-
TrCP expression (Fig. 5f, lane 3–5). These data demonstrated that HAUSP counteracts β-
TrCP-mediated REST ubiquitination through specific deubiquitination.
As HAUSP knockdown reduced REST protein levels and promoted NPC differentiation, we
further examined whether ectopic expression of Wt-HAUSP overrides shHAUSP-induced
differentiation. Because the B5 shHAUSP targets the 3’-end non-coding region of
endogenous HAUSP mRNA while the Wt-HAUSP and Mt-HAUSP constructs do not
contain the 3’-end non-coding sequence, mRNA from Wt-HAUSP or Mt-HAUSP is not
targeted by B5 shHAUSP. Thus, we were able to simultaneously knock down endogenous
HAUSP and overexpress Wt-HAUSP or Mt-HAUSP in NPCs. Immunofluorescent staining
showed that ectopic expression of Wt-HAUSP but not the Mt-HAUSP almost fully rescued
the differentiation phenotype induced by knockdown of endogenous HAUSP with the B5
shHAUSP (Fig. 6a, b). Immunoblotting confirmed that ectopic expression of Wt-HAUSP
but not the Mt-HAUSP attenuated TUJ1 expression induced by knockdown of endogenous
HAUSP (Fig. 6c). Moreover, deubiquitination assay demonstrated that expression of Wt-
HAUSP but not the Mt-HAUSP attenuated the REST ubiquitination induced by B5
shHAUSP (Fig. 6d), showing that Wt-HAUSP functioned through deubiquitination to
stabilize REST and rescued the differentiation phenotype induced by knockdown of
endogenous HAUSP. These data demonstrate that expression of functional HAUSP is
critical for preventing REST ubiquitination and suppressing NPC differentiation.
Huang et al. Page 4
Nat Cell Biol
. Author manuscript; available in PMC 2011 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
A consensus binding site (310-PYSS-313) of human REST is required for the HAUSP-
mediated REST deubiquitination
Our data demonstrate that HAUSP directly deubiquitinates REST. As the known HAUSP
substrates usually contain a consensus sequence (P/AXXS) for the HAUSP-mediated
specific deubiquitination21, 22, we sought to identify the HAUSP consensus binding
sequences on the human REST protein. Human REST contains five P/AXXS sequences
(Supplementary Information, Fig. S5). To determine which sequence is critical for the
HAUSP-mediated REST deubiquitination, we sequentially mutated each of the critical
amino acid Ser to Ala (SA) of these potential sites and expressed mutant REST constructs
in NPCs. We found that only the S313A mutation of 310-PYSS-313 of human REST
disrupted the HAUSP-mediated REST deubiquitination (Fig. 7a, b). In contrast, a similar
mutation (S1042A) on other potential sites such as 1039-PQES-1042 did not prevent the
HAUSP-mediated REST deubiquitination, and S313A/S1042A double mutations (Flag-
REST-AA) showed similar effect to the S313A single mutation on suppressing the HAUSP-
mediated REST deubiquitination (Fig. 7b). Thus, we have identified the consensus HAUSP
binding site (310-PYSS-313) on human REST that is required for the HAUSP-mediated
REST deubiquitination. These data further support that REST deubiquitination is
specifically mediated by HAUSP.
HAUSP and β-TrCP function as oppositional counterparts to control REST protein stability
at post-translational level
As both HAUSP and REST protein levels gradually decline and the E3 ubiquitin ligase β-
TrCP increases during NPC differentiation (Fig. 1), we next examined the roles of reduced
HAUSP and increased β-TrCP in regulating REST ubiquitination during neural
differentiation. Under RA-induced differentiation of NPCs, REST poly-ubiquitination
increased with reduction of total REST protein levels (Fig. 7c; Supplementary Information,
Fig. S6a). Because HAUSP knockdown increased REST ubiquitination (Fig. 5c) and β-
TrCP promoted REST ubiquitination (Fig. 5e, f), it is likely that both decreased HAUSP and
increased β-TrCP contribute to increased REST ubiquitination during neuronal
differentiation. We hypothesized that HAUSP functions as a critical counterbalance to β-
TrCP to inhibit REST ubiquitination and maintain NPCs. Thus, we examined the effects of
knocking down both HAUSP and β-TrCP to determine REST ubiquitination during NPC
differentiation. To initiate neuronal differentiation, NPCs were treated with RA for 48 hours
and transduced with NT shRNA, shHAUSP, β-TrCP – targeting shRNA (shβ-TrCP), or both
shHAUSP and shβ-TrCP. Cells were treated with RA long enough to initiate differentiation
but assessed before terminal differentiation so both HAUSP and β-TrCP were expressed. As
expected, shHAUSP increased REST ubiquitination (Fig. 7d, lane 1, 2), while shβ-TrCP
decreased REST ubiquitination (Fig. 7d, lane 1, 3). Simultaneous targeting of HAUSP and
β-TrCP caused intermediate levels of REST ubiquitination (Fig. 7d, lane 2–4), suggesting
that both HAUSP and β-TrCP regulate REST ubiquitination during NPC differentiation.
Collectively, these data demonstrated HAUSP deubiquitinase and β-TrCP ubiquitin E3
ligase function as oppositional counterparts (“Ying-Yang”) to control REST protein levels
(Fig. 8). To further confirm this important point, we reconstituted a regulatory system by
overexpressing Myc-REST and HA-ubiquitin in combination with HAUSP, Flag-β-TrCP or
both HAUSP and Flag-β-TrCP in 293T cells that have minimal expression of endogenous
REST and HAUSP. Thus, we were able to directly assess REST regulation by the β-TrCP-
mediated ubiquitination and the HAUSP-mediated deubiquitination in a defined cellular
system. Consistently, expression of HAUSP reduced REST poly-ubiquitination
(Supplementary Information, Fig. S6b, lanes 2, 1), and overexpression of β-TrCP increased
REST ubiquitination (Supplementary Information, Fig. S6b, lanes 3, 2). Importantly,
overexpression of HAUSP with β-TrCP together abolished the increased REST
ubiquitination induced by β-TrCP overexpression (Supplementary Information, Fig. S6b,
Huang et al. Page 5
Nat Cell Biol
. Author manuscript; available in PMC 2011 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
lanes 4, 3). Taken together, these data further validated that both HAUSP-mediated
deubiquitination and the β-TrCP-mediated ubiquitination serve opposite roles to control
REST protein stability at the post-translational level. HAUSP stabilizes REST protein
through deubiquitination and promotes NPC maintenance, while β-TrCP mediates REST
degradation through ubiquitination and stimulates NPC differentiation. The balance between
ubiquitination and deubiquitination may determine the net REST protein levels and cellular
fate. This post-translational regulatory mechanism of REST is crucial for determining NPC
maintenance or differentiation.
DISCUSSION
We now demonstrate that HAUSP stabilizes REST through deubiquitination and promotes
maintenance of NPCs. HAUSP was originally identified to be associated with viral proteins
such as ICP0 (Herpesvirus) and EBNA1 (EB virus) during viral infection17, 18, 22. A series
of elegant studies have demonstrated that HAUSP also regulates the stability and functions
of several important proteins under normal and stress conditions20, 21, 2325. Our study
expands the roles of the HAUSP deubiquitinase in the maintenance of NPCs by stabilizing
REST to repress neuronal differentiation. Although HAUSP has several molecular targets,
the ability to rescue the effects of HAUSP modulation on NPC maintenance and
differentiation by REST expression indicates that HAUSP functions largely through REST
to prevent NPC differentiation. However, it is possible that HAUSP may also control other
stem cell transcription factors for the maintenance of other stem cells.
REST is a key transcriptional repressor of neuronal differentiation genes13, 7, 8 to prevent
NPC differentiation18. In accordance with its role in silencing both neuronal and non-
neuronal genes, REST is also essential for embryonic development and for a number of
cellular responses8, 26, 27. REST has also been implicated in the regulation of mitotic
fidelity in non-neural cells6 and was proposed as a tumor suppressor16. Although there is
controversy over the requirement of REST for maintaining ESC pluripotency2830, the
important role of REST in repressing neuronal differentiation and promoting NPC
maintenance has been extensively demonstrated18. Our study suggested that post-
translational control of REST by ubiquitination and deubiquitination is critical for regulating
REST protein levels to determine cellular fate of NPCs.
