congratulations to the winners of the mark a smith prize › wp-content › uploads › ...(green)...

1
The 2018 Mark A. Smith Prize Winner CONGRATULATIONS TO for his paper Synaptic activity induces input-specific rearrangements in a targeted synaptic protein interaction network Jonathan D. Lautz The 2017 Mark A. Smith Prize Winner for his paper An isogenic blood-brain barrier model comprising brain endothelial cells, astrocytes, and neurons derived from human induced pluripotent stem cells Scott Glen Canfield CONGRATULATIONS TO 19 - RM000526 Find out more about the Prize and past Awardees onlinelibrary.wiley.com/page/ journal/14714159/homepage/ mark_a_smith_prize.htm MORE ABOUT THE PRIZE The Mark A. Smith Prize recognizes the contribution of an outstanding young scientist to an exceptional research paper published in the Journal of Neurochemistry. The Editors’ Award, instigated in 2009, was renamed Mark A. Smith prize to pay tribute to Mark’s long service to the Journal as a Handling Editor and Deputy Chief Editor. From 2019 onwards, any first author less than 8 years post PhD will qualify. Don’t miss the poster presentations! e Mark A. Smith Prize 2015 / 2016 Awardees e deubiquitinating enzyme USP46 regulates AMPA receptor ubiquitination and trafficking Yuda Huo, Natasha Khatri, Qingming Hou, James Gilbert, Guan Wang, Heng-Ye Man Department of Biology, Boston University, Boston, Massachusetts, USA Department of Pharmacology and Experimental erapeutics, Boston University School of Medicine, Boston, Massachusetts, USA Email: [email protected] Abstract Background Hypothesis References Regulation of AMPAR accumulation at synapse is the major molecular mechanism underlying the expression of synaptic plasticity, a process contributing to the execution of higher brain functions including learning and memory, and leading to neurological disorders and neurodegenera- tive disease as well ( Huganir & Nicoll, 2013). e amount of functional AMPARs can be regulated by rapid trafficking of receptors to and from the synaptic surface via vesicle-mediated membrane insertion, internaliza- tion and recycling (Malinow & Malenka, 2002; Newpher & Ehlers, 2008; Song & Huganir 2002; Wang, Gilbert, & Man, 2012). In addition to re- ceptor translocation, a change in synaptic receptor level can result from a change in AMPAR degradation and synthesis rate, or a rebalance between these two processes (A. Lin et al., 2011; Schwarz, Hall, & Patrick, 2010). Ubiquitination on certain proteins has been shown to regulate neuronal development and synaptic plasticity (Speese, Trotta, Rodesch, Aravamu- dan, & Broadie, 2003; Yi & Ehler, 2007; Zhao, Hedge, & Marin, 2003). Among membrane proteins including neurotransmitter receptors, ubi- quitination serves as a major signal to trigger endocytosis and direct the internalized protein to lysosome and/or proteasome for degradation (A. Lin et al., 2011; A. W. Lin & Man, 2013; Schwarz et al., 2010). Our previous study has shown that AMPARs are subject to Nedd4-media- ted ubiquitination, leading to a reduction in cell-surface receptor ex- pression and suppressed synaptic transmission (A. Lin et al., 2011; A. Lin & Man, 2014). Additionally, a recent study has demonstrated that all AMPAR subunits are subject to ubiquitination, which is regulated by neuronal activity (Widagdo et al. 2015). Ubiquitination is a rever- sible process mediated via the addition of ubiquitin by E3 ligases and the removal of ubiquitin meidties via deubiquitinating enzymes (DUB) (Nijman et al. 2005; Komander et al. 2009; Reyes-Turcu et al. 2009). However, the deubiquitination of AMPARs remains largely unknown. AMPAR trafficking plays a crucial role for the expression of synaptic plasticity. Our previous work has shown that AMPARs are subject to protein ubiquitination leading to receptor internalization and degra- dation. However, whether ubiquitination is the final stage of AMPAR turnover remains unclear. We hypothesize that the ubiquitin chain on AMPARs could be removed by deubiquitinase (DUB) to counterba- lance receptor endocytosis and bypass the proteasomal degradation. A B C D E F H A B C A B C A B C D E * peptide(Aβ) is a pathological hallmark of Alzheimer’s disease (AD) edthrough the sequential cleavage of amyloid precursor protein (APP) cretases. Hypoxia is a known risk factor for AD and stimulates γ-secretase; however, the underlying mechanisms remain unclear. In showedthat