Accepted Manuscript cIAPs promote the proteasomal degradation of mutant SOD1 linked to familial amyotrophic lateral sclerosis Jin Sun Choi, Kidae Kim, Do Hee Lee, Sayeon Cho, Jae Du Ha, Byoung Chul Park, Sunhong Kim, Sung Goo Park, Jeong-Hoon Kim PII:
S0006-291X(16)31743-0
DOI:
10.1016/j.bbrc.2016.10.065
Reference:
YBBRC 36612
To appear in:
Biochemical and Biophysical Research Communications
Received Date: 4 October 2016 Accepted Date: 19 October 2016
Please cite this article as: J.S. Choi, K. Kim, D.H. Lee, S. Cho, J.D. Ha, B.C. Park, S. Kim, S.G. Park, J.-H. Kim, cIAPs promote the proteasomal degradation of mutant SOD1 linked to familial amyotrophic lateral sclerosis, Biochemical and Biophysical Research Communications (2016), doi: 10.1016/ j.bbrc.2016.10.065. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
ACCEPTED MANUSCRIPT cIAPs promote the proteasomal degradation of mutant SOD1 linked to familial amyotrophic lateral sclerosis
RI PT
Jin Sun Choi1, Kidae Kim1,4, Do Hee Lee7, Sayeon Cho8, Jae Du Ha6, Byoung Chul Park1,4, Sunhong Kim1,5*, Sung Goo Park1,3*, and Jeong-Hoon Kim 2,3*
1
2
Personalized Genomic Medicine
SC
Disease Target Structure Research Center,
Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB),
M AN U
Daejeon 305-333, Republic of Korea 3
4
Department of Functional Genomics,
5
Department of Bio-Analytical Science,
Department of Biomolecular Science, University of Science and Technology (UST),
Daejeon 305-350, Republic of Korea 6
TE D
Bio & Drug Discovery Division, Korea Research Institute of Chemical Technology,
141 Gajeong-ro, Yuseong, Daejeon 305-600, Republic of Korea 7
8
EP
Department of Biotechnology, Seoul Women’s University, Seoul, Republic of Korea
AC C
College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
* Corresponding authors. Sunhong Kim, Disease Target structure Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon 34141, Republic of Korea. Tel: +82-42-860-4278; Fax: +82-42-860-4269; E-mail addresses:
[email protected]
1
ACCEPTED MANUSCRIPT Sung Goo Park, Disease Target structure Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon 34141, Republic of Korea. Tel: +82-42-860-4262; Fax: +82-42-860-4269; E-mail addresses:
[email protected]
RI PT
Jeong-Hoon Kim, Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon 34141, Republic of Korea, Tel: +82-42-860-4264; Fax: +82-42-860-4269; E-mail addresses:
SC
[email protected]
M AN U
Contributions
J S Choi and K D Kim: performed experiments; J S Choi, S Cho, S Kim, B C Park, S G Park, J H Kim; analyzed data; J S Choi, K D Kim, S Kim, J D Ha, S Cho, S G Park,
TE D
B C Park, J H Kim; designed experiments; J S Choi, J H Kim wrote the paper.
Competing financial interests
AC C
EP
The authors declare no competing financial interests.
2
ACCEPTED MANUSCRIPT ABSTRACT Although the ubiquitin–proteasome system is believed to play an important role in the pathogenesis of familial amyotrophic lateral sclerosis (FALS), caused by
RI PT
mutations in Cu/Zn-superoxide dismutase 1 (SOD1), the mechanism of how mutant SOD1 protein is regulated in cells is still poorly understood. Here we have demonstrated that cellular inhibitor of apoptosis proteins (cIAPs) are specifically
SC
associated with FALS-linked mutant SOD1 (mSOD1) and that this interaction promotes the ubiquitin-dependent proteasomal degradation of mutant SOD1. By
M AN U
utilizing cumate inducible SOD1 cells, we also showed that knock-down or pharmacologic depletion of cIAPs leads to H2O2 induced cytotoxicity in mSOD1 expressing cells. Altogether, our results reveal a novel role of cIAPs in FALS-
AC C
EP
TE D
associated mutant SOD1 regulation.