We have demonstrated that HAUSP specifically stabilizes REST protein through
deubiquitination and a critical consensus site (310-PYSS-313) on human REST is required
for the HAUSP-mediated REST deubiquitination. It has been reported that the association of
TRF2 (telomere repeat factor 2) with REST also prevents the ubiquitin proteasome-mediated
degradation of REST31. However, as TRF2 does not have deubiquitinase activity, TRF2 is
unlikely to deubiquitinate REST. Whether TRF2 modulates HAUSP or β-TrCP function to
indirectly affect REST stability is under investigation. Our study establishes the
ubiquitination-deubiquitination system as a critical post-translational control mechanism to
regulate the key stem cell transcriptional factors and thus determine stem cell maintenance
or differentiation. Based on our results and previous studies5, 6, we propose that
deubiquitination and ubiquitination system works as “Ying-Yang” control system to regulate
REST protein levels (Fig. 8) with the net balance between the HAUSP-mediated
deubiquitination and β-TrCP-mediated ubiquitination controlling REST protein levels to
direct cell fate. The relative activity of deubiquitination and ubiquitination for REST may
define the maintenance of “stemness” or initiation of neuronal differentiation. When the
HAUSP-mediated deubiquitination overrides β-TrCP-mediated ubiquitination, REST is
stabilized to suppress differentiation and promote NPC maintenance. In contrast, when the
β-TrCP-mediated ubiquitination exceeds HAUSP-mediated deubiquitination, REST is
targeted for degradation, which promotes cell differentiation by releasing repression of
Huang et al. Page 6
Nat Cell Biol
. Author manuscript; available in PMC 2011 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
differentiation-associated genes. This paradigm of reciprocal yoked post-translational
control may be present in other stem cell regulatory networks to permit cells to efficiently
and finely tune the levels of critical factors that have potent cellular effects in excess or in
shortage.
In conclusion, our study uncovered a crucial post-translational control mechanism to
regulate the key stem cell transcription factor for cell fate determination of NPCs. NPCs
have been proposed as regenerative resources for some CNS diseases. As the use of these
cells requires sufficient proliferation to supply requisite cell numbers while permitting
appropriate differentiation for the stem cell-based therapies, understanding the molecular
mechanisms controlling NPC maintenance and differentiation will have significant impacts
on improving the treatment of neural degenerative diseases.
METHODS
Cells and cell culture
15167 and 17231 NPCs (derived from human fetal brains, Lonza) and the ENStemA NPCs
(derived from a human embryonic stem cell line, Chemicon/Millipore) were cultured in
neurobasal medium supplemented with B27 and epidermal growth factor (EGF)/basic
fibroblast growth factor (bFGF) as described in our previous reports32, 33. These cells were
cultured either in neurosphere suspension or in attached monolayer on the tissue culture
dishes coated with the BD Matrigel hESC-qualified matrix (BD Bioscience). To induce
differentiation, cells were treated with 1 µM of all-trans retinoic acid (RA, Sigma-Aldrich)
for the indicated time.
Immunoblotting and immunofluorescent staining
These basic methods were performed as described in our previous reports3234. Anti-
human REST monoclonal antibody (murine Clone 7D1.3, Millipore), rabbit polyclonal anti-
REST antibodies (Abcam, Santa Cruz), rabbit polyclonal anti-HAUSP antibody (Abcam),
anti-human HAUSP monoclonal antibody (Bethyl laboratories), anti-β-TrCP antibodies
(Santa Cruz), anti-Flag (Sigma-Aldrich), anti-HA (Sigma-Aldrich), anti-TUJ1 (Covance),
anti-Nestin (Millipore), anti-Myc tag (Sigma-Aldrich), anti-Ubiquitin (P4D1, Santa Cruz)
and anti-α-Tubulin (Sigma-Aldrich) antibodies were used for immunoblotting,
immunoprecipitation or immunofluorescent staining. Briefly, for immunofluorescent
staining NPCs cultured on the BD Matrigel-coated cover glasses were induced by 1 µM of
RA for differentiation or infected with lentiviruses expressing HAUSP-targeting shRNA
(shHAUSP) or non-targeting (NT) control shRNA, fixed in 4% PFA (paraformaldehyde) for
15 minutes, permeablized, blocked by blocking buffer containing 0.1% BSA (bovine serum
albumin) and 0.3% triton X-100 for 30 minutes, incubated with the primary antibodies over
night at 4 °C, incubated with the fluorescence-conjugated secondary antibodies (Invitrogen)
at room temperature for 1 hour. Nuclei were counterstained with DAPI.
Knockdown by lentiviral vector-mediated shRNA
shRNA clones for knocking down human HAUSP, REST and β-TrCP were obtained by
screening lentivirus shRNA sets (Mission shRNA, Sigma-Aldrich). The production of
lentivirus and viral infection of NPCs were performed as previously described33, 34.
Lentiviral infection efficiency in NPCs was determined with the lentiviruses expressing GFP
(green-fluorescent protein). When the MOI (Multiplicity of Infection) of 3 was used for the
infection, the majority (>93%) of NPCs were infected by the viruses and expressed GFP
(Supplementary Information, Fig. S7).
Huang et al. Page 7
Nat Cell Biol
. Author manuscript; available in PMC 2011 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Generations of plasmid constructs
Human cDNA clones for REST (BC132859), β-TrCP (MGC: 40028) and ubiquitin (MGC:
8385) were obtained from Open Biosystems. pCI-neo HAUSP was acquired from B.
Vogelstein through Addgene. Open reading frames of these cDNAs were PCR-amplified
using Platinum High Fidelity PCR Supermix (Invitrogen) according to manufacturer’s
protocol and then subcloned into XbaI and BamHI sites of lentivector pLCMV-Neo (a kind
gift of Dr. Peter Chumakov) with either Flag, HA, His or Myc tag coding sequences in
frame and verified by sequencing. All specific mutations were generated by using
QuikChange® II XL Site-Directed Mutagenesis Kit (Stratagene) according to
manufacturer’s instruction. The catalytic dead mutant (223CS) HAUSP (Mt-HAUSP)
was generated with PCR primers 5’-GAA TCA GGG AGC GAC TTC TTA CAT GAA
CAG CCT GC-3’ and 5’-GCA GGC TGT TCA TGT AAG AAG TCG CTC CCT GAT
TC-3’. The REST-S313A mutant (HAUSP binding site mutation) were generated with PCR
primers 5’-CTT TGT CCT TAC TCA GCT TCT CAG AAG ACT CATC-3’ and 5’-GAT
GAG TCT TCT GAG AAG CTG AGT AAG GAC AAA G-3’.
In vivo ubiquitination and deubiquitination assay in NPCs
Human NPCs (before and after HAUSP or β-TrCP knockdown), RA-induced differentiated
cells, or the cells overexpressing HAUSP, β-TrCP, REST or targeting shRNAs subjected for
the ubiquitination assays were treated with the proteasome inhibitor MG132 (20 µM, Sigma-
Aldrich) for 6 hours and then harvested for immunoprecipitation (IP) with anti-REST, anti-
Flag, anti-HA, or anti-Ubiquitin antibody followed immunobloting with an anti-Ubiquitin,
anti-REST, or anti-HA antibody. Briefly, cell lysates (500 µg of total protein) were
incubated with 1 µg of anti-REST antibody (H-290, Santa Cruz), anti-Ubiquitin antibody
(P4D1, Santa Cruz), or normal rabbit IgG with constant rotation overnight at +4°C.
Immunocomplexes were captured by 20 µl of Protein A/G Plus Agarose beads (Santa Cruz)
for 1 hour at +4°C, washed 3 times with ice-cold lysis buffer and eluted in 2× loading buffer
by boiling for 10 minutes, and then analyzed by immunoblotting. Proteins were resolved on
Tris-acetate gels (Invitrogen), blotted onto PVDF (polyvinylidene) membranes and probed
by antibodies specific to REST (Millipore) and Ubiquitin (P4D1, Santa Cruz).
In vitro deubiquitination assay
The
in vitro
deubiquitination assay was performed as described6. Myc-REST-WT was
immunoprecipitated with anti-REST antibody from 293T cells transfected with Myc-REST-
WT plasmid. Flag-β-TrCP, Flag-USP1, Flag-WT-HAUSP and Flag-Mt-HAUSP were
purified by the same way.
In vitro
ubiquitination assay was performed in a 25 µl mixture
including the indicated purified proteins and 50 mM Tris-HCl pH 7.6, 5 mM MgCl2, 0.6
mM dithiothreitol, 2 mM ATP, 1.5 ng/µl His-E1, 10 ng/µl His-Ubc3, 10 ng/µl His-Ubc5, 2.5
µg/µl Ubiquitin, and 1 µM ubiquitin aldehyde (all from Boston Biochem). Then the reactions
were incubated at 30°C for 1 hour and analyzed by anti-REST Western blot.