Dual-specificity phosphatase 26 (DUSP26) regulatesough changes in subcellular localization of the γ-secretase complex teC99 under hypoxic conditions. DUSP26 was identified as a novel gulator from a genome-wide functional screen using a cDNA rary. The phosphatase activity of DUSP26 was required for the β42generation throughγ-secretase but this regulation did not affect of the γ-secretase complex. Interestingly, DUSP26 induced the of C99 in the axons by stimulating anterograde transport of C99- es. Additionally, DUSP26 induced c-Jun N-terminal kinase (JNK) amyloid-precursor-protein (APP) processing and axonal transport of ypoxicconditions, DUSP26 expression levels were elevated together vation, and treatment with JNK inhibitor SP600125, or the DUSP26 -87877, reduced hypoxia-induced generation by diminishing king of C99 to the axons. Finally, we observed enhanced DUSP26 d JNK activation in the hippocampus of AD patients. Our results DUSP26 mediates hypoxia-induced generation through JNK ealinga new regulator of γ-secretase-mediated APP processing under tions. Dual-specificity phosphatase 26 (DUSP26) stimulates Aβ42 generation by promoting Amyloid Precursor Protein axonal transport during hypoxia Abstract Hypothesis Summary Figure 1. DUSP26 increases γ-secretase-mediated C99 (APP) processing for Aβ generation. Figure 2. Phosphatase activity of DUSP26 is required for the regulation of APP processing. Figure 3. DUSP26 activity regulates subcellular distribution of C99 by stimulating anterograde axonal transport. Figure 4. DUSP26 promotes γ-secretase-mediated APP cleavage throu activation. Figure 5. Hypoxic stress increases γ-secretase-mediated APP processin via DUSP26. Figure 6. DUSP26 expression is elevated in the hippocampus of AD pa amyloid-beta peptide (Aβ) in the brain is a hallmark of Alzheimer’s disease 2001). is generated from amyloid-β precursor protein (APP) through agebyβ- andγ-secretases (Holtzman et al. 2011). 42 is associated with AD posits of this form in the brains of AD patients (Iwatsubo et al. 1994). s highlyimplicated in AD pathogenesis, the mechanism that determines thed by γ-secretase remains unclear. Recently, studies showed that γ-secretase subcellular localization affect the42 production ratio (Kanatsu et al. 2014). ellular localization of γ-secretase and its substrate might be important in e specificdetails associated with the mechanism remain elusive. es showed that oxidative stress, including hypoxia and formation of highly species (ROS), is an AD risk factor (Smith et al. 2000, Misonou et al. 2000). elevatedby hypoxic injury resulting from cerebral ischemia or stroke (Bazan et &Gustafson 2006). Moreover, γ-secretase activity, which is a rate-limiting step n, increases under oxidative stress through c-Jun N-terminal kinase (JNK) et al. 2008). Despite the correlation between oxidative stress andgeneration, echanismthat determines how oxidative stress regulatesγ-secretase activity and remainsunknown. typhosphatase 26 (DUSP26) is an atypical phosphatase that dephosphorylates rosine and phosphoserine/phosphothreonine residues within one substrate 2009). DUSP26 may regulate cell proliferation in neuronal cells (Wang et al. l. 2008) and function as both a tumor suppressor (Vasudevan et al. 2005, Wang maet al. 2009) and an oncogene, depending upon the cellular context (Yu et al. therole of DUSP26 in non-dividing cells, such as neurons, is not understood. widefunctional screen using a cell-based assay (Han et al. 2014), we identified -secretase activator. γ-secretase activity, which is a rate-limiting step in ases under oxidative stress through c-Jun N-terminal kinase (JNK) activation 8). However, the regulatory mechanism that determines how oxidative stress etase activity and APP processing remains unknown. We hypothesize that a JNK-activated shift in the subcellular localization of γ-secretase and C99 y toaxons in neuronal cells for 42 generation during hypoxic stress. References Background The Mark A. Smith Prize 2016 Awardees Sunmin Jung, Jihoon Nah, Jonghee Han, Seon-Guk Choi, Hyunjoo Kim, Jaesang Park, Ha-Kyung Pyo and Yong-Keun JungResearch Laboratory, School of Biological Science, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 151-747, Korea; E-mail: Congratulations to the winners of the Mark A Smith Prize Join us at the Opening Ceremony to see the prize awarded to the 2017 and 2018 winners and to hear them present their work.