3
ACCEPTED MANUSCRIPT INTRODUCTION A hallmark of neurodegenerative diseases such as Parkinson’s disease, Huntington’s disease, Alzheimer’s disease (AD), and Amyotrophic lateral sclerosis (ALS) is the
RI PT
deposit of ubiquitin-positive protein aggregates in the brains of affected patients (1). ALS is an adult neurodegenerative disease caused by the selective loss of motor neurons in the cerebral cortex, brain stem, and spinal cord. It is characterized by
SC
progressive paralysis, selective neuronal death, and eventual death of the affected individuals. Although most ALS cases are sporadic with unknown causes, 10 % are
M AN U
inherited, in what is known as familial ALS (FALS). The best characterized form of FALS (approximately 20 % of cases) is the dominant mutation in the gene encoding Cu/Zn-superoxide dismutase (SOD1) (2). SOD1 is an abundant antioxidant enzyme, originally identified in the central nervous system, and which catalyzes the
TE D
conversion of superoxide to water and hydrogen peroxide (3). It has been shown that motor neuron death in mice expressing FALS-linked mutant SOD is due to a ‘‘toxic gain-of-function’’ caused by SOD1 mutations (4). At the molecular level, mutant
EP
SOD1 proteins are rapidly degraded by proteasomes, whose inhibition can induce aggresome formation and subsequent motor neuron death (5). Several E3 ligases
AC C
involved in the proteasomal degradation of mutant SOD1 have been identified. Dorfin, a RING-finger type E3 ubiquitin ligase, protects neuronal cells from the toxic effects of mutant SOD1 proteins by proteasomal degradation (6). Previously, we showed that Hsc70-interacting protein (CHIP), a co-chaperone and quality-control E3 ligase, promotes the degradation of mutant SOD1 (7). Another research group showed that Dorfin-CHIP chimeric proteins are able to degrade mutant SOD1 with high efficiency (8). To define the mechanism underlying the ubiquitin-dependent 4
ACCEPTED MANUSCRIPT degradation of mutant SOD1 and its implications in inclusion body formation, it is therefore crucial to understand how these mutant SOD1 proteins are ubiquitinated and which ubiquitination components are involved in this process.
RI PT
The inhibitor of apoptosis protein (IAP) family is an endogenous and negative regulator of apoptosis (9). IAPs are characterized by the presence of the N-terminal baculovirus IAP repeat (BIR) domain (10). Initially, IAP proteins have been shown to
SC
inhibit apoptosis by directly preventing proteolytic cleavage of caspases (10, 11). Subsequently, some IAP proteins such as cIAP1, cIAP2 and XIAP have been
M AN U
demonstrated to possess a RING finger domain which provides them with E3 ubiquitin ligase activity (12). It has been well documented that cIAP1 and cIAP2 regulate the nuclear factor-κB (NF-κB) signaling pathway by means of their E3 ligase activity. In the tumor necrosis factor alpha (TNFα)-mediated canonical NF-kB
TE D
pathways, cIAPs are recruited to TNF receptor 1- and 2-associated complexes through direct interaction with TNF-associated factors 2 (TRAF2) (13). The recruited cIAPs mono-ubiquitinate receptor interacting serine-threonine kinase 1 (RIPK1),
EP
subsequently leading to the activation of canonical NF-kB pathway (14). On the other hand, under unstimulated conditions, cIAPs together with TRAF1 and TRAF2
AC C
promote the degradation of NF-κB inducing kinase (NIK), suppressing the noncanonical NF-κB signaling target genes, including pro-apoptotic genes (15). Intriguingly, it has been shown that XIAP, a representative of IAPs, is involved in mutant SOD protein induced cell death. The activation of apoptotic caspases such as caspase-12, caspase-9, and caspase-3 have been observed in a transgenic model of ALS (16) and the forced expression of XIAP prolongs survival of ALS transgenic mice by directly inhibiting caspases (16-18). However, in spite of the 5
ACCEPTED MANUSCRIPT structural similarity between XIAP and cIAPs, the relationship between cIAPs and ALS is unknown. In the present study, we demonstrate that cIAPs selectively bind to mutant
RI PT
SOD1 proteins, and that this interaction facilitates the ubiquitination of mutant SOD1, leading to the degradation of mutant SOD1. Furthermore, we show that knock-down or pharmacologic depletion of cIAPs induces H2O2 induced cytotoxicity in mutant
SC
SOD1 expressing cells. Our findings implicate cIAPs as regulators of FALSassociated mutant SOD1 and provide an evidence that cIAPs could be therapeutic
AC C
EP
TE D
M AN U
target for FALS.
6
ACCEPTED MANUSCRIPT MATERIALS AND METHODS
Cell culture, transfections, and reagents HEK293T cells (#CRL-11268, ATCC) and HeLa cells (#CCL-2, ATCC) were
RI PT
cultured in Dulbecco’s Modified Eagle Medium (DMEM) (GIBCO) supplemented with 10% fetal bovine serum, and penicillin/streptomycin (100 U/mL) in humidified incubators at 37 °C with 5 % CO2. Transfection of HEK293T cells and HeLa cells
SC
with various expression vectors was carried out using Lipofectamine (Invitrogen) and X-tremeGENE (Roche). Cumate solution (QM100A-1) was obtained from System
M AN U
Biosciences (SBI). BV6 (S7463) was obtained from Selleckchem. CellTiter-Glo® Luminescent Cell Viability Assay (G7571) and DeadEnd™ Fluorometric TUNEL System (G3250) were purchased from Promega Corporation. Complete protease
TE D
inhibitor cocktail (13760700) was purchased from Roche.