REST-HAUSP interaction
REST Immunoprecipitation
: 293T cells were transfected with plasmids expressing Myc-
REST and Flag-HAUSP using Lipofectamine and Plus reagent (Invitrogen) according to
manufacturer’s recommendations. Cells were harvested in lysis buffer (50 mM Tris-HCl, pH
7.4, 150 mM NaCl, 5 mM EDTA, 1% NP-40 supplemented with protease inhibitors) and
lysates were pre-cleared by centrifugation at 15k
g
for 15 minutes. 500 µg of total protein
lysate were subjected to immunoprecipitation with 2 µg of α-REST (NRSF) antibody
(H-290, Santa Cruz) or normal rabbit IgG for 3 hours at 4°C. Immunocomplexes were
captured by 20 µl of protein A/G Plus agarose beads (Santa Cruz) for 1 hour at 4°C, washed
5 times with lysis buffer, eluted in 2× Laemmli buffer by boiling for 5 minutes and resolved
Huang et al. Page 8
Nat Cell Biol
. Author manuscript; available in PMC 2011 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
by SDS-PAGE.
HA (HAUSP) Immuoprecipitation
: 293T cells were transfected with
plasmids expressing HA-HAUSP (or empty vector) and Myc-REST using Lipofectamine
and Plus reagent (Invitrogen) according to manufacturer’s instructions. Cells were harvested
in lysis buffer (50 mM Tris-HCl pH 7.4, 150 mM NaCl, 5 mM EDTA, 1% NP-40 with
protease inhibitors) and lysates were precleared by centrifugation at 15k
g
for 15 minutes.
500 µg of total protein lysate were incubated with 25 µl of EZview Red Anti-HA Affinity
Gel (Sigma-Aldrich) for 3 hours at 4°C and washed 5 times with lysis buffer. Captured
proteins were eluted in 2× Laemmli buffer by boiling for 5 minutes and resolved by SDS-
PAGE.
REST ubiquitination and deubiquitination assays in 293T
293T cells were transfected using Lipofectamine and Plus reagent (Invitrogen) to express
HA-Ub, Myc-REST, Flag-β-TrCP, HAUSP or corresponding empty vectors as indicated.
Prior to harvesting cells were treated with 20 µM MG-132 proteasome inhibitor for 4 hours.
Cells were harvested in lysis buffer (50 mM Tris-HCl, pH 7.4, 150 mM NaCl, 5 mM EDTA,
1% NP-40 supplemented with protease inhibitors) and lysates were precleared by
centrifugation at 15k
g
for 15 minutes. 500 µg of total protein lysate were incubated with 25
µl of EZview Red Anti-HA Affinity Gel (Sigma-Aldrich) for 3 hours at 4°C with constant
rotation, beads were washed 5 times in lysis buffer and captured proteins were eluted in 2×
Laemmli buffer by boiling for 5 minutes.
Real Time PCR
mRNA samples were isolated from 15167 or ENStemA NPCs infected with lentiviruses
expressing HAUSP-targeting shRNA or non-targeting control shRNA for 72 hours, and then
subjected for real-time PCR (RT-PCR) analysis using the following primer pair: HAUSP
(Forward Primer: 5’-ACT TTG AGC CAC AGC CCG GTA ATA-3’, Reverse Primer: 5’-
GCC TTG AAC ACA CCA GCT TGG AAA-3’); REST (Forward Primer: 5’-CGC CAT
GCA AGA CAG GTT CAC AAT-3’, Reverse Primer: 5’-AGC TGC ATA GTC ACA TAC
AGG GCA-3’); Co-REST (Forward Primer: 5’-AAC GGG ACA ATC TTG GCA TGT
TGG-3’, Reverse Primer: 5’-AGA GCC TGT TCC ATG TTG TAC CCA-3’) and TUJ1
(Forward Primer: 5’-ATC AGC AAG GTG CGT GAG GAG TAT-3’, Reverse Primer: 5’-
TCG TTG TCG ATG CAG TAG GTC TCA-3’) in the 7900HT Fast Real-Time PCR
System (AB Applied Biosystems).
Statistical analysis
All grouped data are presented as mean ± standard deviation (s.d.). Difference between
groups was assessed by one-way ANOVA or one-way ANOVA on ranks tests. SigmaStat
Software (Version 3.5) was used for all statistical analyses.
Supplementary Material
Refer to Web version on PubMed Central for supplementary material.
Acknowledgments
We thank members in Dr. Rich’s laboratory for helpful discussions and suggestions. We are also grateful to the
Flow Cytometry Core, Imaging Core and Central Cell Services at Cleveland Clinic Lerner Research Institute for
their help and services. This work was supported by a research fund from Cleveland Clinic Foundation and a NIH
grant (NS070315) to S.B.
Huang et al. Page 9
Nat Cell Biol
. Author manuscript; available in PMC 2011 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
References
1. Chong JA, et al. REST: a mammalian silencer protein that restricts sodium channel gene expression
to neurons. Cell. 1995; 80:949–957. [PubMed: 7697725]
2. Schoenherr CJ, Anderson DJ. The neuron-restrictive silencer factor (NRSF): a coordinate repressor
of multiple neuron-specific genes. Science. 1995; 267:1360–1363. [PubMed: 7871435]
3. Ballas N, Grunseich C, Lu DD, Speh JC, Mandel G. REST and its corepressors mediate plasticity of
neuronal gene chromatin throughout neurogenesis. Cell. 2005; 121:645–657. [PubMed: 15907476]
4. Abrajano JJ, et al. Differential deployment of REST and CoREST promotes glial subtypes
specificcatio and oligodendrocyte lineage maturation. PLoS One. 2010; 4:e7665. [PubMed:
19888342]
5. Westbrook TF, et al. SCFβ-TRCP controls oncogenic transformation and neural differentiation
through REST degradation. Nature. 2008; 452:370–374. [PubMed: 18354483]
6. Guardavaccaro D, et al. Control of chromosome stability by the β-TrCP-REST-Mad2 axis. Nature.
2008; 452:365–369. [PubMed: 18354482]
7. Ballas N, Mandel G. The many faces of REST oversee epigenetic programming of neuronal genes.
Curr. Opin. Neurobiol. 2005; 15:500–506. [PubMed: 16150588]
8. Johnson R, et al. REST regulates distinct transcriptional networks in embryonic and neural stem
cells. PLoS Biol. 2008; 6:2205–2219.
9. Zuccato C, et al. Huntingtin interacts with REST/NRSF to modulate the transcription of NRSE-
controlled neuronal genes. Nature Genet. 2003; 35:76–83. [PubMed: 12881722]
10. Lepagnol-Bestel AM, et al. Huntingtin interacts with REST/NRSF to modulate the transcription of
NRSE-controlled neuronal genes. Hum. Mol. Genet. 2009; 18:1405–1414. [PubMed: 19218269]
11. Tahiliani M, et al. The histone H3K4 demethylase SMCX links REST target genes to X-linked
mental retardation. Nature. 2007; 447:601–605. [PubMed: 17468742]
12. Canzonetta C, et al. DYRK1A-dosage imbalance perturbs NRSF/REST levels, deregulating
pluripotency and embryonic stem cell fate in Down syndrome. Am. J. Hum. Genet. 2008; 83:388–
400. [PubMed: 18771760]
13. Lawinger P, et al. The neuronal repressor REST/NRSF is an essential regulator in
medulloblastoma cells. Nature Med. 2000; 6:826–831. [PubMed: 10888935]
14. Su X, et al. Abnormal expression of REST/NRSF and Myc in neural stem/progenitor cells causes
cerebellar tumors by blocking neuronal differentiation. Mol. Cell Biol. 2006; 26:1666–1678.
[PubMed: 16478988]
15. Fuller GN, et al. Many human medulloblastoma tumors overexpress repressor element-1 silencing
transcription (REST)/neuron-restrictive silencer factor, which can be functionally countered by
REST-VP16. Mol. Cancer Ther. 2005; 4:343–349. [PubMed: 15767543]
16. Westbrook T, et al. A genetic screen for candidate tumor suppressors identifies REST. Cell. 2005;
121:837–848. [PubMed: 15960972]
17. Holowaty MN, et al. Protein profiling with Epstein-Barr nuclear antigen-1 reveals an interaction
with the herpesvirus-associated ubiquitin-specific protease HAUSP/USP7. J. Biol. Chem. 2003;
278:29987–29994. [PubMed: 12783858]
18. Boutell C, Canning M, Orr A, Everett RD. Reciprocal activities between herpes simplex virus type
1 regulatory protein ICP0, a ubiquitin E3 ligase, and ubiquitin-specific protease USP7. J. Virol.
