CORE Metadata, citation and similar papers at core.ac.uk

Provided by Elsevier - Publisher Connector

Biochimica et Biophysica Acta 1843 (2014) 2838–2842

Contents lists available at ScienceDirect

Biochimica et Biophysica Acta

journal homepage: www.elsevier.com/locate/bbamcr

Review Ubiquitin proteasome system-mediated degradation of synaptic : An update from the postsynaptic side

Nien-Pei Tsai ⁎

Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA

article info abstract

Article history: The ubiquitin proteasome system is one of the principle mechanisms for the regulation of homeostasis in Received 5 July 2014 mammalian cells. In dynamic cellular structures such as neuronal synapses, ubiquitin proteasome system and Received in revised form 10 August 2014 protein translation provide an efficient way for cells to respond promptly to local stimulation and regulate Accepted 11 August 2014 neuroplasticity. The majority of research related to long-term plasticity has been focused on the postsynapses Available online 16 August 2014 and has shown that ubiquitination and subsequent degradation of specific proteins are involved in various

Keywords: activity-dependent plasticity events. This review summarizes recent achievements in understanding Ubiquitination ubiquitination of postsynaptic proteins and its impact on synapse plasticity and discusses the direction for Synapse advancing future research in the field. E3 ligase © 2014 Elsevier B.V. All rights reserved. Plasticity Proteasome Neuronal activity

1. Introduction the central nervous system such as ischemia or hypoxia [6,7],and neurodegenerative diseases [8–10], this review will primarily focus on Synapses and synapse plasticity play critical roles in determining ubiquitination-mediated degradation of synaptic or synapse-related the connectivity and strength of the neural circuit. Multiple layers of proteins in physiological conditions. Due to the increased number regulation provide efficient management of synaptic transmission of recent studies showing activity-dependent regulation of UPS at during neural development and in response to stimulation (activity). postsynapses, the focus of this review will be on the events that occur The regulation that takes place locally, at or near the synapses, has in postsynapses. received tremendous attention in the past decade. This is mainly because regulation at the local level provides not only a quicker 2. Proteasome and protein ubiquitination response to stimulation but also the specificity and selectivity for regu- lation of different neurons, axons, dendrites, and even synapses [1–5]. In eukaryotic cells proteasomes are protein complexes whose Local regulation can be divided into two categories based on whether function is to degrade misfolded proteins and certain other proteins it affects protein content. 1) Regulation that does not affect protein con- destined to be eliminated [11]. A small regulatory protein known as tent can occur through signaling cascades to mediate protein–protein ubiquitin is covalently linked to proteins that are to be degraded interaction, protein trafficking, and posttranslational modifications by the proteasome. This tagging reaction, which is called protein (except for ubiquitination). 2) Regulation that affects total or specific ubiquitination, is triggered by a series of enzymatic reactions (Fig. 1). protein content can occur through local protein translation or turnover. Initially, E1 ubiquitin-activating enzyme binds to a ubiquitin molecule Of these, local protein turnover has not been well studied but is emerg- and then catalyzes a reaction to link ubiquitin onto E1 through a ing as an important field. This is particularly true for proteasome- thioester linkage. Secondly, E2 ubiquitin-conjugating enzyme transfers mediated degradation, as the ubiquitin proteasome system (UPS) the ubiquitin from E1 to an active site on E2 through a transes- provides spatial and target-specific degradation, which are ideal for terification reaction. Finally, E3 ligase, which interacts with both the dynamic and heterogeneous compartments such as synapses. Although substrate and E2, catalyzes the transfer of ubiquitin from E2 to the UPS is strongly associated with many pathophysiological conditions in substrate [11]. Currently several hundred E3 ligases have been identi- fied; substantially these provide the specificity of ubiquitination for particular proteins. ⁎ Department of Molecular and Integrative Physiology, School of Molecular and Cellular Many studies have elucidated the importance of proteasomes in reg- Biology, University of Illinois at Urbana-Champaign, 407 South Goodwin Ave., Urbana, IL – 61801, USA. Tel.: +1 217 244 5620; fax: +1 217 333 1133. ulating functional and structural synapse plasticity [3,12 18]. It is not E-mail address: [email protected]. surprising that efforts to better understand the role of proteasomes

http://dx.doi.org/10.1016/j.bbamcr.2014.08.006 0167-4889/© 2014 Elsevier B.V. All rights reserved. N.-P. Tsai / Biochimica et Biophysica Acta 1843 (2014) 2838–2842 2839