Upload: others

Post on 26-Jan-2021

1 views

Category:

Documents


0 download

TRANSCRIPT

  • The 2018 Mark A. Smith Prize Winner

    CONGRATULATIONS TO

    for his paper Synaptic activity induces input-specific rearrangements in a targeted

    synaptic protein interaction network

    Jonathan D. Lautz

    The 2017 Mark A. Smith Prize Winner

    for his paper An isogenic blood-brain barrier model comprising brain endothelial cells,

    astrocytes, and neurons derived from human induced pluripotent stem cells

    Scott Glen CanfieldCONGRATULATIONS TO

    19 - RM000526

    Find out more about the Prize and past Awardees

    onlinelibrary.wiley.com/page/ journal/14714159/homepage/ mark_a_smith_prize.htm

    MORE ABOUT THE PRIZEThe Mark A. Smith Prize recognizes the contribution of an outstanding young scientist to an exceptional research paper published in the Journal of Neurochemistry.

    The Editors’ Award, instigated in 2009, was renamed Mark A. Smith prize to pay tribute to Mark’s long service to the Journal as a Handling Editor and Deputy Chief Editor. From 2019 onwards, any first author less than 8 years post PhD will qualify.

    Don’t miss the poster presentations!

    The Mark A. Smith Prize 2015 / 2016 AwardeesThe deubiquitinating enzyme USP46 regulates AMPA receptor ubiquitination and traffickingYuda Huo, Natasha Khatri, Qingming Hou, James Gilbert, Guan Wang, Heng-Ye ManDepartment of Biology, Boston University, Boston, Massachusetts, USADepartment of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts, USAEmail: [email protected]

    Abstract

    Background

    Hypothesis

    Summary

    References

    Regulation of AMPAR accumulation at synapse is the major molecular mechanism underlying the expression of synaptic plasticity, a process contributing to the execution of higher brain functions including learning and memory, and leading to neurological disorders and neurodegenera-tive disease as well ( Huganir & Nicoll, 2013). The amount of functional AMPARs can be regulated by rapid trafficking of receptors to and from the synaptic surface via vesicle-mediated membrane insertion, internaliza-tion and recycling (Malinow & Malenka, 2002; Newpher & Ehlers, 2008; Song & Huganir 2002; Wang, Gilbert, & Man, 2012). In addition to re-ceptor translocation, a change in synaptic receptor level can result from a change in AMPAR degradation and synthesis rate, or a rebalance between these two processes (A. Lin et al., 2011; Schwarz, Hall, & Patrick, 2010).Ubiquitination on certain proteins has been shown to regulate neuronal development and synaptic plasticity (Speese, Trotta, Rodesch, Aravamu-dan, & Broadie, 2003; Yi & Ehler, 2007; Zhao, Hedge, & Marin, 2003). Among membrane proteins including neurotransmitter receptors, ubi-quitination serves as a major signal to trigger endocytosis and direct the internalized protein to lysosome and/or proteasome for degradation (A. Lin et al., 2011; A. W. Lin & Man, 2013; Schwarz et al., 2010). Our previous study has shown that AMPARs are subject to Nedd4-media-ted ubiquitination, leading to a reduction in cell-surface receptor ex-pression and suppressed synaptic transmission (A. Lin et al., 2011; A. Lin & Man, 2014). Additionally, a recent study has demonstrated that all AMPAR subunits are subject to ubiquitination, which is regulated by neuronal activity (Widagdo et al. 2015). Ubiquitination is a rever-sible process mediated via the addition of ubiquitin by E3 ligases and the removal of ubiquitin meidties via deubiquitinating enzymes (DUB) (Nijman et al. 2005; Komander et al. 2009; Reyes-Turcu et al. 2009). However, the deubiquitination of AMPARs remains largely unknown.