2005; 79:12342–12354. [PubMed: 16160161]
19. Andres ME, et al. CoREST: a functional corepressor required for regulation of neural-specific gene
expression. Proc. Natl. Acad. Sci. USA. 1999; 96:9873–9878. [PubMed: 10449787]
20. Van der Horst A, et al. FOXO4 transcriptional activity is regulated by monoubiquitination and
USP7/HAUSP. Nature Cell Biol. 2006; 8:1064–1073. [PubMed: 16964248]
21. Sheng Y, et al. Molecular recognition of p53 and MDM2 by USP7/HAUSP. Nature Struct. Mol.
Biol. 2006; 13:285–291. [PubMed: 16474402]
22. Saridakis V, et al. Structure of the p53 binding domain of HAUSP/USP7 bound to Epstein-Barr
nuclear antigen 1 implications for EBV-mediated immortalization. Mol. Cell. 2005; 18:25–36.
[PubMed: 15808506]
Huang et al. Page 10
Nat Cell Biol
. Author manuscript; available in PMC 2011 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
23. Li M, Brooks CL, Kon N, Gu W. A dynamic role of HAUSP in the p53-Mdm2 pathway. Mol.
Cell. 2004; 13:879–886. [PubMed: 15053880]
24. Cummins JM, et al. Tumour suppression: disruption of HAUSP gene stabilizes p53. Nature. 2004;
428:6982.
25. Song MS, et al. The deubiquitinylation and localization of PTEN are regulated by a HAUSP-PML
network. Nature. 2008; 455:813–817. [PubMed: 18716620]
26. Jorgensen HF, et al. REST selectively represses a subset of RE1-containing neuronal genes in
mouse embryonic stem cells. Development. 2009; 136:715–721. [PubMed: 19201947]
27. Chen ZF, Paquette AJ, Anderson DJ. NRSF/REST is required in vivo for repression of multiple
neuronal target genes during embryogenesis. Nature Genet. 1998; 20:136–142. [PubMed:
9771705]
28. Singh SK, Kagalwala MN, Parker-Thornburg J, Adams H, Majumder S. REST maintains self-
renewal and pluripotency of embryonic stem cells. Nature. 2008; 453:223–227. [PubMed:
18362916]
29. Buckley NJ, Johnson R, Sun YM, Stanton LW. Is REST a regulator of pluripotency? Nature. 2009;
457:E5–E6. [PubMed: 19242418]
30. Yamada Y, Aoki H, Kunisada T, Hara A. Rest promotes the early differentiation of mouse ESCs
but is not required for their maintenance. Cell Stem Cell. 2010; 6:10–15. [PubMed: 20085738]
31. Zhang P, et al. Nontelomeric TRF2-REST interaction modulates neuronal gene silencing and fate
of tumor and stem cells. Curr. Biol. 2008; 18:1489–1494. [PubMed: 18818083]
32. Bao S, et al. Cancer stem cells promote radioresistance of malignant glioma through preferential
activation of DNA damage checkpoint and repair. Nature. 2006; 444:756–760. [PubMed:
17051156]
33. Bao S, et al. Targeting cancer stem cells through L1CAM suppresses glioma growth. Cancer Res.
2008; 68:6043–6048. [PubMed: 18676824]
34. Li Z, et al. Hypoxia-inducible factors regulate tumorigenic capacity of glioma stem cells. Cancer
Cell. 2009; 15:501–513. [PubMed: 19477429]
Huang et al. Page 11
Nat Cell Biol
. Author manuscript; available in PMC 2011 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 1.
HAUSP and REST protein levels decline coordinately during neuronal differentiation. (a)
Immunoblotting of HAUSP, β-TrCP, REST and TUJ1 (a neuronal differentiation marker)
during differentiation. 15167 NPCs (neural stem/progenitor cells derived from a fetal brain
by Lonza) were induced by all-trans retinoic acid (RA) to undergo cellular differentiation for
the indicated times. HAUSP and REST protein levels gradually decreased, while the β-TrCP
E3 ubiquitin ligase and the TUJ1 (type III β-tubulin, a REST target gene) levels increased
during NPC differentiation. (b) Immunofluorescent staining confirmed that both HAUSP
(red) and REST (green) protein levels declined during NPC differentiation. 15167 NPCs
Huang et al. Page 12
Nat Cell Biol
. Author manuscript; available in PMC 2011 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
were induced by RA to undergo differentiation for the indicated times, then fixed and
immunostained with anti-HAUSP and anti-REST specific antibodies. Nuclei were
counterstained with DAPI (blue). (c) Immunofluorescent staining showed that HAUSP (red)
decreased but the neuronal differentiation marker TUJ1 (green) increased during
differentiation. ENStemA NPCs (derived from a human ES cell line by Chemicon/
Millipore) were differentiated by RA treatment for the indicated times, fixed and stained
with anti-HAUSP and anti-TUJ1 specific antibodies. Nuclei were counterstained with DAPI
(blue). Uncropped images of blots are shown in Supplementary Information, Fig. S8.
Huang et al. Page 13
Nat Cell Biol
. Author manuscript; available in PMC 2011 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 2.
HAUSP knockdown reduces REST protein levels in NPCs. (a, b) Immunoblotting showed
that HAUSP knockdown by two distinct shRNAs (B2 and B5) decreased protein levels of
REST but not CoREST in ENStemA (a) and 15167 (b) NPCs. NPCs were infected with
lentiviruses expressing shHAUSP or non-targeting (NT) control shRNA for 48 hours, whole
cell lysates were harvested for immunoblotting with the specific antibodies as indicated. (c,
d) Immunofluorescent staining confirmed that HAUSP knockdown reduced REST levels in
ENStemA (c) and 17231 (d) NPCs. Cells were cultured and attached on cover glasses coated
with BD Matrigel hESC-qualified matrix, infected with lentiviruses expressing shHAUSP or
Huang et al. Page 14
Nat Cell Biol
. Author manuscript; available in PMC 2011 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
control NT shRNA, treated without (c) or with (d) puromycin to select for infected cells,
fixed and immunostained with anti-HAUSP and anti-REST specific antibodies. HAUSP was
labeled in green, and REST was labeled in red. Nuclei were counterstained with DAPI
(blue). Nuclei with reduced HAUSP and REST proteins are indicated by arrows in c. All
Puromycin-selected cells infected with lentiviruses expressing HAUSP-targeting shRNA
showed reduced HAUSP and REST protein levels in d. Lentiviral infection efficiency in
NPCs with GFP-expressing lentiviruses is shown in Supplementary Information, Fig. S7.
Uncropped images of blots are shown in Supplementary Information, Fig. S8.
Huang et al. Page 15
Nat Cell Biol
. Author manuscript; available in PMC 2011 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 3.
HAUSP knockdown promotes neural differentiation and decreases NPC self-renewal, and
REST overexpression rescues the differentiation phenotype induced by HAUSP knockdown.
(a) Targeting HAUSP with shRNA promotes neuronal differentiation. 15167 NPCs were
infected with lentiviruses expressing shHAUSP (B5 clone) or non-targeting (NT) shRNA for
126 hours and immuno-stained for Nestin (an NPC maker, in red) and TUJ1 (a neuronal
differentiation marker, in green). (b) Quantified data from a confirmed that HAUSP
knockdown increased neuronal lineage specification. The fraction of cells expressing TUJ1
(green) significantly (p < 0.001) increased and the fraction of cells expressing Nestin (red)
Huang et al. Page 16
Nat Cell Biol
. Author manuscript; available in PMC 2011 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
decreased after HAUSP knockdown in the NPCs. Data are presented as means ± s.d. [n = 3
(200 cells/experiment)]. (c) Immunofluorescent staining showed that ectopic REST
expression rescued the differentiation phenotype induced by HAUSP knockdown. 17231
NPCs were transfected with Flag-REST or vector control, and infected with shHAUSP (B5
clone) or NT shRNA lentiviruses for 126 hours and immuno-stained for Nestin (red) and
TUJ1 (green). (d) Quantified data from c indicated that ectopic expression of REST
significantly (p < 0.001) attenuated the increased fraction of cells expressing TUJ1 induced
by HAUSP knockdown. Data are presented as means ± s.d. [n = 3 (200 cells/experiment)].
(e) Immunoblotting confirmed that ectopic expression of REST repressed the TUJ1
expression induced by HAUSP knockdown. 17231 NPCs were transfected with Flag-REST
or vector, and infected with shHAUSP (B5 clone) or NT shRNA lentiviruses for 96 hours,
and immunoblotted with specific antibodies against HAUSP, Flag, TUJ1 and α-Tubulin. (f)
Neurosphere formation assay showed that HAUSP knockdown reduced NPC self-renewal
potential. 15167 NPCs were infected with shHAUSP or NT shRNA lentiviruses and allowed
to form neurospheres in serum-free suspension culture. HAUSP knockdown reduced the
neurosphere size and induced the attachment of neurosphere on the uncoated dishes. (g)
Quantified data from f confirmed that HAUSP knockdown with two specific shRNAs (B2
and B5) significantly (p < 0.001) decreased the number of neurosphere formation of 15167
NPCs. Data are means ± s.d. (n = 3). Uncropped images of blots are shown in
Supplementary Information, Fig. S8.