regulated indirectly by ubiquitination through the Wiskott-Aldrich syndrome protein (WASP) family member WASH-dependent mecha- nism [28]. Therefore, it would be an important topic to understand whether F-actin level and polymerization at synapses can also be regulated by ubiquitination-related mechanisms as that would likely in turn affect synapse plasticity. Besides cytoskeleton proteins, scaffold proteins are another group of structural components in PSD that have the ability to affect the structure and complexity of PSD depending on their protein interaction network. Among several PSD-associated scaffold proteins regulated by UPS, PSD-95 is of particular interest because it is the best studied member of the membrane-associated guanylate kinase (MAGUK) family of PDZ domain-containing proteins [29].Thefirst study that identified PSD-95 as being poly-ubiquitinated linked it to NMDA-induced AMPA-receptor endocytosis [30]. This report also identified the ubiquitin E3 ligase specific for PSD-95 as murine double minute-2 (Mdm2), the well-known E3 ligase widely studied for its role in ubiquitinating the tumor suppressor p53 and regulating tumor- Fig. 1. The general ubiquitination pathway. The pathway starts with an ATP-dependent igenesis [31]. Additionally this study reported a potential PEST sequence activation of Ubiquitin (Ub) by an E1 ubiquitin-activating enzyme (E1). An E2 (a peptide sequence linked to protein degradation that is rich in conjugating-enzyme (E2) then catalyzes the transfer of a ubiquitin from an E1 to an E2. When a substrate is brought by an E3 ligase (E3) to a Ub-conjugated E2, the Ub is , , serine, and threonine) [32] in Mdm2-mediated transferred to the substrate on specific lysine residues. Depending on the functional ubiquitination; however, the function of this sequence in neuronal domain in E3 ligases, this step can be a direct Ub transfer from E2 to the substrate or a cells is currently unconfirmed. This is primarily because the PEST- two-step process involving a transient binding of E3s to the Ub. As this same deletion mutant of PSD-95 showed an abnormal dendritic distribution ubiquitination pathway repeats, a protein substrate can be poly-ubiquitinated. [33] and could not be used for studying the physical contact between PSD-95 and Mdm2 under physiological conditions. Despite this, the have been focused on the identification of E3 ligases and the substrates fact of PSD-95 ubiquitination is not in doubt as many follow-up studies involved in synapse plasticity, since this is the rate-limiting step during have confirmed this observation and suggested several regulatory protein ubiquitination as well as the step where specificity comes into mechanisms underlying Mdm2-mediated PSD-95 ubiquitination. play. Several studies have linked different E3 ligases to the regulation These include the action of cyclin-dependent kinase 5 (Cdk5) [34], of synapse plasticity and the associated animal behavior [19–21]. the level of lipoprotein receptor LRP1 [35], and the activation of Studies have also uncovered ubiquitinated proteins in postsynapses by activity-dependent transcription factor myocyte enhancer factor 2 both candidate-based [22] and unbiased proteomic approaches [23] (MEF2) [36]. The latter study also connected Mdm2-mediated PSD-95 (Table 1). However, because of the number of E3 ligases and all their ubiquitination to the deficiency in activity-dependent synapse elimina- possible substrates, linking the critical E3 ligase and its substrate to tion observed in the mouse model of fragile X syndrome (FXS), the most synapse plasticity is still considered an understudied subject. common genetic form of mental retardation and autism [37,38]. Another important scaffold protein known to be ubiquitinated is 3. Ubiquitination of postsynaptic scaffold proteins Homer1a. Homers are a family of scaffold proteins with 3 major isoforms (Homer1, Homer2, and Homer3), which are thought to medi- The postsynaptic density (PSD) is a complex of proteins localized in ate synapse signaling and are involved in learning and memory [39]. the postsynaptic membrane that provides functional and structural Although Homers are known to be associated with UPS [40],thus support for excitatory synaptic transmission [24]. The size, integrity, far, Homer1a is the only isoform that has been confirmed as being and content of PSD are known to affect the strength of the synaptic ubiquitinated [40–42]. Homer1a, a short splice variant of Homer1, transmission [24]. The cytoskeleton protein is one of the major groups lacks a coiled-coil domain for oligomerization [43] and has been of components identified in the PSD [25]. Among those identified, suggested to be a competitor of full-length Homers in multiple signaling the filament actin (F-actin) plays the most critical role in mediating pathways [43,44]. The ubiquitination of Homer1a occurs at an 11-amino dendritic spine morphology and function [26,27]. Although it is unclear acid region in its C-terminal end [42] and is thought to play a role in whether F-actin can be ubiquitinated, F-actin