    AMPAR trafficking plays a crucial role for the expression of synaptic plasticity. Our previous work has shown that AMPARs are subject to protein ubiquitination leading to receptor internalization and degra-dation. However, whether ubiquitination is the final stage of AMPAR turnover remains unclear. We hypothesize that the ubiquitin chain on AMPARs could be removed by deubiquitinase (DUB) to counterba-lance receptor endocytosis and bypass the proteasomal degradation.

    Huganir R. L. and Nicoll R. A. (2013) AMPARs and synaptic plasticity:the last 25 years. Neuron 80, 704–717. doi: 10.1016/j.neuron.2013.10.025.Komander D., Clague M. J. and Urbe S. (2009) Breaking the chains: structure and function of the deubiquitinases. Nat. Rev. Mol. Cell Biol. 10, 550-563. doi: 10.1155/2012/892749.Lin A., Hou Q., Jarzylo L., Amato S., Gilbert J., Shang F. and Man H. Y. (2011) Nedd4-mediated AMPA recep-tor ubiquitination regulates receptor turnover and trafficking. J. Neurochem. 119, 27-39. doi:10.1111/j.1471-4159.2011.07221.x.Lin A. W. and Man H. Y. (2013) Ubiquitination of neurotransmitter receptors and postsynaptic scaffolding prote-ins. Neural. Plast. 2013, 432057. doi: 10.1155/2013/432057.Lin A. and Man H. Y. (2014) Endocytic adaptor epidermal growth factor receptor substrate 15(Eps15) is involved in the trafficking of ubiquitinated alpha-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid receptors. J. Biol. Chem. 289. 24652-24664. doi: 10.1074/jbc.M114.582114.Malinow R. and Malenka R. C. (2002) AMPA receptor trafficking and synaptic plasticity. Annu. Rev. Neurosci. 25, 103-126. doi: 10.1146/annurev. Neuro. 25.112701.142758.Newpher T. M. and Ehlers M. D. (2008) Glutamate receptor dynamics in dendritic microdomains. Neuron. 58, 472-497. doi: 10.1016/j.neuron.2008.04.030.Nijman S. M., Luna-Vargas M. P., Velds A., Brummelkamp T.R., Dirac A. M., Sixma T. K. and Bernards R. (2005) A genomic and functional inventory of deubiquitinating enzymes. Cell 123, 773-786. doi: 10.1016/j.cell.2005.11.007.Schwarz L. A., Hall B. J. and Patrick G. N. (2010) Activity-dependent ubiquitination of GluA1 mediates a dis-tinct AMPA receptor endocytosis and sorting pathway. J. Neurosci. 30, 16718-16729. doi: 10.1523/JNEUROS-CI.3686-10.2010.Song I. and Huganir R. L. (2002) Regulation of AMPA receptors during synaptic plasticity. Trends Neurosci. 25, 578-588.Speese S. D., Trotta N., Rodesch C. K., Aravamudan B. and Broadie K. (2003) The ubiquitin proteasome system actutely regulates presynaptic protein turnover and synaptic efficacy. Curr. Biol. 13, 899-910.Wang G., Gilbert J. and Man H. Y. (2012) AMPA receptor trafficking in homeostatic synaptic plasticity: functio-nal molecules and signaling cascades. Neural Plast. 2012, 825464. doi: 10.1155/2012/825364.Yi J. J. and Ehlers M. D. (2007) Emerging roles for ubiquitin and protein degradation in neuronal function. Phar-macol. Rev. 59, 14-39. doi: 10.1124/pr.59.1.4.Zhao Y., Hegde A. N. and Martin K. C. (2003) The ubiquitin proteasome system fucntions as an inhibitory cons-traint on synaptic strengthening. Curr. Biol. 13, 887-898.Reyes-Turcu F. E., Ventii K. H. and Wikinson K. D. (2009) Regulation and cellular roles of ubiquitin-specific deu-