Immunoprecipitation and western blot analysis For immunoprecipitation experiments, cells were lysed in lysis buffer (20 mM
EP
Tris–HCl [pH 7.5] containing 150 mM NaCl, 1% (v/v) IGEPAL, 10 % (w/v) glycerol, and 1 mM complete protease inhibitor cocktail). Prepared cell lysates were mixed
AC C
with anti-FLAG M2 affinity gel or anti-HA-agarose at 4 °C for 4 h with gentle agitation or were incubated for 2 h at 4 °C with appropriate antibodies with gentle rotation. Subsequently, protein G plus/protein A agarose was added to each sample and incubated at 4 °C for 2 h with rocking. The beads were collected by centrifugation, washed three times with lysis buffer, and then boiled in SDS-PAGE sample buffer for 5 min to elute the antigen complex from the affinity gel. Samples were resolved by
7
ACCEPTED MANUSCRIPT SDS-PAGE, transferred to nitrocellulose membranes (10401116, GE Healthcare Life Sciences), and analyzed by immunoblotting using appropriate antibodies. .
RI PT
Establishment of inducible cell lines PiggyBac transposon Mammalian expression vector constructs were obtained as follows. Wild type SOD1 (WT) and mutant SOD1 (G93A) were subcloned into the
SC
FLAG-tagged PB Cumate switch Transposon vector (System Biosciences). HeLa cells were transfected with PB Transposon plasmid DNA and PiggyBac Transposase
M AN U
plasmid (System Biosciences) with X-tremeGENE (Roche). To establish stable inducible cell lines, cells were split after three days at various confluences (1:60 or 1:600 dilution) and placed in media containing 1 µg/ml puromycin. After two weeks of puromycin selection, cumate solution was added (System Biosciences) to induce
siRNA knockdown
TE D
FLAG-SOD1 (WT and G93A) expression.
EP
RNA interference was carried out using specific siRNA duplexes to silence cIAP1 and cIAP2 in HEK293T or HeLa cells. Chemically synthesized siRNA
used
AC C
duplexes were purchased from Bioneer Corporation. The following sequences were for
all
experiments: cIAP1; 5′-GUCACUGCUUGUUAUGCAU-3′, cIAP2; 5′ CAGUUA CCCUCAUCUACUU-3′. Delivery of sicIAPs or scrambled control into the cells was carried out using the Neon® transfection system (Invitrogen) according to the manufacturer’s instructions; cells were then incubated for 24–48 h before analysis.
8
ACCEPTED MANUSCRIPT Cell viability assay and TUNEL assay To perform the cell viability assay, SOD1-G93A cells pretreated with siRNA or with BV6, were incubated in 1mM H2O2 for 0, 6, and 9 h. Cell viability was assessed
RI PT
using Cell TiterGlo (CTG) mixture as recommended by the supplier. The fragmented DNA of apoptotic cells was measured by TUNEL, using the DeadEnd Fluorometric TUNEL System (Promega) in accordance with the
SC
manufacturer’s instructions. In brief, SOD1-G93A cells were seeded in 35 mm glassbottom dishes (MatTek Corp., Ashland, MA, USA) with addition of cumate. The cell
M AN U
plate without cumate addition was assayed as the experimental control. After 24 h, the cells were fixed with 4 % formaldehyde in PBS for 25 min at 4 °C and then permeabilized with 0.2 % Triton X-100 for 5 min. The cells were incubated with TdT Reaction Mix for 1 h at 37 °C, washed with distilled water, and incubated for 30 min
TE D
with 2× SSC at room temperature. The stained, mounted cells were counterstained with DAPI. Fluorescence was detected using a Zeiss LSM 510 META confocal
AC C
EP
microscope (Zeiss, Thornwood, NY, USA).
9
ACCEPTED MANUSCRIPT RESULTS
cIAPs interact with mutant SOD1 As the survival of mutant SOD1 expressing mice was affected by the expression
RI PT
of XIAP, an IAP family protein, we hypothesized that cIAPs could play a role in regulation of mutant SOD1. Therefore, we tested the interaction between cIAP1 or cIAP2 and wildtype SOD1 or FALS associated mutant SOD1 (G93A) using co-
SC
immunoprecipitation experiments. To this end, either FLAG tagged wildtype SOD1 or G93A SOD1 was expressed with Myc tagged cIAP1 or cIAP2 in HEK293 cells.
M AN U
SOD1 proteins were immunoprecipitated using anti-FLAG M2 agarose and the resulting immune complexes were analyzed by immunoblotting with anti-Myc antibody. Of interest, mutant SOD1 selectively interacted with cIAP1 and cIAP2, whereas wildtype SOD1 did not interact with cIAP1, nor cIAP2 (Figure 1A and
TE D
Supplementary Figure S1A). In the reciprocal co-immunoprecipitation experiment, specific association of mutant SOD1 and cIAP1 was also observed (Figure 1B),
EP
implying that the cIAPs interact with FALS associated mutant SOD1.