polymerization can be synapse plasticity [45]. The ubiquitination of other Homers has not yet been confirmed; however, given the number of Homers known to be associated with synapse plasticity and related behavioral tests [46,47], Table 1 it is highly possible that UPS-mediated turnover occurs with other The summary of ubiquitinated proteins in the postsynapses described in this article. Homer family members. Postsynaptic protein Protein name E3 ligase Associated Other PSD proteins reportedly regulated by UPS include Shank [22], fi category identi ed references GKAP [22,23,48], and AKAP79/109 [22]. The ubiquitination and the Scaffold protein PSD-95 Mdm2 [30,33–36] subsequent levels of Shank, GKAP, and AKAP79/109 were shown to all Homer1a [40–42] be regulated in a similar fashion by neuronal activity [22]. Treatment Shank [22] of cortical neuronal cultures with tetrodotoxin (TTX), an inhibitor GKAP Trim3 [22,23,48] fi AKAP79/109 [22] of neuronal action potential ring, reduced the ubiquitination and Receptor protein/subunit AMPAR (GluA1 subunit) Nedd4-1 [52] increased the levels of all three proteins; whereas, treatment of the AMPAR (GluA2 subunit) [53] cultures with bicuculline, an antagonist of γ-aminobutyric acid NMDAR (NR2B subunit) Mib2 [57] (GABA) receptor A, GABA(A) receptor, resulted in the reverse effect. mGluR1a Siah1a [40,58] mGluR5 Siah1a [58] However, these manipulations of neuronal activity produced completely GABA(A) [59,60] opposite effects on other scaffold proteins, such as PSD-95 and Homers Signaling molecule Arc Ube3a, Triad3a [67–70] [22,36,45]. This differential regulation suggests that the activity- CaMKII [23] induced ubiquitination of scaffold proteins might occur differently in SPAR β-TRCP [76] different regions within the PSD, such as the region closer to the 2840 N.-P. Tsai / Biochimica et Biophysica Acta 1843 (2014) 2838–2842 member where PSD-95 primarily locates and the region closer to cyto- triggering and being activated by various signaling cascades, it would plasm where Shank is concentrated [49]. This scenario will suggest an be of particular interest to understand 1) under what circumstances activity-dependent rearrangement of PSD structures within the same mGluRs are ubiquitinated and 2) what the consequences of the synapse. A real-time, imaging-based experiment to determine the ubiquitination of mGluRs are. level of these scaffold proteins in synapses during activity manipulation Another postsynaptic receptor known to be ubiquitinated is the might give us an insight into how scaffold proteins are differentially GABA(A) receptor. GABA(A) receptor is the major ligand-gated ion regulated. channel that mediates fast synaptic inhibition. A study has shown that chronic inhibition of neuronal activity increases the level of the 4. Ubiquitination of neurotransmitter receptors GABA(A) receptor ubiquitination and reduces its cell surface stability, while increasing neuronal activity results in reverse effects [59]. Along Neurotransmitter receptors at the postsynaptic cells are the main with the same logic, the trafficking of GABA(A) receptor subunit into players in regulating synaptic transmission. Multiple receptors have lysosome for degradation is also regulated by ubiquitination [60].This been identified in an unbiased screening for ubiquitinated proteins regulation occurs particularly in the intracellular domain of the γ2 [23]. Among them, glutamate receptors are of particular interest subunit. In summary, these data provide another mechanism to regu- due to their importance for synapse plasticity and their role in late synapse plasticity through ubiquitination on neurotransmitter neurodevelopmental and psychiatric disorders. Consequently, the regu- receptors. lation of glutamate receptors by ubiquitination has become a focus of study. 5. Ubiquitination of other signaling molecules One of the most well-known glutamate-gated receptors is the AMPA receptor, which mediates fast synaptic transmission. The insertion and Synapse plasticity is regulated by various signaling pathways removal of AMPA receptor subunits are two of the major mechanisms especially in neuronal activity events. Among many signaling molecules involved in synaptic strength and are considered the most well- regulated by UPS, three proteins are currently attracting much attention studied mechanism in long-term synapse plasticity [50,51]. A study to due to their importance at the postsynaptic terminals. The first is the determine if ubiquitination can mediate the functions of the AMPA activity-regulated cytoskeleton-associated protein (Arc) [61]. It is well receptor showed that the AMPA receptor subunit GluA1 is ubiquitinated known that Arc facilitates the internalization of AMPA receptors during AMPA-triggered neuronal activity [52]. The same study also [62,63]. Because of this, the level of Arc has been directly implicated in demonstrated that this ubiquitination most likely occurs at the lysine- many synapse-weakening processes, including in synaptic long-term rich region of the GluA1 C-terminus and involves the E3 ligase depression (LTD) [62,63] and synapse elimination [64].Ube3Awasthe known as the neural-precursor cell-expressed developmentally down- first ubiquitin E3 ligase identified for Arc. Back in the late 1990s, studies regulated gene 4-1 (Nedd4-1). Most importantly, this particular showed that the loss of a maternal copy of the UBE3A gene accounts for ubiquitination event regulates the endocytic properties of GluA1- over 80% of Angelman Syndrome cases, a severe neurodevelopmental containing AMPA receptors rather than leading to direct protein degra- disorder [65,66]. During the past 5 years, multiple studies proposed dation. A subsequent study revealed that promoting neuronal activity, that dysregulation of the AMPA receptor level in synapses triggered by by using either the GABA-A receptor antagonist or AMPA receptor abnormal Arc ubiquitination might lead to the symptoms seen in agonist, induces ubiquitination of the GluA2 subunit of the AMPA recep- Angelman Syndrome [67,68]. However, recent studies questioned if tor and subsequently leads to receptor endocytosis [53]. Although Ube3A is the most critical E3 ligase for Arc [69,70]. The second ubiquitin multiple studies have demonstrated ubiquitin-mediated regulation of E3 ligase identified for Arc is the RING domain ubiquitin ligase Triad3A membrane proteins in neuronal cells [54–56], the AMPA receptor is [69]. In Triad3A-knockdown cells, Arc is accumulated, and the level of the first postsynaptic receptor whose endocytosis has been shown to the surface AMPA receptor is reduced accordingly. Because of the role be regulated by ubiquitination. of Arc in activity-dependent synapse plasticity, Triad3A, therefore, Another critical glutamate-gated ion channel is the NMDA receptor. appears to act as a terminator for Arc-mediated events during neuronal Upon activation, NMDA receptor is permeable by several ions, including activity. calcium. The influx of calcium into postsynaptic cells initiates a number Another signaling protein ubiquitinated at the postsynaptic of signaling cascades that are important for synapse plasticity and terminal is Ca2+/calmodulin-dependent protein kinase II (CaMKII) memory-related processes. Therefore, much effort has been applied to [23]. CaMKII regulates synapse plasticity at postsynaptic terminals understanding the regulation of NMDA receptor functions, including through various mechanisms, including phosphorylating the subunits posttranslational modifications. Several different regulatory mecha- of AMPA and NMDA receptors [71,72], acting as a scaffold component nisms have been identified thus far. It was recently reported that at PSD [73,74], and regulating proteasome movement into dendritic the NR2B subunit of the NMDA receptor can be ubiquitinated by the spines [2,75] (discussed later). Although both the identity of the E3 E3 ligase Mind bomb-2 (Mib2) [57]. Although it is unclear if the ligase specific for CaMKII and the consequences of the ubiquitination ubiquitination of NR2B leads to receptor endocytosis, degradation, or of CaMKII are still unclear, exploring the effects of CaMKII ubiquitination other downstream signaling cascades, the same study demonstrated in synapse plasticity is an important research direction, especially given the possibility that ubiquitination can regulate the function of the the diverse roles played by CaMKII at the synapses. NMDA receptor. The final postsynaptic signaling molecule being ubiquitinated is the In addition to the ionotropic glutamate receptors, such as the AMPA spine-associated Rap guanosine triphosphatase activating protein and NMDA receptors, the metabotropic glutamate receptors (mGluRs) (SPAR) [76]. SPAR is an actin regulatory protein that is critical for are also critical for synapse plasticity, especially in the case of long- controlling dendritic spine morphology. Upon neuronal activity stimu- term depression (LTD). Among the members of two groups of mGluRs, lation, SPAR is phosphorylated by polo-like kinase 2 (Plk2) [77,78], group 1 and 2, two members of group 1 have been reported to which leads to SPAR ubiquitination and degradation, and subsequent be ubiquitinated. A study found that the ubiquitin E3 ligase Seven in synapse weakening [76,78].Theβ-Transducin Repeat Containing absentia homolog 1A (Siah1a) can ubiquitinate both mGluR1a and Protein Ligase (β-TRCP), one of the four subunits in Skp1/Cul1/F-box mGluR5, which leads to their proteasome degradation [58]. A follow- protein (SCF) complex, is identified as the E3 ligase responsible for up study indicated that such ubiquitination, especially of mGluR1a, SPAR ubiquitination. Because of the indispensable functions of SPAR in requires the long isomer Homer3 as a shuttling protein to bring regulating dendritic spine morphology, the ubiquitination of SPAR ubiquitinated mGluR1a to the proteasomes through interaction with has important implications for affecting synapse plasticity, such as the proteasome component S8 ATPase [40]. Because mGluRs are both homeostatic synapse plasticity [77–79]. N.-P. Tsai / Biochimica et Biophysica Acta 1843 (2014) 2838–2842 2841