    Alpha-Amino-3-hydroxy-5-methyl-isoxazole-4-propionic acid receptors (AMPARs) are the primary mediators for inter-neuronal communication and play a crucial role in higher brain functions including learning and me-mory. Our previous work demonstrated that AMPARs are subject to ubiquitination by the E3 ligase Nedd4, re-sulting in EPS15-mediated receptor internalization and Ubiquitin (Ub)-proteasome pathway (UPP)-dependent degradation. Protein ubiquitination is a highly dynamic and reversible process, achieved via the balance between ubiquitination and deubiquitination. However, deubiquitination of mammalian AMPARs and the responsible deubiquitinating enzymes (DUBs) remain elusive. In this study, we identify a DUB, USP46, which specifically removes ubiquitination modification from AMPARs. USP46 is enriched at the synapse and co-localizes with sy-naptic marker proteins. In heterologous cells and neurons, expression of USP46 results in a significant reduction in AMPAR ubiquitination, accompanied with a reduced rate in AMPAR degradation and an increase in AM-PAR synaptic accumulation. By contrast, knockdown of USP46 by RNAi leads to elevated AMPAR ubiquitination and a reduction in AMPAR protein amount in neurons. Consistently, mEPSC recordings show reduced synaptic strength in neurons expressing USP46-selective RNAi. These results demonstrate USP46-mediated regulation of AMPAR ubiquitination and turnover, which may play an important role in synaptic plasticity and brain function.

    PSD95

    USP46

    Merge

    GluA1

    USP46

    Merge

    P2 fr

    actio

    n

    Homo

    gena

    te Su

    pern

    atant

    Akt

    USP46

    Nedd4

    PSD-95

    GluA1

    Hipp

    ocam

    pus

    PFC

    Cere

    bellu

    m

    Who

    le br

    ain ly

    sate

    USP46

    Nedd4

    Tubulin

    DIV0

    USP46

    GluA1

    Tubulin

    DIV3

    DI

    V5

    DIV7

    DI

    V9

    DIV1

    1 DI

    V13

    DIV1

    5 DI

    V17

    DIV1

    9 DI

    V21

    DIV2

    3

    A

    B

    C D

    E

    IB: GluA1

    IB: HA (Ubi)

    IB: Tubulin (lysate)

    250

    150

    100

    GluA1+HA-Ubi

    Vecto

    r

    USP4

    6

    USP1

    2 A

    0.0

    0.2

    0.4

    0.6

    0.8

    1.0

    1.2

    Control USP46 USP12

    No

    rma

    lize

    d U

    biq

    uiti

    n

    inte

    nsi

    ty

    *

    B

    IB: HA (Ubi)

    IB: GluA2

    IB: Tubulin (lysate)

    GluA2+HA-Ubi

    Vector USP12 USP46

    250

    150

    100

    IP: GluA2

    C

    K48 K63 WT K63 K48 R63 R48

    K6 K11 K27 K29 K33

    D

    IB: HA (Ubi)

    IB: GluA1

    IB: Tubulin (lysate)

    GluA1

    Ubi

    K63

    K48

    R63

    R48

    250

    150

    100

    IP: GluA1

    E

    IB: HA (Ubi)

    IB: GluA1 IB: Tubulin (lysate)

    HA-

    Ubi

    K4

    8 K6

    3 H

    A-U

    bi

    K48

    K63

    USP46

    250

    150

    100

    IP: GluA1

    F

    0 1 3 6 9 0 1 3 6 9 GluA1

    Tubulin

    GluA1 GluA1+USP46

    CHX (hr)

    G

    0

    20

    40

    60

    80

    100

    120

    0h 1h 3h 6h 9h

    No

    rma

    lize

    d G

    lur1

    inte

    nsi

    ty

    (%)

    Cyclohexamide treatment time

    GluA1

    GluA1+USP46

    *

    H

    IP: GluA1

    IB: Myc (USP)

    USP46 USP12

    +

    +

    MG132

    IB: Ubi

    +

    +

    IB: Tubulin

    150

    100

    75

    Lys

    ate

    A

    0.0 0.2 0.4 0.6 0.8 1.0 1.2 1.4 1.6 1.8

    Vecto

    r

    USP4

    6

    USP1

    2

    Vecto

    r+MG

    132

    USP4

    6+MG

    132

    USP1

    2+MG

    132

    Norm

    aliz

    ed U

    biq

    uiti

    n in

    tensi

    ty

    *

    * *

    B

    GFP-GluA1 HA-Ubi USP46

    Input In vitro assay

    250KD

    150KD

    100KD

    IB: HA (Ubi)