cIAPs regulate the stability of mutant SOD1
AC C
cIAPs function as an E3 ubiquitin ligase, regulating the stability of numerous cellular proteins through their own RING domain (19). Direct interaction between cIAPs and mutant SOD1 prompted us to examine whether cIAPs might regulate the stability of mutant SOD1. To avoid the co-transfection artefect, we established HeLa cell lines inducibly expressing either wildtype SOD1 or G93A SOD1 using the PiggyBac cumate switch system. Upon cumate treatment, wildtype SOD1 or G93A SOD1 was expressed in the established cell lines (Figure 2A). Using these cell lines, 10
ACCEPTED MANUSCRIPT we examined SOD1 levels when sicIAP1, sicIAP2 or both was treated in the presence of cumate. Wildtype SOD1 level was not changed while G93A SOD1 level increased by treatment of sicIAPs (Figure 2B). It is of note that sicIAP2 showed
RI PT
prominent effects. Similarly, pharmacologic depletion of cIAPs by Smac mimetic BV6, a bivalent IAP antagonist compound which causes the degradation of cIAPs, also induced increased G93A SOD1 level (Figure 2C). To examine whether this effect
SC
was due to proteasomal degradation, cells were treated with the proteasome inhibitor, MG132 and sicIAPs. Upon MG132 treatment, reduced level of G93A SOD1 by
degradation of mutant SOD1.
M AN U
sicIAP1/2 was rescued (Figure 2D), suggesting that cIAPs promote the proteasomal
cIAPs facilitates the ubiquitination of mutant SOD1
In general, proteasomal degradation requires the ubiquitination of target substrates
TE D
(20). Thus, we performed in vivo ubiquitination assay to examine whether cIAPs could promote the ubiquitination of mutant SOD1. FLAG tagged wildtype SOD1 or
EP
G93A SOD1 was expressed with Myc tagged cIAP1 (Myc-cIAP1) and HA tagged ubiquitin (HA-Ub) in HEK293T cells. SOD1 proteins were immunoprecipitated using
AC C
anti-FLAG M2 agarose and the resulting immune complexes were analyzed by immunoblotting with anti-HA and anti-FLAG antibodies. In the absence of exogenous cIAP1, the ubiquitination of G93A SOD1 was marginal, whilst in the presence of exogenous cIAP1, the ubiquitination of G93A SOD1 strongly increased (Figure 3A). However, wildtype SOD1 was not ubiquitinated regardless of cIAP1 (Figure 3A). Similar results were obtained for cIAP2 (Supplementary Figure S1B). Subsequently, endogenous cIAPs were knocked down using siRNA to investigate the level of 11
ACCEPTED MANUSCRIPT mutant SOD1 ubiquitination. Poly-ubiquitinated G93A SOD1 reduced upon treatment with sicIAP1/2, compared to the scrambled control (Figure 3B), implying that cIAPs directly facilitate the ubiquitination of mutant SOD1. As the RING domain of cIAPs is
RI PT
crucial for E3 ligase function, we wondered whether the RING finger domain of cIAP1 is a prerequisite for mutant SOD1 ubiquitination. SOD1-G93A and HAubiquitin were expressed along with wildtype cIAP1 or mutant cIAP1 (H588A) which
resulting
immunocomplex
was
analyzed
by
SC
lacks E3 ligase activity (19). The SOD1 proteins were immunoprecipitated and the immunoblotting.
Although
the
M AN U
overexpression of wildtype cIAP1 augmented the ubiquitination of G93A SOD1, the overexpression of H588A mutant cIAP1 had little effect on the formation of polyubiquitin chains (Figure 3C). This result strongly indicates that RING domain of
TE D
cIAP1 is required for the ubiquitination of mutant SOD1.
Depletion of cIAPs exacerbates H2O2 induced cytotoxicity in mutant SOD1 expressing cells
EP
Previous reports have shown that overexpression of mutant SOD1 leads to cellular toxicity in cultured neuronal cells and transgenic animal models (21) and that
AC C
mutant SOD1 expressing cells exhibit a dose dependent reduction in cell viability when exposed to H2O2 induced oxidative stress (4, 22, 23). We wondered if cIAPs might contribute to mutant SOD1-mediated cytotoxicity upon H2O2 treatment. To investigate, the cell viability assay was carried out using SOD1-G93A cells, with cIAPs depleted by siRNAs or Smac mimetic. Cumate treatment or cIAP1/2 knockdown induced slight cytotoxicity (20 %~40 %), regardless of H2O2 exposure (Figure 4A). When cells were treated with both sicIAP1/2 and cumate, only 20 % of 12
ACCEPTED MANUSCRIPT the population survived following 6 h and 9 h of H2O2 exposure, indicating that cIAPs are necessary for cell survival when mutant SOD1 is overexpressed (Figure 4A). Similar results were observed in experiments with treatment of BV6. Co-treatment of
RI PT
BV6 and cumate induced significant cell death upon H2O2 exposure (Figure 4B). It is of note that treatment of sicIAP1/2 or BV6 did not affect cell viability in cumate induced wildtype SOD1 cells (Figure S2). In addition, we observed that
SC
downregulation of cIAP1/2 induced greater DNA fragmentation in mutant SOD1 expression using TUNEL assay. (Figure 4C and 4D), suggesting that cIAPs modulate
AC C
EP
TE D
M AN U
mutant SOD1-mediated cell death.