6. Regulation of proteasome activity [10] S. Tydlacka, C.E. Wang, X. Wang, S. Li, X.J. Li, Differential activities of the ubiquitin- proteasome system in neurons versus glia may account for the preferential accumulation of misfolded proteins in neurons, J. Neurosci. 28 (2008) Proteasomes are involved in both synapse formation [13,80] and 13285–13295. synapse elimination [81] induced by neuronal activity. Consequently, in [11] M. Schmidt, D. Finley, Regulation of proteasome activity in health and disease, Biochim. Biophys. Acta 1843 (2014) 13–25. addition to the modulation of synaptic function through ubiquitination [12] A.E. Cartier, S.N. Djakovic, A. Salehi, S.M. Wilson, E. Masliah, G.N. Patrick, Regulation and degradation of specific proteins, coordination of proteasome activ- of synaptic structure by ubiquitin C-terminal hydrolase L1, J. Neurosci. 29 (2009) ity in neurons and synapses is another form of regulation. For example, 7857–7868. studies show that the levels of multiple proteasome subunits and [13] A.M. Hamilton, W.C. Oh, H. Vega-Ramirez, I.S. Stein, J.W. Hell, G.N. Patrick, K. Zito, Activity-dependent growth of new dendritic spines is regulated by the proteasome, regulatory genes are controlled by and associated with neuronal plastic- Neuron 74 (2012) 1023–1030. ity through specific transcription factors, such as Zif268 [82],aswellas [14] A.M. Hamilton, K. Zito, Breaking it down: the ubiquitin proteasome system in through signaling molecules and events, such as CaMKII [83] and neuronal morphogenesis, Neural Plast. 2013 (2013) 196848. [15] A.N. Hegde, The ubiquitin-proteasome pathway and synaptic plasticity, Learn. Mem. phosphorylation of Rpt6 [84]. Another form of regulation controls the 17 (2010) 314–327. accessibility of the proteasomes to ubiquitinated proteins in the [16] S.K. Jakawich, R.M. Neely, S.N. Djakovic, G.N. Patrick, M.A. Sutton, An essential synapses. A study has shown that in neuronal synapses the proteasome postsynaptic role for the ubiquitin proteasome system in slow homeostatic synaptic plasticity in cultured hippocampal neurons, Neuroscience 171 (2010) 1016–1031. is able to move from dendritic shafts into dendritic spines during neuro- [17] J.J. Yi, M.D. Ehlers, Ubiquitin and protein turnover in synapse function, Neuron 47 nal activity [85]. This was observed by tracking the movement of (2005) 629–632. GFP-tagged proteasome component Rpt1 [85]. Later, it was shown [18] C. Dong, S.V. Bach, K.A. Haynes, A.N. Hegde, Proteasome modulates positive and negative translational regulators in long-term synaptic plasticity, J. Neurosci. 34 that this sequestration and the subsequent activation of the proteasome (2014) 3171–3182. require auto-phosphorylation of CaMKIIalpha at Ser-286 and phosphor- [19] X. Tian, J. Li, V. Valakh, A. DiAntonio, C. Wu, Drosophila Rae1 controls the abundance ylation of Rpt6 at Ser-120 by CaMKII [75]. These studies suggest that the of the ubiquitin ligase Highwire in post-mitotic neurons, Nat. Neurosci. 14 (2011) 1267–1275. activation of the proteasomes in synapses might promote widespread [20] J.E. Pick, M. Malumbres, E. Klann, The E3 ligase APC/C-Cdh1 is required for destruction in synapses in some neuronal conditions such as long- associative fear memory and long-term potentiation in the amygdala of adult term depression (LTD) [86–88]. mice, Learn. Mem. 20 (2013) 11–20. [21] J.E. Pick, L. Wang, J.E. Mayfield, E. Klann, Neuronal expression of the ubiquitin E3 ligase APC/C-Cdh1 during development is required for long-term potentiation, 7. Conclusions behavioral flexibility, and extinction, Neurobiol. Learn. Mem. 100 (2013) 25–31. [22] M.D. Ehlers, Activity level controls postsynaptic composition and signaling via the ubiquitin-proteasome system, Nat. Neurosci. 6 (2003) 231–242. UPS plays an extremely important role in the control of cellular [23] C.H. Na, D.R. Jones, Y. Yang, X. Wang, Y. Xu, J. Peng, Synaptic protein ubiquitination homeostasis. In addition to the simple degradation of ubiquitinated pro- in rat brain revealed by antibody-based ubiquitome analysis, J. Proteome Res. 11 teins, multiple layers of regulation by UPS through various modulations (2012) 4722–4732. fi [24] S. Okabe, Molecular dynamics of the excitatory synapse, Adv. Exp. Med. Biol. 970 provide temporal and spatial control of speci c proteins. In neuronal (2012) 131–152. cells, the structure and function of individual synapses are usually [25] A. Dosemeci, A.J. Makusky, E. Jankowska-Stephens, X. Yang, D.J. Slotta, S.P. Markey, different and hence require fine-tuned, individual regulation. UPS Composition of the synaptic PSD-95 complex, Mol. Cell Proteomics 6 (2007) fi 1749–1760. provides a way to modify synapses by controlling the quantity of speci c [26] S. Halpain, A. Hipolito, L. Saffer, Regulation of F-actin stability in dendritic spines by proteins. The specificity of this regulation is provided by the hundreds of glutamate receptors and calcineurin, J. Neurosci. 