    IB: GluA1 IB: Myc (USP46)

    +

    + +

    +

    +

    + + Boil

    + + +

    C

    IB: Myc

    IB: Tub

    ShUSP46 + +

    Myc-USP46 Myc-USP12

    ShUSP12

    A shUSP46 shUSP12

    +

    +

    250

    150

    100

    75

    IP: GluA1

    IB: Tubulin (Lysate)

    IB: Ubi

    B

    0.0

    0.5

    1.0

    1.5

    2.0

    2.5

    3.0

    3.5

    Ubi

    Ubi+s

    hUSP

    46

    Ubi+s

    hUSP

    12 N

    orm

    aliz

    ed U

    biqu

    itin

    inte

    nsity

    *

    C

    siSc

    r si

    USP

    46

    siU

    SP12

    Surf GluA1 GFP Merge A

    0.0

    0.2

    0.4

    0.6

    0.8

    1.0

    1.2

    Surfa

    ce G

    luA1

    siScram siUSP46 siUSP12

    **

    B

    siSc

    r si

    USP

    46

    siU

    SP12

    Total GluA1 GFP Merge C

    0.0

    0.2

    0.4

    0.6

    0.8

    1.0

    1.2

    siScram siUSP46 siUSP12

    Tota

    l Glu

    A1

    *

    D

    EGFP

    U

    SP46

    Surf GluA1 GFP Merge

    EGFP

    U

    SP46

    Total GluA1 GFP Merge

    0.0

    0.2

    0.4

    0.6

    0.8

    1.0

    1.2

    1.4

    EGFP USP46

    Surfa

    ce G

    luA1

    **

    0.0

    0.2

    0.4

    0.6

    0.8

    1.0

    1.2

    EGFP USP46

    Tota

    l Glu

    A1

    A B

    C D

    0.00

    2.00

    4.00

    6.00

    8.00

    10.00

    12.00

    14.00

    scrambled siUSP46 siUSP12

    Amplitu

    de(p

    A) **

    0.00

    2.00

    4.00

    6.00

    8.00

    10.00

    12.00

    14.00

    16.00

    18.00

    Control USP46 USP12

    Amplitu

    de(p

    A)

    *

    Control

    USP46

    USP12

    scrambled

    siUSP46

    siUSP12

    A

    B

    C

    D

    E

    2s 10

    pA

    FIGURE 1. UPS46 synaptic distribution and developme.ntal expression.

    FIGURE 3. USP46 deubiquitinates GluA1 in vivo and in vitro.

    FIGURE 4. Knock down of USP46 enhances AMPAR ubiquitination.

    A and B. Double staining of USP46 (Red) and postsynaptic protein PSD-95 (Green) or GluA1 (Green) in cultured DIV14 hippocampal neu-rons. The selected region was enlarged for cla-rity (right). Arrowheads indicate co-localized puncta. Scale bar represents 20 μm. C. USP46 and other proteins including Nedd4, PSD-95 and GluA1 are enriched in the synaptosomal P2 fraction purified from adult rat hippocam-pal tissue. In contrast, Akt is mainly distribut-ed in the solubilized fraction. The same amount of protein (3 μg) was loaded for each lane. D. USP46 expression in different brain regions. USP46 is relatively higher in prefrontal cortex (PFC). The AMPAR E3 ligase Nedd4 and tubu-lin was also probed. E. USP46 expression time course was measured from cultured cortical neurons at varied days in vitro (DIV) after pla-ting. USP46 amounts peaked in the 2nd and 3rd weeks. GluA1 and tubulin were also probed.

    A and B. HEK cells were co-transfected with HA-ubiquitin (HA-Ubi), GFP-GluA1 together with either USP46 or USP12. GFP-GluA1 iso-lated by immunoprecipitation was probed for HA-ubiquitin. Ubiquitin signals (HA-Ubi) were reduced in cells expressing USP46, but not USP12. The same western blot was re-pro-bed with GluA1 antibody to confirm the im-munoprecipitation. Tubulin from cell lysates was probed to indicate equal amount of lysa-tes (N = 3 independent experiments). Bar gra-phes represent Mean ± SEM, *p