13
ACCEPTED MANUSCRIPT DISCUSSION Here we demonstrated that cIAPs selectively bind to FALS-linked mutant SOD1, resulting in its proteasomal degradation. However, this result raises the question of
RI PT
how cIAPs can discriminate between mutant and wildtype SOD1. Of interest, cIAPs possess an ubiquitin-associated (UBA) domain in their center. Therefore, we hypothesized that the UBA domain of cIAPs may be essential for this interaction. To
SC
test this, a UBA domain mutant of cIAP1 (MF/AA) was generated and was used to test the interaction with mutant SOD1. However, UBA mutant cIAP1 also bound to
M AN U
mutant SOD1, suggesting that the UBA domain was not essential for this interaction (Supplementary Figure S3). Previously, the carboxyl terminus of CHIP, a cochaperone and a quality-control E3 ligase, was found to interact with mutant SOD1 and promote mutant SOD1 degradation through direct ubiquitination of its interacting
TE D
partner, heat-shock protein 70 or heat-shock cognate 70 (Hsp/Hsc70) (7). Thus we reasoned that CHIP could mediate the interaction between cIAPs and mutant SOD1, facilitating the degradation of mutant SOD1. However, we failed to observe the
EP
specific interaction between mutant SOD1 and CHIP when mutant SOD1 and CHIP were overexpressed (data not shown). Alternatively, we speculated that the
AC C
association of cIAPs and mutant SOD1 could be mediated by heat-shock proteins, which have been known to be involved in the ubiquitination of abnormal or misfolded proteins (7). This idea is also supported by the fact that cIAP1 regulates cell differentiation through the interaction with Hsp90 (24). SOD1 is an antioxidant enzyme in the cellular antioxidant defense system, scavenging oxygen radicals. Previous studies have shown that FALS-linked mutant SOD1 malfunction, which promotes the production of oxygen radicals, leads to their 14
ACCEPTED MANUSCRIPT intracellular inclusion, which is toxic to motor neurons (25). In our study, we observed that the downregulation of cIAPs by siRNA increases both mutant SOD1 levels and H2O2 induced cell death. Likewise, the smac mimetic compounds (BV6) have been
RI PT
shown to lead to mutant SOD1 accumulation. A recent study has demonstrated that NF-κB signaling pathway is constitutively activated in ALS patients and the SOD1G93A mouse model of ALS (26). Given the fact that cIAP2 is not only a modulator in
SC
the NF-κB signaling pathway, but also a target gene of the NF-κB pathway (27), it is plausible that activated NF-κB signaling pathway induces cIAP2 transcription and
M AN U
subsequent expression, helping to remove misfolded and/or malfunctioning mutant
AC C
EP
TE D
SOD1 proteins, and preventing cell death in ALS patients or SOD1-G93A mice.
15
ACCEPTED MANUSCRIPT Acknowledgments This work was supported by the Korea Research Institute of Bioscience and Biotechnology (KRIBB), a research grant from the National Research Foundation of
AC C
EP
TE D
M AN U
SC
of Science and Technology (CAP-15-11-KRICT).
RI PT
Korea (NRF- 2011-0028172), and a research grant from National Research Council
16
ACCEPTED MANUSCRIPT FIGURE LEGENDS
Figure 1. cIAPs interact with FALS-linked mutant SOD1. (A) FLAG tagged wildtype SOD1 (F-SOD1 (WT)) or FLAG tagged G93A SOD1 (F-SOD1 (G93A)) was with
Myc
immunoprecipitated
tagged
cIAP1
(Myc-cIAP1)
in
HEK293T cells.
RI PT
expressed
The
proteins using FLAG beads were analyzed by immunoblotting
with anti-Myc antibody and anti-FLAG antibody. (B) FLAG tagged wildtype SOD1 (F-
SC
SOD1 (WT)) or FLAG tagged G93A SOD1 (F-SOD1 (G93A)) was expressed with HA tagged cIAP1 (HA-cIAP1) in HEK293T cells. The immunoprecipitated proteins using
M AN U
HA beads were analyzed by immunoblotting with anti-FLAG antibody and anti-HA antibody.
Figure 2. cIAPs promote the degradation of FALS-linked mutant SOD1. (A)
TE D
Establishment of HeLa cell lines inducibly expressing wildtype SOD1 (SOD1-WT) or G93A SOD1 (SOD1-G93A). Expression of SOD1 proteins was detected by immunoblotting following cumate treatment. (B) SOD1-WT or SOD1-G93A cells were
EP
transfected with siRNA specific for cIAP1, cIAP2 or scrambled control and the SOD1 proteins were induced by cumate. After 24 h, the cell lysates were analyzed by
AC C
immunoblotting using anti-FLAG, anti-cIAP1, anti-cIAP2, and anti-GAPDH antibodies. (C) SOD1-G93A cells were pretreated with cumate for 24 h and then cells were treated with or without Smac mimetic BV6 (1 µΜ) for an additional 7 h. The cell lysates were then analyzed by immunoblotting with anti-FLAG, anti-cIAP1, and antiGAPDH antibodies. (D) SOD1-G93A cells transfected with sicIAP1, sicIAP2 or scrambled control were treated with cumate. After 24 h, the cells were then treated with or without a proteasome inhibitor, MG132 (10 µΜ), for an additional 8 h. The cell 17
ACCEPTED MANUSCRIPT lysates were analyzed by immunoblotting using anti-FLAG, anti-cIAP1, anti-cIAP2, and anti-GAPDH antibodies.