18 (1998) 9835–9844. E3 ligases and their specific substrates. However, current literature in [27] P. Hotulainen, C.C. Hoogenraad, Actin in dendritic spines: connecting dynamics to – the field is still in need of a systematic approach to the identification function, J. Cell Biol. 189 (2010) 619 629. [28] Y.H.Hao,J.M.Doyle,S.Ramanathan,T.S.Gomez,D.Jia,M.Xu,Z.J.Chen,D.D.Billadeau, and analysis of the network of E3 ligases and protein ubiquitination in M.K. Rosen, P.R. Potts, Regulation of WASH-dependent actin polymerization and the central nervous system. Future efforts in such a direction will lead protein trafficking by ubiquitination, Cell 152 (2013) 1051–1064. to a broader understanding of protein ubiquitination in the central [29] C. Oliva, P. Escobedo, C. Astorga, C. Molina, J. Sierralta, Role of the MAGUK protein family in synapse formation and function, Dev. Neurobiol. 72 (2012) 57–72. nervous system and, hopefully, to facilitation of our understanding of [30] M. Colledge, E.M. Snyder, R.A. Crozier, J.A. Soderling, Y. Jin, L.K. Langeberg, H. Lu, the regulation of synapse plasticity. M.F. Bear, J.D. Scott, Ubiquitination regulates PSD-95 degradation and AMPA receptor surface expression, Neuron 40 (2003) 595–607. [31] M. Wade, Y.C. Li, G.M. Wahl, MDM2, MDMX and p53 in oncogenesis and cancer Acknowledgment therapy, Nat. Rev. Cancer 13 (2013) 83–96. [32] R.D. Meyer, S. Srinivasan, A.J. Singh, J.E. Mahoney, K.R. Gharahassanlou, N. Rahimi, PEST motif serine and tyrosine phosphorylation controls vascular endothelial This work was supported by the start-up fund provided by the Depart- growth factor receptor 2 stability and downregulation, Mol. Cell. Biol. 31 (2011) ment of Molecular and Integrative Physiology, School of Molecular and 2010–2025. Cellular Biology, University of Illinois at Urbana-Champaign. [33] S. Bhattacharyya, V. Biou, W. Xu, O. Schluter, R.C. Malenka, A critical role for PSD-95/AKAP interactions in endocytosis of synaptic AMPA receptors, Nat. Neurosci. 12 (2009) 172–181. References [34] M.J.Bianchetta,T.T.Lam,S.N.Jones,M.A.Morabito,Cyclin-dependentkinase5 regulates PSD-95 ubiquitination in neurons, J. Neurosci. 31 (2011) [1] G. Leal, D. Comprido, C.B. Duarte, BDNF-induced local protein synthesis and synaptic 12029–12035. plasticity, Neuropharmacology 76 Pt C (2014) 639–656. [35] C. Nakajima, A. Kulik, M. Frotscher, J. Herz, M. Schafer, H.H. Bock, P. May, Low [2] B. Bingol, M. Sheng, Deconstruction for reconstruction: the role of in density lipoprotein receptor-related protein 1 (LRP1) modulates N-methyl-D- neural plasticity and disease, Neuron 69 (2011) 22–32. aspartate (NMDA) receptor-dependent intracellular signaling and NMDA-induced [3] A.N. Hegde, Ubiquitin-proteasome-mediated local protein degradation and synaptic regulation of postsynaptic protein complexes, J. Biol. Chem. 288 (2013) plasticity, Prog. Neurobiol. 73 (2004) 311–357. 21909–21923. [4] L. Liu-Yesucevitz, G.J. Bassell, A.D. Gitler, A.C. Hart, E. Klann, J.D. Richter, S.T. Warren, [36] N.P. Tsai, J.R. Wilkerson, W. Guo, M.A. Maksimova, G.N. DeMartino, C.W. Cowan, B. Wolozin, Local RNA translation at the synapse and in disease, J. Neurosci. 31 K.M. Huber, Multiple autism-linked genes mediate synapse elimination via (2011) 16086–16093. proteasomal degradation of a synaptic scaffold PSD-95, Cell 151 (2012) [5] S.A. Swanger, G.J. Bassell, Dendritic protein synthesis in the normal and diseased 1581–1594. brain, Neuroscience 232C (2012) 106–127. [37] B.S. Abrahams, D.H. Geschwind, Advances in autism genetics: on the threshold of a [6] C.L. Liu, M.E. Martone, B.R. Hu, Protein ubiquitination in postsynaptic densities after new neurobiology, Nat. Rev. Genet. 9 (2008) 341–355. transient cerebral ischemia, J. Cereb. Blood Flow Metab. 24 (2004) 1219–1225. [38] R.J. Kelleher III, M.F. Bear, The autistic neuron: troubled translation? Cell 135 (2008) [7] G.E. Saraceno, R. Castilla, G.E. Barreto, J. Gonzalez, R.A. Kolliker-Frers, F. Capani, 401–406. Hippocampal dendritic spines modifications induced by perinatal asphyxia, Neural [39] L. Foa, R. Gasperini, Developmental roles for Homer: more than just a pretty scaffold, Plast. 2012 (2012) 873532. J. Neurochem. 108 (2009) 1–10. [8] L. Campello, J. Esteve-Rudd, N. Cuenca, J. Martin-Nieto, The ubiquitin-proteasome [40] K. Rezvani, K. Baalman, Y. Teng, M.P. Mee, S.P. Dawson, H. Wang, M. De Biasi, system in retinal health and disease, Mol. Neurobiol. 47 (2013) 790–810. R.J. Mayer, Proteasomal degradation of the metabotropic glutamate receptor [9] L.S. Chin, J.A. Olzmann, L. Li, Parkin-mediated ubiquitin signalling in aggresome 1alpha is mediated by Homer-3 via the proteasomal S8 ATPase: signal formation and autophagy, Biochem. Soc. Trans. 38 (2010) 144–149. transduction and synaptic transmission, J. Neurochem. 122 (2012) 24–37. 2842 N.-P. Tsai / Biochimica et Biophysica Acta 1843 (2014) 2838–2842