Huang et al. Page 17
Nat Cell Biol
. Author manuscript; available in PMC 2011 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 4.
Real-time PCR (RT-PCR) analysis indicated that reduced HAUSP expression by shRNA did
not significantly alter REST mRNA expression but increased TUJ1 (a REST target gene)
mRNA levels. ENStemA NPCs were targeted with shHAUSP (B5 clone) or control non-
targeting (NT) shRNA for 72 hours through lentiviral infection. RNA samples were
prepared for RT-PCR analysis with specific primers for HAUSP, REST, Co-REST and
TUJ1. HAUSP mRNA was significantly down-regulated but REST mRNA levels were not
significantly affected. Data are presented as means ± s.d. (n = 3).
Huang et al. Page 18
Nat Cell Biol
. Author manuscript; available in PMC 2011 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 5.
HAUSP mediates REST deubiquitination to regulate REST protein levels. (a, b)
Immunoprecipitation (IP) showed that HAUSP and REST interact in NPCs. Cell lysates of
ENStemA NPCs were immunoprecipitated with anti-REST (mAb) or anti-HAUSP antibody
or IgG control and then immunoblotted with anti-HAUSP and anti-REST (rabbit polyclonal)
antibodies. (c) Ubiquitination assays showed that HAUSP knockdown increased REST
ubiquitination in NPCs. 17231 NPCs were infected with lentiviruses expressing shHAUSP
or non-targeting (NT) shRNA for 48 hours and then treated with the proteasome inhibitor
MG132 for 6 hours before harvest for IP. Cell lysates were immunoprecipitated with an anti-
Huang et al. Page 19
Nat Cell Biol
. Author manuscript; available in PMC 2011 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
REST or anti-Ubiquitin specific antibody or control IgG, and then immunobloted with anti-
Ubiquitin or anti-REST specific antibody. Both IP with anti-REST antibody and the
reciprocal IP with anti-Ubiquitin antibody confirmed that HAUSP knockdown increased
REST poly-ubiquitination. (d) Ectopic expression of wild-type HAUSP (Wt-HAUSP) but
not the catalytic dead mutant (223CS) HAUSP (Mt-HAUSP) reduced REST
ubiquitination. 17231 NPCs were transfected with the Flag-tagged Wt-HAUSP (Wt), Mt-
HAUSP (Mt) or vector (V) control through lentiviral infection for 36 hours, then treated
with the proteasome inhibitor MG132 for 6 hours, and subjected for analysis of REST
ubiquitination. Ectopic expression of Wt-HAUSP but not Mt-HAUSP reduced REST
ubiquitination in the NPCs. (e) In vitro deubquitination assay showed that the β-TrCP-
mediated REST ubiquitination was specifically inhibited by Wt-HAUSP (lane 3) but not by
the Mt-HAUSP (a catalytic dead mutant, lane 4) or control deubiquitinase USP1 (lane 5).
Flag-Wt-HAUSP, Flag-Mt-HAUSP, Myc-REST, HA-β-TrCP and Flag-USP1 were
individually overexpressed in 293T cells, and then purified with the specific antibody or the
corresponding tag antibody for this assay. (f) In vivo deubiquitination assay confirmed that
the β-TrCP-mediated REST ubiquitination was specifically attenuated by the Wt-HAUSP
but not the catalytic dead Mt-HAUSP. 293T cells were transfected with the indicated sets of
plasmids for 48 hours, treated with the proteasome inhibitor MG132 for 6 hours and then
subjected for analysis of REST ubiquitination in the samples expressing different set of
proteins as indicated. Uncropped images of blots are shown in Supplementary Information,
Fig. S8.
Huang et al. Page 20
Nat Cell Biol
. Author manuscript; available in PMC 2011 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 6.
Neuronal differentiation induced by knockdown of endogenous HAUSP was rescued by
ectopic expression of wild-type HAUSP but not the catalytic dead HAUSP mutant. As the
B5 shHAUSP clone targets 3’-end non-coding region of endogenous HAUSP mRNA,
ectopic expression of the wild-type and mutant HAUSP that lack the 3’-end non-coding
region was not affected by the B5 HAUSP shRNA. (a) Immunofluorescent staining showed
that ectopic expression of wild-type HAUSP (Wt-HAUSP) but not the catalytic dead mutant
HAUSP (Mt-HAUSP) rescued the neuronal differentiation phenotype induced by
knockdown of endogenous HAUSP. 17231 NPCs were transfected with Flag-tagged Wt-
Huang et al. Page 21
Nat Cell Biol
. Author manuscript; available in PMC 2011 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
HAUSP or Mt-HAUSP, or vector control through lentiviral infection, and infected with
lentiviruses expressing shHAUSP (B5 clone) or control non-targeting (NT) shRNA for 96
hours and immuno-stained with specific antibodies against Nestin (red) and TUJ1 (green).
Ectopic expression of Wt-HAUSP but not the Mt-HAUSP suppressed the expression of
neuronal marker TUJ1 that was induced by knockdown of endogenous HAUSP. (b)
Quantified data from a indicated that ectopic expression of the Wt-HAUSP but not the Mt-
HAUSP in NPCs almost fully rescued the differentiation phenotype induced by knockdown
of the endogenous HAUSP. Data are means ± s.d. [n = 4 (200 cells/experiment)]. (c)
Immunoblotting validated that ectopic expression of Wt-HAUSP but not the Mt-HAUSP
resulted in suppression of TUJ1 expression. 17231 NPCs were transfected with Flag-tagged
Wt-HAUSP or Mt-HAUSP, or vector control through lentiviral infection, and infected with
lentiviruses expressing shHAUSP (B5 clone) or NT shRNA for 72 hours and immuno-
stained with specific antibodies against REST, Flag, TUJ1 and α-Tubulin (loading control).
(d) Deubiquitination assay showed that ectopic expression of Wt-HAUSP but not the
catalytic dead Mt-HAUSP attenuated the REST ubiquitination induced by knockdown of
endogenous HAUSP. 15167 NPCs were transfected with Flag-tagged Wt-HAUSP or Mt-
HAUSP, or vector control through lentiviral infection, and infected with lentiviruses
expressing shHAUSP (B5 clone) or NT shRNA for 48 hours, treated with the proteasome
inhibitor MG132 for 6 hours, immunoprecipitated (IP) with anti-REST specific antibody or
the IgG control, and immunobloted with anti-Ubiquitin specific antibody. Uncropped
images of blots are shown in Supplementary Information, Fig. S8.
Huang et al. Page 22
Nat Cell Biol
. Author manuscript; available in PMC 2011 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 7.
A consensus site of human REST (310-PYSS-313) is required for the HAUSP-mediated
REST deubquitination and HAUSP counteracts β-TrCP-mediated REST ubiquitination. (a)
Diagram of human REST protein. A consensus sequence (310-PYSS-313) of human REST
required for the HAUSP-mediated REST deubiquitination and a critical mutation (S313A)
on this site that disrupts the deubiquitination are shown. (b) In vivo deubiquitination assay
showed that a critical mutation (S313A) on the consensus sequence (310-PYSS-313) of
human REST attenuated the HAUSP-mediated REST deubiquitination (see lanes 2–4),
while a similar mutation (S1042A) on another potential site (1039-PQES-1042) did not alter
the HAUSP-mediated REST deubiquitination (see lane 3–5). S313A/S1042A double
mutations (Flag-REST-AA) and S313A single mutation showed similar effect on the
HAUSP-mediated REST deubiquitination (see lane 4–6). 293T cells were transfected with
the indicated set of expression plasmids, and treated with the proteasome inhibitor MG132
Huang et al. Page 23
Nat Cell Biol
. Author manuscript; available in PMC 2011 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
for 6 hours, and then subjected for analysis of REST ubiquitination. (c) REST ubiquitination
increased during neuronal differentiation. 17231 NPCs differentiation was induced by all-
trans retinoic acid (RA) for four days, treated with MG132 for 6 hours, and harvested for a
ubiquitination assay. Cell lysates from NPCs or differentiated cells were subjected to
immunoprecipitation with anti-REST antibody (Rabbit) and immunoblotted with anti-
Ubiquitin and the anti-REST antibodies (mAb). (d) Double knockdown analysis confirmed
that REST protein is controlled by both β-TrCP-mediated ubiquitination and the HAUSP-
mediated deubiquitination in NPCs. 15167 NPCs were treated with RA for only 24 hours to
initiate differentiation, and transduced with shHAUSP, shβ-TrCP, both shHAUSP and shβ-
TrCP, or non-targeting (NT) shRNA for 36 hours through lentiviral infection, treated with
MG132 for 6 hours, and harvested for ubiquitination assessment. HAUSP knockdown alone
increased REST ubiquitination (lanes 1, 2), while β-TrCP knockdown alone reduced REST
ubiqutination (lanes 1, 3). However, HAUSP and β-TrCP double knockdown restored REST
ubiquitination that was reduced by β-TrCP knockdown and abolishes REST ubiquitination
induced by HAUSP knockdown (lanes 1–4). Uncropped images of blots are shown in
Supplementary Information, Fig. S8.