RI PT
Figure 3. cIAPs facilitate the ubiquitination of FALS-linked mutant SOD1. (A) FLAG tagged wildtype SOD1 (F-SOD1 (WT)) or G93A mutant SOD1 (F-SOD1 (G93A)) was expressed with Myc tagged cIAP1 (Myc-cIAP1) and HA tagged
SC
ubiquitin (HA-Ub) in HEK293T cells. After 24 h, the cells were treated with a proteasome inhibitor, MG132 (10 µΜ), for an additional 8 h. The immunoprecipitated
M AN U
proteins using FLAG beads were analyzed by immunoblotting using anti-HA antibody and anti-FLAG antibody (top two panels). The expression level was determined using anti-Myc and anti-FLAG antibodies (bottom two panels). (B) HeLa cells were transfected with siRNA specific for cIAP1, cIAP2 or scrambled control for 24 h. The
TE D
next day, cells were transfected with FLAG tagged wildtype SOD1 (F-SOD1 (WT)) or mutant SOD1 (F-SOD1 (G93A)) and HA tagged ubiquitin (HA-Ub). After 24 h, the cells were treated with a proteasome inhibitor, MG132 (10 µΜ), for an additional 8 h. immunoprecipitated
proteins
using
FLAG
beads
were
analyzed
by
EP
The
immunoblotting with anti-HA antibody and anti-FLAG antibody (top two panels). The
AC C
expression level was determined using anti-cIAP1, anti-cIAP2, and anti-FLAG antibodies (bottom three panels). (C) FLAG tagged mutant SOD1 (F-SOD1 (G93A)) was expressed with Myc tagged cIAP1 (Myc-cIAP1) or Myc tagged H588A (MycH588A) and HA tagged ubiquitin (HA-Ub) in HEK293T cells. After 24 h, the cells were treated with a proteasome inhibitor, MG132 (10 µΜ), for an additional 8 h. The immunoprecipitated using FLAG beads were analyzed by immunoblotting with antiHA antibody and anti-FLAG antibody (top two panels). The expression level was 18
ACCEPTED MANUSCRIPT determined using anti-Myc and anti-FLAG antibodies (bottom two panels)
Figure 4: Depletion of cIAPs exacerbates H2O2 induced cytotoxicity in mutant
RI PT
SOD1 expressing cells. (A) SOD-G93A cells electroporated with siRNA specific for cIAP1, cIAP2 or scrambled control were pretreated with cumate for 24 h and the cells were then treated with H2O2 (1 mM) for 0, 6, and 9 h and cell viabilities were
SC
measured by the CellTiter-Glo assay. Data are presented as mean ± SEM (error bars) of three independent experiments, as indicated (*P < 0.05, **P < 0.01). (B) SOD-
M AN U
G93A cells pretreated with or without cumate for 24 h were treated with or without Smac mimetic BV6 (1 µΜ) for an additional 7 h, and cells were treated with H2O2 (1 mM) for 0, 6 and 9 h before the CellTiter-Glo assay was performed. Data are presented as mean ± SEM (error bars) of three independent experiments, as
TE D
indicated (*P < 0.05, **P < 0.01). (C) SOD-G93A cells electroporated with siRNA specific for cIAP1, cIAP2 or scrambled control were pretreated with cumate for 24 h and the cells were then treated with H2O2 (1 mM) for 3 h, and the TUNEL assay was
EP
carried out. The images of TUNEL positive cells were captured by a fluorescence microscope (400×). (D) Quantitation of the results from (C). Data are presented as
AC C
mean ± SEM (error bars) of three independent experiments, as indicated (**P < 0.01).
19
ACCEPTED MANUSCRIPT REFERENCES
1.
Alves-Rodrigues, A., Gregori, L., and Figueiredo-Pereira, M. E. (1998) Ubiquitin, cellular inclusions and their role in neurodegeneration. Trends in neurosciences 21, 516-520
2.
Brown, R. H., Jr. (1995) Superoxide dismutase in familial amyotrophic lateral sclerosis:
3.
RI PT
models for gain of function. Current opinion in neurobiology 5, 841-846
Yim, M. B., Chock, P. B., and Stadtman, E. R. (1993) Enzyme function of copper, zinc superoxide dismutase as a free radical generator. The Journal of biological chemistry 268, 4099-4105
4.