[41] H. Ageta, A. Kato, Y. Fukazawa, K. Inokuchi, H. Sugiyama, Effects of proteasome [65] T. Kishino, M. Lalande, J. Wagstaff, UBE3A/E6-AP mutations cause Angelman inhibitors on the synaptic localization of Vesl-1S/Homer-1a proteins, Brain Res. syndrome, Nat. Genet. 15 (1997) 70–73. Mol.BrainRes.97(2001)186–189. [66] T. Matsuura, J.S. Sutcliffe, P. Fang, R.J. Galjaard, Y.H. Jiang, C.S. Benton, J.M. Rommens, [42] H. Ageta, A. Kato, S. Hatakeyama, K. Nakayama, Y. Isojima, H. Sugiyama, Regulation A.L. Beaudet, De novo truncating mutations in E6-AP ubiquitin-protein ligase gene of the level of Vesl-1S/Homer-1a proteins by ubiquitin-proteasome proteolytic (UBE3A) in Angelman syndrome, Nat. Genet. 15 (1997) 74–77. systems, J. Biol. Chem. 276 (2001) 15893–15897. [67] C.Cao,M.S.Rioult-Pedotti,P.Migani,C.J.Yu,R.Tiwari,K.Parang,M.R.Spaller, [43] J.A.Ronesi,K.A.Collins,S.A.Hays,N.P.Tsai,W.Guo,S.G.Birnbaum,J.H.Hu, D.J. Goebel, J. Marshall, Impairment of TrkB-PSD-95 signaling in Angelman P.F.Worley,J.R.Gibson,K.M.Huber,Disrupted Homer scaffolds mediate syndrome, PLoS Biol. 11 (2013) e1001478. abnormal mGluR5 function in a mouse model of fragile X syndrome, Nat. [68] P.L. Greer, R. Hanayama, B.L. Bloodgood, A.R. Mardinly, D.M. Lipton, S.W. Flavell, Neurosci. 15 (2012) 431–440 (S431). T.K. Kim, E.C. Griffith,Z.Waldon,R.Maehr,H.L.Ploegh,S.Chowdhury,P.F. [44] G.C. Zhang, L.M. Mao, X.Y. Liu, N.K. Parelkar, A. Arora, L. Yang, M. Hains, E.E. Fibuch, Worley, J. Steen, M.E. Greenberg, The Angelman Syndrome protein Ube3A J.Q. Wang, In vivo regulation of Homer1a expression in the striatum by cocaine, regulates synapse development by ubiquitinating arc, Cell 140 (2010) 704–716. Mol. Pharmacol. 71 (2007) 1148–1158. [69] A.M. Mabb, H.S. Je, M.J. Wall, C.G. Robinson, R.S. Larsen, Y. Qiang, S.A. Correa, M.D. [45] K. Rezvani, Y. Teng, D. Shim, M. De Biasi, Nicotine regulates multiple synaptic Ehlers, Triad3A regulates synaptic strength by ubiquitination of arc, Neuron 82 proteins by inhibiting proteasomal activity, J. Neurosci. 27 (2007) 10508–10519. (2014) 1299–1316. [46] C. Li, S. Dong, H. Wang, Y. Hu, Microarray analysis of gene expression changes in the [70] S. Kuhnle, B. Mothes, K. Matentzoglu, M. Scheffner, Role of the ubiquitin ligase brains of NR2B-induced memory-enhanced mice, Neuroscience 197 (2011) E6AP/UBE3A in controlling levels of the synaptic protein Arc, Proc. Natl. Acad. Sci. 121–131. U. S. A. 110 (2013) 8888–8893. [47] D.M. Tiruchinapalli, M.G. Caron, J.D. Keene, Activity-dependent expression of [71] A.R.Halt,R.F.Dallapiazza,Y.Zhou,I.S.Stein,H.Qian,S.Juntti,S.Wojcik,N.Brose, ELAV/Hu RBPs and neuronal mRNAs in seizure and cocaine brain, J. Neurochem. A.J. Silva, J.W. Hell, CaMKII binding to GluN2B is critical during memory consolidation, 107 (2008) 1529–1543. EMBO J. 31 (2012) 1203–1216. [48] A.Y. Hung, C.C. Sung, I.L. Brito, M. Sheng, Degradation of postsynaptic scaffold GKAP [72] M. Wyszynski, J. Lin, A. Rao, E. Nigh, A.H. Beggs, A.M. Craig, M. Sheng, Competitive and regulation of dendritic spine morphology by the TRIM3 ubiquitin ligase in rat binding of alpha-actinin and calmodulin to the NMDA receptor, Nature 385 hippocampal neurons, PLoS One 5 (2010) e9842. (1997) 439–442. [49] C. Sala, V. Piech, N.R. Wilson, M. Passafaro, G. Liu, M. Sheng, Regulation of dendritic [73] H. Okuno, K. Akashi, Y. Ishii, N. Yagishita-Kyo, K. Suzuki, M. Nonaka, T. Kawashima, spine morphology and synaptic function by Shank and Homer, Neuron 31 (2001) H. Fujii, S. Takemoto-Kimura, M. Abe, R. Natsume, S. Chowdhury, K. Sakimura, 115–130. P.F. Worley, H. Bito, Inverse synaptic tagging of inactive synapses via dynamic [50] H.W. Kessels, R. Malinow, Synaptic AMPA receptor plasticity and behavior, Neuron interaction of Arc/Arg3.1 with CaMKIIbeta, Cell 149 (2012) 886–898. 61 (2009) 340–350. [74] A. Sanz-Clemente, J.A. Gray, K.A. Ogilvie, R.A. Nicoll, K.W. Roche, Activated CaMKII [51] M.E. Wolf, Regulation of AMPA receptor trafficking in the nucleus accumbens by couples GluN2B and casein kinase 2 to control synaptic NMDA receptors, Cell Rep. dopamine and cocaine, Neurotox. Res. 18 (2010) 393–409. 3(2013)607–614. [52] L.A. Schwarz, B.J. Hall, G.N. Patrick, Activity-dependent ubiquitination of GluA1 [75] B. Bingol, C.F. Wang, D. Arnott, D. Cheng, J. Peng, M. Sheng, Autophosphorylated mediates a distinct AMPA receptor endocytosis and sorting pathway, J. Neurosci. CaMKIIalpha acts as a scaffold to recruit proteasomes to dendritic spines, Cell 140 30 (2010) 16718–16729. (2010) 567–578. [53] M.P. Lussier, Y. Nasu-Nishimura, K.W. Roche, Activity-dependent ubiquitination of [76] X.L. Ang, D.P. Seeburg, M. Sheng, J.W. Harper, Regulation of postsynaptic RapGAP the AMPA receptor subunit GluA2, J. Neurosci. 