Huang et al. Page 24
Nat Cell Biol
. Author manuscript; available in PMC 2011 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 8.
“Ying-Yang” control model of REST protein at the post-translational level. Both HAUSP-
mediated deubiquitination (“Ying”) and β-TrCP-mediated ubiquitination (“Yang”) regulate
REST protein levels in NPCs. HAUSP deubiquitinase stabilizes REST protein to promote
NPC maintenance. In contrast, the β-TrCP E3 ubiquitin ligase mediates REST ubiquitination
and degradation to promote neuronal differentiation. The net balance between the β-TrCP-
mediated ubiquitination and the HAUSP-mediated deubiquitination controls REST protein
levels and determine cellular fate.
Huang et al. Page 25
Nat Cell Biol
. Author manuscript; available in PMC 2011 August 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript

Supplementary resource (1)

... REST binds to RE-1, a highly conserved 21-23 bp DNA sequence located in several neuronal genes, and causes silencing of the transcription of target genes by recruiting specific co-repressors [7]. During neural differentiation, REST expression decreases rapidly and is maintained at low levels in mature neurons [8,9]. The aberrant expression of the REST protein has been associated with a variety of tumors and neurodegenerative disorders [10]. ...
... Several reports suggest that REST undergo posttranslational regulation by a ubiquitination system to maintain cellular proteostasis [9,12,14]. During differentiation of neural precursor cells, the REST protein level is decreased due to rapid degradation through the proteasomal degradation pathway by the E3 ubiquitin ligase complex SCF-β-TrCP [9,14]. ...
... Several reports suggest that REST undergo posttranslational regulation by a ubiquitination system to maintain cellular proteostasis [9,12,14]. During differentiation of neural precursor cells, the REST protein level is decreased due to rapid degradation through the proteasomal degradation pathway by the E3 ubiquitin ligase complex SCF-β-TrCP [9,14]. In contrast, the SCFβ-TrCP E3 ligase expression is increased in differentiated neuroblastoma tumors, indicating a negative correlation between the expression of REST and SCF-β-TrCP E3 ligase [9,12]. ...
Article
Full-text available
Background: The repressor element-1 silencing transcription factor (REST), a master transcriptional repressor, is essential for maintenance, self-renewal, and differentiation in neuroblastoma. An elevated expression of REST is associated with impaired neuronal differentiation, which results in aggressive neuroblastoma formation. E3 ligases are known to regulate REST protein abundance through the 26 S proteasomal degradation pathway in neuroblastoma. However, deubiquitinating enzymes (DUBs), which counteract the function of E3 ligase-mediated REST protein degradation and their impact on neuroblastoma tumorigenesis have remained unexplored. Methods: We employed a CRISPR/Cas9 system to perform a genome-wide knockout of ubiquitin-specific proteases (USPs) and used western blot analysis to screen for DUBs that regulate REST protein abundance. The interaction between USP3 and REST was confirmed by immunoprecipitation and Duolink in situ proximity assays. The deubiquitinating effect of USP3 on REST protein degradation, half-life, and neuronal differentiation was validated by immunoprecipitation, in vitro deubiquitination, protein-turnover, and immunostaining assays. The correlation between USP3 and REST expression was assessed using patient neuroblastoma datasets. The USP3 gene knockout in neuroblastoma cells was performed using CRISPR/Cas9, and the clinical relevance of USP3 regulating REST-mediated neuroblastoma tumorigenesis was confirmed by in vitro and in vivo oncogenic experiments. Results: We identified a deubiquitinase USP3 that interacts with, stabilizes, and increases the half-life of REST protein by counteracting its ubiquitination in neuroblastoma. An in silico analysis showed a correlation between USP3 and REST in multiple neuroblastoma cell lines and identified USP3 as a prognostic marker for overall survival in neuroblastoma patients. Silencing of USP3 led to a decreased self-renewal capacity and promoted retinoic acid-induced differentiation in neuroblastoma. A loss of USP3 led to attenuation of REST-mediated neuroblastoma tumorigenesis in a mouse xenograft model. Conclusion: The findings of this study indicate that USP3 is a critical factor that blocks neuronal differentiation, which can lead to neuroblastoma. We envision that targeting USP3 in neuroblastoma tumors might provide an effective therapeutic differentiation strategy for improved survival rates of neuroblastoma patients.
... The identified REST gene regulatory networks and biological functions agree with observations of a key role of REST as a repressor of neuronal genes in non-neuronal cells. REST depletion promotes neuronal differentiation [56], while REST stabilization promotes maintenance of NPCs [57]. However, ablation of REST expression to 1% of wild type levels appeared to impede the development of NSCs, NPCs and neurons [58]. ...
Article
Full-text available
The RE1-silencing transcription factor (REST) acts either as a repressor or activator of transcription depending on the genomic and cellular context. REST is a key player in brain cell differentiation by inducing chromatin modifications, including DNA methylation, in a proximity of its binding sites. Its dysfunction may contribute to oncogenesis. Mutations in IDH1/2 significantly change the epigenome contributing to blockade of cell differentiation and glioma development. We aimed at defining how REST modulates gene activation and repression in the context of the IDH mutation-related phenotype in gliomas. We studied the effects of REST knockdown, genome wide occurrence of REST binding sites, and DNA methylation of REST motifs in IDH wild type and IDH mutant gliomas. We found that REST target genes, REST binding patterns, and TF motif occurrence proximal to REST binding sites differed in IDH wild-type and mutant gliomas. Among differentially expressed REST targets were genes involved in glial cell differentiation and extracellular matrix organization, some of which were differentially methylated at promoters or gene bodies. REST knockdown differently impacted invasion of the parental or IDH1 mutant glioma cells. The canonical REST-repressed gene targets showed significant correlation with the GBM NPC-like cellular state. Interestingly, results of REST or KAISO silencing suggested the interplay between these TFs in regulation of REST-activated and repressed targets. The identified gene regulatory networks and putative REST cooperativity with other TFs, such as KAISO, show distinct REST target regulatory networks in IDH-WT and IDH-MUT gliomas, without concomitant DNA methylation changes. We conclude that REST could be an important therapeutic target in gliomas. Supplementary Information The online version contains supplementary material available at 10.1186/s40478-024-01779-y.
... Deletion of adenine at -240, which represented the major allele in dogs, but was present at much lower frequencies in wolves and virtually absent in coyotes, was predicted to remove binding sites for REST and ZNF423, both of which regulate neuron differentiation [7]. REST is a transcriptional repressor that is expressed in neural progenitor cells, neurons of the prefrontal cortex, in hippocampal pyramidal neurons, dentate gyrus granule neurons and cerebellar Purkinje and granule neurons [21,33]. As with TRNP1, the level of expression of REST regulates the migration of radial glia during neocortical development [36]. ...
Article
Full-text available
Cortical folding is related to the functional organization of the brain. The TMF-1 regulated protein (TRNP1) regulates the expansion and folding of the mammalian cerebral cortex, a process that may have been accelerated by the domestication of dogs. The objectives of this study were to sequence the TRNP1 gene in dogs and related canid species, provide evidence of its expression in dog brain and compare the genetic variation within dogs and across the Canidae. The gene was located in silico to dog chromosome 2. The sequence was experimentally confirmed by amplifying and sequencing the TRNP1 exonic and promoter regions in 72 canids (36 purebred dogs, 20 Gray wolves and wolf-dog hybrids, 10 coyotes, 5 red foxes and 1 Gray fox). Results A partial TRNP1 transcript was isolated from several regions in the dog brain. Thirty genetic polymorphisms were found in the Canis sp. with 17 common to both dogs and wolves, and only one unique to dogs. Seven polymorphisms were observed only in coyotes. An additional 9 variants were seen in red foxes. Dogs were the least genetically diverse. Several polymorphisms in the promoter and 3'untranslated region were predicted to alter TRNP1 function by interfering with the binding of transcriptional repressors and miRNAs expressed in neural precursors. A c.259_264 deletion variant that encodes a polyalanine expansion was polymorphic in all species studied except for dogs. A stretch of 15 nucleotides that is found in other mammalian sequences (corresponding to 5 amino acids located between Pro58 and Ala59 in the putative dog protein) was absent from the TRNP1 sequences of all 5 canid species sequenced. Both of these aforementioned coding sequence variations were predicted to affect the formation of alpha helices in the disordered region of the TRNP1 protein. Conclusions Potentially functionally important polymorphisms in the TRNP1 gene are found within and across various Canis species as well as the red fox, and unique differences in protein structure have evolved and been conserved in the Canidae compared to all other mammalian species.