Yim, M. B., Kang, J. H., Yim, H. S., et al., (1996) A gain-of-function of an amyotrophic lateral
SC
sclerosis-associated Cu,Zn-superoxide dismutase mutant: An enhancement of free radical formation due to a decrease in Km for hydrogen peroxide. Proceedings of the National
Academy of Sciences of the United States of America 93, 5709-5714
Urushitani, M., Kurisu, J., Tsukita, K., et al., (2002) Proteasomal inhibition by misfolded
M AN U
5.
mutant superoxide dismutase 1 induces selective motor neuron death in familial amyotrophic lateral sclerosis. Journal of neurochemistry 83, 1030-1042 6.
Niwa, J., Ishigaki, S., Hishikawa, N., et al., (2002) Dorfin ubiquitylates mutant SOD1 and prevents mutant SOD1-mediated neurotoxicity. The Journal of biological chemistry 277, 36793-36798
Choi, J. S., Cho, S., Park, S. G., et al., (2004) Co-chaperone CHIP associates with mutant
TE D
7.
Cu/Zn-superoxide dismutase proteins linked to familial amyotrophic lateral sclerosis and promotes their degradation by proteasomes. Biochemical and biophysical research
communications 321, 574-583 8.
Ishigaki, S., Niwa, J., Yamada, S., et al., (2007) Dorfin-CHIP chimeric proteins potently
EP
ubiquitylate and degrade familial ALS-related mutant SOD1 proteins and reduce their cellular toxicity. Neurobiology of disease 25, 331-341 9.
Deveraux, Q. L., and Reed, J. C. (1999) IAP family proteins--suppressors of apoptosis. Genes
10.
AC C
& development 13, 239-252
Roy, N., Deveraux, Q. L., Takahashi, R., et al., (1997) The c-IAP-1 and c-IAP-2 proteins are
direct inhibitors of specific caspases. The EMBO journal 16, 6914-6925
11.
Deveraux, Q. L., Leo, E., Stennicke, H. R., et al., (1999) Cleavage of human inhibitor of
apoptosis protein XIAP results in fragments with distinct specificities for caspases. The
EMBO journal 18, 5242-5251 12.
Yang, Y. L., and Li, X. M. (2000) The IAP family: endogenous caspase inhibitors with multiple biological activities. Cell research 10, 169-177
13.
Rothe, M., Pan, M. G., Henzel, W. J., et al., (1995) The TNFR2-TRAF signaling complex contains two novel proteins related to baculoviral inhibitor of apoptosis proteins. Cell 83, 1243-1252 20
ACCEPTED MANUSCRIPT 14.
Bertrand, M. J., Milutinovic, S., Dickson, K. M., et al., (2008) cIAP1 and cIAP2 facilitate cancer cell survival by functioning as E3 ligases that promote RIP1 ubiquitination.
Molecular cell 30, 689-700 15.
Zarnegar, B. J., Wang, Y., Mahoney, D. J., et al., (2008) Noncanonical NF-kappaB activation requires coordinated assembly of a regulatory complex of the adaptors cIAP1, cIAP2, TRAF2 and TRAF3 and the kinase NIK. Nature immunology 9, 1371-1378 Inoue, H., Tsukita, K., Iwasato, T., et al., (2003) The crucial role of caspase-9 in the disease
RI PT
16.
progression of a transgenic ALS mouse model. The EMBO journal 22, 6665-6674 17.
Ishigaki, S., Liang, Y., Yamamoto, M., et al., (2002) X-Linked inhibitor of apoptosis protein is involved in mutant SOD1-mediated neuronal degeneration. Journal of neurochemistry 82, 576-584
Wootz, H., Hansson, I., Korhonen, L., et al., (2006) XIAP decreases caspase-12 cleavage and
SC
18.
calpain activity in spinal cord of ALS transgenic mice. Experimental cell research 312, 19.
M AN U
1890-1898
Yang, Y., Fang, S., Jensen, J. P., et al., (2000) Ubiquitin protein ligase activity of IAPs and their degradation in proteasomes in response to apoptotic stimuli. Science 288, 874-877
20.
Amerik, A., Swaminathan, S., Krantz, B. A., et al., (1997) In vivo disassembly of free polyubiquitin chains by yeast Ubp14 modulates rates of protein degradation by the proteasome. The EMBO journal 16, 4826-4838
21.
Mishra, A., Maheshwari, M., Chhangani, D., et al., (2013) E6-AP association promotes SOD1 1323
22.
TE D
aggresomes degradation and suppresses toxicity. Neurobiology of aging 34, 1310 e1311Shibata, N., Hirano, A., Yamamoto, T., et al., (2000) Superoxide dismutase-1 mutationrelated neurotoxicity in familial amyotrophic lateral sclerosis. Amyotrophic lateral sclerosis
and other motor neuron disorders : official publication of the World Federation of 23.
EP
Neurology, Research Group on Motor Neuron Diseases 1, 143-161 Richardson, K., Allen, S. P., Mortiboys, H., et al., (2013) The effect of SOD1 mutation on
AC C
cellular bioenergetic profile and viability in response to oxidative stress and influence of mutation-type. PloS one 8, e68256
24.