31 (2011) 3077–3081. SPAR by Polo-like kinase 2 and the SCFbeta-TRCP ubiquitin ligase in hippocampal [54] A.B. Fotia, J. Ekberg, D.J. Adams, D.I. Cook, P. Poronnik, S. Kumar, Regulation of neurons, J. Biol. Chem. 283 (2008) 29424–29432. neuronal voltage-gated sodium channels by the ubiquitin-protein ligases Nedd4 [77] D.T. Pak, S. Yang, S. Rudolph-Correia, E. Kim, M. Sheng, Regulation of dendritic spine and Nedd4-2, J. Biol. Chem. 279 (2004) 28930–28935. morphology by SPAR, a PSD-95-associated RapGAP, Neuron 31 (2001) 289–303. [55] T. Jespersen, M. Membrez, C.S. Nicolas, B. Pitard, O. Staub, S.P. Olesen, I. Baro, [78] D.P. Seeburg, M. Feliu-Mojer, J. Gaiottino, D.T. Pak, M. Sheng, Critical role of CDK5 H. Abriel, The KCNQ1 potassium channel is down-regulated by ubiquitylating and Polo-like kinase 2 in homeostatic synaptic plasticity during elevated activity, enzymes of the Nedd4/Nedd4-like family, Cardiovasc. Res. 74 (2007) 64–74. Neuron 58 (2008) 571–583. [56] G.N. Patrick, B. Bingol, H.A. Weld, E.M. Schuman, Ubiquitin-mediated proteasome [79] Y. Chen, P. Yuanxiang, T. Knopfel, U. Thomas, T. Behnisch, Hippocampal LTP triggers activity is required for agonist-induced endocytosis of GluRs, Curr. Biol. 13 (2003) proteasome-mediated SPAR degradation in CA1 neurons, Synapse 66 (2012) 2073–2081. 142–150. [57] R. Jurd, C. Thornton, J. Wang, K. Luong, K. Phamluong, V. Kharazia, S.L. Gibb, D. Ron, [80] A. DiAntonio, L. Hicke, Ubiquitin-dependent regulation of the synapse, Annu. Rev. Mind bomb-2 is an E3 ligase that ubiquitinates the N-methyl-D-aspartate receptor Neurosci. 27 (2004) 223–246. NR2B subunit in a phosphorylation-dependent manner, J. Biol. Chem. 283 (2008) [81] A. Erturk, Y. Wang, M. Sheng, Local pruning of dendrites and spines by caspase-3- 301–310. dependent and proteasome-limited mechanisms, J. Neurosci. 34 (2014) 1672–1688. [58] K. Moriyoshi, K. Iijima, H. Fujii, H. Ito, Y. Cho, S. Nakanishi, Seven in absentia [82] A.B. James, A.M. Conway, B.J. Morris, Regulation of the neuronal proteasome by homolog 1A mediates ubiquitination and degradation of group 1 metabotropic Zif268 (Egr1), J. Neurosci. 26 (2006) 1624–1634. glutamate receptors, Proc. Natl. Acad. Sci. U. S. A. 101 (2004) 8614–8619. [83] S.N. Djakovic, L.A. Schwarz, B. Barylko, G.N. DeMartino, G.N. Patrick, Regulation of [59] R.S. Saliba, G. Michels, T.C. Jacob, M.N. Pangalos, S.J. Moss, Activity-dependent the proteasome by neuronal activity and calcium/calmodulin-dependent protein ubiquitination of GABA(A) receptors regulates their accumulation at synaptic kinase II, J. Biol. Chem. 284 (2009) 26655–26665. sites, J. Neurosci. 27 (2007) 13341–13351. [84] S.N.Djakovic,E.M.Marquez-Lona,S.K.Jakawich,R.Wright,C.Chu,M.A.Sutton, [60] I.L. Arancibia-Carcamo, E.Y. Yuen, J. Muir, M.J. Lumb, G. Michels, R.S. Saliba, G.N. Patrick, Phosphorylation of Rpt6 regulates synaptic strength in hippocampal T.G. Smart, Z. Yan, J.T. Kittler, S.J. Moss, Ubiquitin-dependent lysosomal neurons, J. Neurosci. 32 (2012) 5126–5131. targeting of GABA(A) receptors regulates neuronal inhibition, Proc. Natl. [85] B. Bingol, E.M. Schuman, Activity-dependent dynamics and sequestration of Acad.Sci.U.S.A.106(2009)17552–17557. proteasomes in dendritic spines, Nature 441 (2006) 1144–1148. [61] E. Korb, S. Finkbeiner, Arc in synaptic plasticity: from gene to behavior, Trends [86] D. Fioravante, R.Y. Liu, J.H. Byrne, The ubiquitin-proteasome system is necessary for Neurosci. 34 (2011) 591–598. long-term synaptic depression in Aplysia, J. Neurosci. 28 (2008) 10245–10256. [62] M.W. Waung, K.M. Huber, Protein translation in synaptic plasticity: mGluR-LTD, [87] C.M. Forrest, L.G. Darlington, T.W. Stone, Involvement of the proteasome and Fragile X, Curr. Opin. Neurobiol. 19 (2009) 319–326. caspase activation in hippocampal long-term depression induced by the serine [63] M.W. Waung, B.E. Pfeiffer, E.D. Nosyreva, J.A. Ronesi, K.M. Huber, Rapid translation protease subtilisin, Neuroscience 231 (2013) 233–246. of Arc/Arg3.1 selectively mediates mGluR-dependent LTD through persistent [88] L. Hou, M.D. Antion, D. Hu, C.M. Spencer, R. Paylor, E. Klann, Dynamic translational increases in AMPAR endocytosis rate, Neuron 59 (2008) 84–97. and proteasomal regulation of fragile X mental retardation protein controls [64] J.R.Wilkerson,N.P.Tsai,M.A.Maksimova,H.Wu,N.P.Cabalo,K.W.Loerwald, mGluR-dependent long-term depression, Neuron 51 (2006) 441–454. J.B.Dictenberg,J.R.Gibson,K.M.Huber, A role for dendriticmGluR5-mediated local translation of Arc/Arg3.1 in MEF2-dependent synapse elimination, Cell Rep. 7 (2014) 1589–1600.