... Deletion of adenine at -240, which represented the major allele in dogs, but was present at much lower frequencies in wolves and virtually absent in coyotes, was predicted to remove binding sites for REST and ZNF423, both of which regulate neuron differentiation [7]. REST is a transcriptional repressor that is expressed in neural progenitor cells, neurons of the prefrontal cortex, in hippocampal pyramidal neurons, dentate gyrus granule neurons and cerebellar Purkinje and granule neurons [21,33]. As with TRNP1, the level of expression of REST regulates the migration of radial glia during neocortical development [36]. ...
Article
Cortical folding is related to the functional organization of the brain. The TMF-1 regulated protein (TRNP1) regulates the expansion and folding of the mammalian cerebral cortex, a process that may have been accelerated by the domestication of dogs. The objectives of this study were to sequence the TRNP1 gene in dogs and related canid species, provide evidence of its expression in dog brain and compare the genetic variation within dogs and across the Canidae. The gene was located in silico to dog chromosome 2. The sequence was experimentally confirmed by amplifying and sequencing the TRNP1 exonic and promoter regions in 72 canids (36 purebred dogs, 20 Gy wolves and wolf-dog hybrids, 10 coyotes, 5 red foxes and 1 Gy fox).A partial TRNP1 transcript was isolated from several regions in the dog brain. Thirty genetic polymorphisms were found in the Canis sp. with 17 common to both dogs and wolves, and only one unique to dogs. Seven polymorphisms were observed only in coyotes. An additional 9 variants were seen in red foxes. Dogs were the least genetically diverse. Several polymorphisms in the promoter and 3'untranslated region were predicted to alter TRNP1 function by interfering with the binding of transcriptional repressors and miRNAs expressed in neural precursors. A c.259_264 deletion variant that encodes a polyalanine expansion was polymorphic in all species studied except for dogs. A stretch of 15 nucleotides that is found in other mammalian sequences (corresponding to 5 amino acids located between Pro58 and Ala59 in the putative dog protein) was absent from the TRNP1 sequences of all 5 canid species sequenced. Both of these aforementioned coding sequence variations were predicted to affect the formation of alpha helices in the disordered region of the TRNP1 protein.Potentially functionally important polymorphisms in the TRNP1 gene are found within and across various Canis species as well as the red fox, and unique differences in protein structure have evolved and been conserved in the Canidae compared to all other mammalian species.
... Within the TME, the highly immunosuppressive Forkhead box protein P3 (Foxp3) + Tregs can limit the antitumor responses presented by effective T cells [53,54]. It has been demonstrated that higher levels of USP7 facilitate the growth of tumors by modifying the immunosuppressive properties of Foxp3 + Treg cells [55][56][57] . In the ex vivo T eff suppression test by van Loosdregt et al., USP7 could interact with Foxp3 in Tregs; and USP7 knockdown hindered Tregs' functions [57]. ...
Article
Full-text available
Tumors have evolved in various mechanisms to evade the immune system, hindering the antitumor immune response and facilitating tumor progression. Immunotherapy has become a potential treatment strategy specific to different cancer types by utilizing multifarious molecular mechanisms to enhance the immune response against tumors. Among these mechanisms, the ubiquitin–proteasome system (UPS) is a significant non-lysosomal pathway specific to protein degradation, regulated by deubiquitinating enzymes (DUBs) that counterbalance ubiquitin signaling. Ubiquitin-specific proteases (USPs), the largest DUB family with the strongest variety, play critical roles in modulating immune cell function, regulating immune response, and participating in antigen processing and presentation during tumor progression. According to recent studies, the expressions of some USP family members in tumor cells are involved in tumor immune escape and immune microenvironment. This review explores the potential of targeting USPs as a new approach for cancer immunotherapy, highlighting recent basic and preclinical studies investigating the applications of USP inhibitors. By providing insights into the structure and function of USPs in cancer immunity, this review aims at assisting in developing new therapeutic approaches for enhancing the immunotherapy efficacy.
... USP7 is a member of a deubiquitinating enzyme family that contains more than 90 genes 58 . Recently, USP7 was reported to be an important regulator of transcription factors 39,59,60 . Our studies indicated an accumulation of USP7 in the nucleus during the proangiogenic function of ACEI in ECs, where it binds to Sp1/Sp3 and protects Sp1/Sp3 from proteasomal degradation. ...
Article
Full-text available
Angiogenesis is a critical pathophysiological process involved in organ growth and various diseases. Transcription factors Sp1/Sp3 are necessary for fetal development and tumor growth. Sp1/Sp3 proteins were downregulated in the capillaries of the gastrocnemius in patients with critical limb ischemia samples. Endothelial-specific Sp1/Sp3 knockout reduces angiogenesis in retinal, pathological, and tumor models and induced activation of the Notch1 pathway. Further, the inactivation of VEGFR2 signaling by Notch1 contributes to the delayed angiogenesis phenotype. Mechanistically, endothelial Sp1 binds to the promoter of Notch1 and inhibits its transcription, which is enhanced by Sp3. The proangiogenic effect of ACEI is abolished in Sp1/Sp3-deletion male mice. We identify USP7 as an ACEI-activated deubiquitinating enzyme that translocated into the nucleus binding to Sp1/Sp3, which are deacetylated by HDAC1. Our findings demonstrate a central role for endothelial USP7-Sp1/Sp3-Notch1 signaling in pathophysiological angiogenesis in response to ACEI treatment.
Article
Selective degradation of cellular proteins by the ubiquitin -proteasome system is one of the key regulatory mechanisms in eukaryotic cells. Accumulating data indicate that the ubiquitin - proteasome system is involved in the regulation of fundamental processes in mammalian stem cells, including proliferation, differentiation, cell migration, aging and programmed cell death. Regulation can be carried out either by proteolytic degradation of key transcription factors and signaling pathway proteins, or by posttranslational modifications of target proteins with ubiquitin or other ubiquitin-like modifiers. Studies of the molecular mechanisms of proteostasis maintenance in stem cells are of great importance for the development of new therapeutic approaches aimed at the treatment of autoimmune and neurodegenerative diseases, cancer and other socially significant pathologies. This review covers current data on the function of the ubiquitin-proteasome system in stem cells.
Article
Embryonic stem cells (ESCs), which are derived from the undifferentiated inner cell mass of the embryo, can differentiate every cell type of the body regarding their pluripotency. Therefore, human or mouse ESCs can be used as an unlimited cell source for numerous researches or therapeutical approaches. However, pluripotency maintenance of ESCs during in vitro culture is challenging because of their endless differentiation capacity. In the current study, the effect of USP7 on pluripotency maintenance of mouse ESCs (mESCs) has been investigated with the help of cell viability assay, morphological analysis, alkaline phosphatase (ALP) staining, qPCR analysis, and Western Blotting. 600 nM P5091 application, which showed no significant toxicity in mESCs, increased the total ubiquitinated protein amount as a proof of the accomplishment of proper USP7 inhibition. Morphological analysis and ALP activity evaluation indicated that dual inhibition of GSK3 and MEK together with leukemia inhibitory factor (LIF) treatment protects the pluripotency in presence of active USP7 enzyme. Yet, inactivation of USP7 reduced the ALP activity and altered the cell morphology in each treatment group. This morphological change and decreased ALP activity refer to differentiated mESCs. These findings were supported by gene expression and protein analysis. Gene expressions and protein amounts of pluripotency related Oct4, Nanog, c-Myc, Sox2 and Klf4 transcription factors decreased significantly after USP7 inhibition. Together with this observation, a remarkable reduction in β-Catenin expression was also noticed. It was also observed that USP7 inactivation shortens the half-live of β-Catenin and GSK3β proteins. This study demonstrates that USP7 activation is crucial for proper pluripotency maintenance, which is provided through β-Catenin stabilization.
Article
Full-text available
FOXO (Forkhead box O) transcription factors are important regulators of cellular metabolism, cell-cycle progression and cell death. FOXO activity is regulated by multiple post-translational modifications, including phosphorylation, acetylation and polyubiquitination. Here, we show that FOXO becomes monoubiquitinated in response to increased cellular oxidative stress, resulting in its re-localization to the nucleus and an increase in its transcriptional activity. Deubiquitination of FOXO requires the deubiquitinating enzyme USP7/HAUSP (herpesvirus-associated ubiquitin-specific protease), which interacts with and deubiquitinates FOXO in response to oxidative stress. Oxidative stress-induced ubiquitination and deubiquitination by USP7 do not influence FOXO protein half-life. However, USP7 does negatively regulate FOXO transcriptional activity towards endogenous promoters. Our results demonstrate a novel mechanism of FOXO regulation and indicate that USP7 has an important role in regulating FOXO-mediated stress responses.