Didelot, C., Lanneau, D., Brunet, M., et al., (2008) Interaction of heat-shock protein 90 beta
isoform (HSP90 beta) with cellular inhibitor of apoptosis 1 (c-IAP1) is required for cell differentiation. Cell death and differentiation 15, 859-866
25.
Ratovitski, T., Corson, L. B., Strain, J., et al., (1999) Variation in the biochemical/biophysical properties of mutant superoxide dismutase 1 enzymes and the rate of disease progression in familial amyotrophic lateral sclerosis kindreds. Human molecular genetics 8, 1451-1460
26.
Casciati, A., Ferri, A., Cozzolino, M., et al., (2002) Oxidative modulation of nuclear factorkappaB in human cells expressing mutant fALS-typical superoxide dismutases. Journal of
neurochemistry 83, 1019-1029 21
ACCEPTED MANUSCRIPT Stehlik, C., de Martin, R., Binder, B. R., et al., (1998) Cytokine induced expression of porcine inhibitor of apoptosis protein (iap) family member is regulated by NF-kappa B.
EP
TE D
M AN U
SC
RI PT
Biochemical and biophysical research communications 243, 827-832
AC C
27.
22
ACCEPTED MANUSCRIPT
A F-SOD1 (G93A) Myc-cIAP1
IP: FLAG
- + - - + + + +
- + - - + + + +
70 kDa
α-Myc
RI PT
F-SOD1 (WT)
Lysate
25 kDa
Lysate HA-cIAP1 F-SOD1 (G93A) F-SOD1 (WT)
+ +
+ -
IP: HA
+ + -
+ +
25 kDa
+ -
+ + -
M AN U
B
SC
α-FLAG
α-FLAG
70 kDa
AC C
EP
TE D
α-HA
Figure 1
ACCEPTED MANUSCRIPT
A
B F-SOD1 (WT)
cumate
-
+
-
control sicIAP1 sicIAP2
+
25 kDa
α-FLAG
+ - - - + - + - - + +
25 kDa
α-GAPDH
35 kDa
F-SOD1 (G93A)
+ - - - + - + - - + +
RI PT
SOD1-WT SOD1-G93A
SC
70 kDa
M AN U
70 kDa
α-FLAG α-cIAP1 α-cIAP2 α-GAPDH
35 kDa
D
25 kDa
+ +
AC C
70 kDa
+
EP
1uM BV6 FLAG-G93A
TE D
C
35 kDa
MG132 control
sicIAP1/2 FLAG-G93A 25 kDa
+ +
+ +
+ + +
+ + + α-FLAG
α-FLAG 70 kDa
α-cIAP1
70 kDa
α-cIAP2
35 kDa
α-GAPDH
α-cIAP1 α-GAPDH
Figure 2
ACCEPTED MANUSCRIPT
A
B control
F-SOD1 (G93A) F-SOD1 (WT) HA-Ub
+ +
+ +
+ - + + + +
+ + +
sicIAP1/2 FLAG-WT FLAG-G93A HA-ub
(kDa)
(kDa)
+ + +
+ + +
170 130
170 130
α-HA
100
100
IP: FLAG
70
25
70
SC
IP: FLAG
+ + +
RI PT
+ -
Myc-cIAP1
+ + +
α-FLAG
M AN U
25
(kDa)
α-HA
α-FLAG
(kDa)
α-Myc
70
Lysate 25
α-FLAG
C
Myc-cIAP1 HA-ub
- - - - + - + + + + +
70
α-cIAP2
25
α-FLAG
+ + +
EP
FLAG-G93A
+ + -
α-cIAP1
TE D
Myc-H588A
Lysate
70
170 kDa 130 kDa
AC C
100 kDa
IP: FLAG
α-HA
70 kDa
25 kDa
α-FLAG 25 kDa
Lysate 70 kDa
α-FLAG α-MYC
Figure 3
ACCEPTED MANUSCRIPT
A
B G93A (1mM H2O2)
G93A (1mM H2O2)
140 **
** *
**
80
control
60
sicIAP1/2 cumate+control
40
cumate+sicIAP1/2
0
100
* **
80 60 40
DMSO BV6 cumate+DMSO cumate+BV6
0
6 Time(Hrs)
9
0
M AN U
0
6 Time(Hrs)
9
D
cumate+control cumate+sicIAP1/2
AC C
G93A (1mM H2O2) Apoptosis rate (%)
sicIAP1/2
EP
control
TE D
C
DAPI
**
20
20
TUNEL
*
ns
RI PT
100
*
ns
ns
ns
SC
*
*
*
*
Cell viability (%)
Cell viability (%)
120
*
120
120 100
**
80 60 40 20 0
Figure 4
ACCEPTED MANUSCRIPT cIAPs selectively interact with FALS-linked mutant SOD1. cIAPs-mutant SOD1 interaction promotes ubiquitination and degradation of mutant SOD1. Knock-down or pharmacologic depletion of cIAPs leads to H2O2 induced cytotoxicity in mutant SOD1 expressing cells.
AC C
EP
TE D
M AN U
SC
RI PT
Our results reveal a novel role of cIAPs in FALS-associated mutant SOD1 regulation.