Article
Full-text available
Background: The repressor element-1 (RE1) silencing transcription factor/neuron-restrictive silencer factor (REST/NRSF) is a master transcriptional regulator that binds to numerous genomic RE1 sites where it acts as a molecular scaffold for dynamic recruitment of modulatory and epigenetic cofactors, including corepressor for element-1-silencing transcription factor (CoREST). CoREST also acts as a hub for various cofactors that play important roles in epigenetic remodeling and transcriptional regulation. While REST can recruit CoREST to its macromolecular complex, CoREST complexes also function at genomic sites independently of REST. REST and CoREST perform a broad array of context-specific functions, which include repression of neuronal differentiation genes in neural stem cells (NSCs) and other non-neuronal cells as well as promotion of neurogenesis. Despite their involvement in multiple aspects of neuronal development, REST and CoREST are not believed to have any direct modulatory roles in glial cell maturation. Methodology/principal findings: We challenged this view by performing the first study of REST and CoREST in NSC-mediated glial lineage specification and differentiation. Utilizing ChIP on chip (ChIP-chip) assays, we identified distinct but overlapping developmental stage-specific profiles for REST and CoREST target genes during astrocyte (AS) and oligodendrocyte (OL) lineage specification and OL lineage maturation and myelination, including many genes not previously implicated in glial cell biology or linked to REST and CoREST regulation. Amongst these factors are those implicated in macroglial (AS and OL) cell identity, maturation, and maintenance, such as members of key developmental signaling pathways and combinatorial transcription factor codes. Conclusions/significance: Our results imply that REST and CoREST modulate not only neuronal but also glial lineage elaboration. These factors may therefore mediate critical developmental processes including the coupling of neurogenesis and gliogenesis and neuronal-glial interactions that underlie synaptic and neural network plasticity and homeostasis in health and in specific neurological disease states.
Article
Full-text available
The molecular mechanisms that lead to the cognitive defects characteristic of Down syndrome (DS), the most frequent cause of mental retardation, have remained elusive. Here we use a transgenic DS mouse model (152F7 line) to show that DYRK1A gene dosage imbalance deregulates chromosomal clusters of genes located near neuron-restrictive silencer factor (REST/NRSF) binding sites. We found that Dyrk1a binds the SWI/SNF complex known to interact with REST/NRSF. The mutation of a REST/NRSF binding site in the promoter of the REST/NRSF target gene L1cam modifies the transcriptional effect of Dyrk1a-dosage imbalance on L1cam. Dyrk1a dosage imbalance perturbs Rest/Nrsf levels with decreased Rest/Nrsf expression in embryonic neurons and increased expression in adult neurons. Using transcriptome analysis of embryonic brain subregions of transgenic 152F7 mouse line, we identified a coordinated deregulation of multiple genes that are responsible for dendritic growth impairment present in DS. Similarly, Dyrk1a overexpression in primary mouse cortical neurons induced severe reduction of the dendritic growth and dendritic complexity. We propose that DYRK1A overexpression-related neuronal gene deregulation via disturbance of REST/NRSF levels, and the REST/NRSF-SWI/SNF chromatin remodelling complex, significantly contributes to the neural phenotypic changes that characterize DS.
Article
Full-text available
REST is a transcriptional repressor that targets a group of neuronal genes in non-neuronal cells. In embryonic stem (ES) cells, REST has been implicated in controlling the expression of transcription factor genes that are crucial for lineage determination and for maintaining ES cell potential. Here, we asked whether REST directly regulates neural-specifying genes in mouse ES cells using siRNA-mediated REST knockdown and ES cells that lack functional REST protein as a result of gene targeting. Loss of REST did not affect the expression of any of ten transcription factor genes known to promote neural commitment and did not affect the expression of several microRNAs, including miR-21, a putative REST target in ES cells. REST-deficient ES cells retained the ability to self-renew and to undergo appropriate differentiation towards mesoderm, endoderm and ectoderm lineages upon LIF withdrawal. Genome-wide expression profiling showed that genes that were deregulated in the absence of REST were preferentially expressed in the brain and highly enriched for the presence of canonical REST binding sites (RE1). Chromatin immunoprecipitation studies confirmed these genes as direct targets of REST in ES cells. Collectively, these data show that REST selectively silences a cohort of neuronal genes in ES cells.
Article
Full-text available
Author Summary Embryonic stem cells have the unique and defining property of pluripotency: the ability to differentiate into all cell types. Key transcription factors form interconnected gene regulatory networks that control pluripotency and differentiation. Recently, the transcriptional repressor RE1-silencing transcription factor (REST) was implicated in the maintenance of pluripotency. This was surprising, given that REST has long been known as an essential regulator of neurodevelopment. How does REST regulate pluripotency? Does REST have distinct cohorts of binding sites and target genes in different developmental contexts? To address these questions, we made whole-genome maps of REST binding sites in two mouse stem cell types: embryonic (ESC) and neural (NSC) stem cells. These data were compared with each other and with gene expression data from cells in which REST activity was inhibited. The target genes were almost completely distinct in the two cell types. Surprisingly, we found that REST recruitment has two approximately equal components: common sites across all cells and an ESC-specific component. These pluripotency-associated sites are enriched for particular classes of genes, including those mediating the Wnt signaling pathway, which is an essential regulator of pluripotency.
Article
Highlights ► Rest is not required for mouse ESC maintenance► Rest promotes the early differentiation of mouse ESCs► Rest connects to the Oct3/4-Sox2-Nanog core regulatory circuitry in ESCs
Article
Glioblastomas are lethal cancers characterized by florid angiogenesis promoted in part by glioma stem cells (GSCs). Because hypoxia regulates angiogenesis, we examined hypoxic responses in GSCs. We now demonstrate that hypoxia-inducible factor HIF2alpha and multiple HIF-regulated genes are preferentially expressed in GSCs in comparison to non-stem tumor cells and normal neural progenitors. In tumor specimens, HIF2alpha colocalizes with cancer stem cell markers. Targeting HIFs in GSCs inhibits self-renewal, proliferation, and survival in vitro, and attenuates tumor initiation potential of GSCs in vivo. Analysis of a molecular database reveals that HIF2A expression correlates with poor glioma patient survival. Our results demonstrate that GSCs differentially respond to hypoxia with distinct HIF induction patterns, and HIF2alpha might represent a promising target for antiglioblastoma therapies.
Article
Establishment and maintenance of the pluripotent state of ESCs is a key issue in stem cell biology and regenerative medicine, and consequently identification of transcription factors that regulate ESC pluripotency is an important goal. Singh et al. claim that the transcriptional repressor REST is such a regulator and that a 50% reduction of REST in ESCs leads to activation of a specific microRNA, miR-21, and that this subsequently results in loss of pluripotency markers and a reciprocal gain in some lineage-specific differentiation markers. In contrast, we show that, in haplodeficient Rest(+/-) ESCs, we detected no change in pluripotency markers, no precocious expression of differentiated neuronal markers and no interaction of REST with miR-21. It is vital that identification of factors that regulate pluripotency is based on robust, consistent data, and the contrast in data reported here undermines the claim by Singh et al. that REST is such a regulator.
Article
Removal of TRF2, a telomere shelterin protein, recapitulates key aspects of telomere attrition including the DNA-damage response and cell-cycle arrest [1]. Distinct from the response of proliferating cells to loss of TRF2 [2, 3], in rodent noncycling cells, TRF2 inhibition promotes differentiation and growth [4, 5]. However, the mechanism that couples telomere gene-silencing features [6-8] to differentiation programs has yet to be elucidated. Here we describe an extratelomeric function of TRF2 in the regulation of neuronal genes mediated by the interaction of TRF2 with repressor element 1-silencing transcription factor (REST), a master repressor of gene networks devoted to neuronal functions [9-12]. TRF2-REST complexes are readily detected by coimmunoprecipitation assays and are localized to aggregated PML-nuclear bodies in undifferentiated pluripotent human NTera2 stem cells. Inhibition of TRF2, either by a dominant-negative mutant or by RNA interference, dissociates TRF2-REST complexes resulting in ubiquitin-proteasomal degradation of REST. Consequentially, REST-targeted neural genes (L1CAM, beta3-tubulin, synaptophysin, and others) are derepressed, resulting in acquisition of neuronal phenotypes. Notably, selective damage to telomeres without affecting TRF2 levels causes neither REST degradation nor cell differentiation. Thus, in addition to protecting telomeres, TRF2 possesses a novel role in stabilization of REST thereby controlling neural tumor and stem cell fate.