Available online at www.sciencedirect.com

ScienceDirect NMDA receptor dysfunction in autism spectrum disorders Eun-Jae Lee1, Su Yeon Choi2 and Eunjoon Kim2,3 Abnormalities and imbalances in neuronal excitatory and inhibitory synapses have been implicated in diverse neuropsychiatric disorders including autism spectrum disorders (ASDs). Increasing evidence indicates that dysfunction of NMDA receptors (NMDARs) at excitatory synapses is associated with ASDs. In support of this, human ASD-associated genetic variations are found in genes encoding NMDAR subunits. Pharmacological enhancement or suppression of NMDAR function ameliorates ASD symptoms in humans. Animal models of ASD display bidirectional NMDAR dysfunction, and correcting this deficit rescues ASD-like behaviors. These findings suggest that deviation of NMDAR function in either direction contributes to the development of ASDs, and that correcting NMDAR dysfunction has therapeutic potential for ASDs. Addresses 1 Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea 2 Department of Biological Sciences, KAIST, Daejeon 305-701, Republic of Korea 3 Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon 305-701, Republic of Korea Corresponding author: Kim, Eunjoon ([email protected])

Among known synaptic proteins implicated in ASD are metabotropic glutamate receptors (mGluRs). Functional enhancement and suppression of mGluR5 are associated with fragile X syndrome and tuberous sclerosis, respectively, which share autism as a common phenotype [5,6]. More recently, ionotropic glutamate receptors, namely NMDA receptors (NMDARs) and AMPA receptors (AMPARs), have also been implicated in ASDs [7]. In this review, we will focus on NMDA receptors and summarize evidence supporting the hypothesis that NMDAR dysfunction contributes to ASDs, and, by extension, that correcting NMDAR dysfunction has therapeutic potential for ASDs.

ASD-related human NMDAR genetic variants Clinical studies on ASDs have identified genetic variants of NMDAR subunit genes. Specifically, de novo mutations have been identified in the GRIN2B gene, encoding the GluN2B subunit [8,9,10–12]. In addition, SNP analyses have linked both GRIN2A (GluN2A subunit) and GRIN2B with ASDs [13]. Because assembled NMDARs contain four subunits, each with distinct properties [14], ASD-related GRIN2A/GRIN2B variants likely alter the functional properties of NMDARs and/or NMDAR-dependent plasticity.

Current Opinion in Pharmacology 2015, 20:8–13 This review comes from a themed issue on Neurosciences Edited by Pierre Paoletti and Jean-Philippe Pin For a complete overview see the Issue and the Editorial http://dx.doi.org/10.1016/j.coph.2014.10.007 1471-4892/# 2014 Elsevier Ltd. All rights reserved.

Introduction Autism spectrum disorders (ASDs) represent neurodevelopmental disorders characterized by two core symptoms; (1) impaired social interaction and communication, and (2) restricted and repetitive behaviors, interests, and activities. ASDs affect 1% of the population, and are considered to be highly genetic in nature [1,2]. A large number (600) of ASD-related genetic variations have been identified (sfari.org), and target gene functions are apparently quite diverse. However, some fall onto common pathways, including synaptic function and chromosome remodeling [3,4], suggesting that core mechanisms may exist. Current Opinion in Pharmacology 2015, 20:8–13

NMDAR modulators in ASD therapy Pharmacological modulation of NMDAR function can improve ASD symptoms. D-cycloserine (DCS), an NMDAR agonist, significantly ameliorates social withdrawal [15] and repetitive behavior [16] in individuals with ASD. These results suggest that reduced NMDAR function may contribute to the development of ASDs in humans. Elevated NMDAR function is also implicated in ASDs. Memantine, an NMDAR antagonist, and its analogue amantadine improve ASD-related symptoms including social deficits, inappropriate language, stereotypy, cognitive impairments, lethargy, irritability, inattention, and hyperactivity [17,18]. These results, together with the DCS results, highlight the importance of a normal range of NMDAR function, and suggest that deviation of NMDAR function in either direction leads to ASD. This concept is in line with the emerging view that synaptic function within a normal range is important and its deviation causes ASDs and intellectual disability [6,19].

Animal models of ASD with NMDAR dysfunction Animal studies have provided further support for the contribution of NMDAR dysfunction to ASDs. As is www.sciencedirect.com

NMDAR dysfunction and ASDs Lee, Choi and Kim 9

the case in humans, positive or negative modulators of NMDARs can normalize ASD-like phenotypes in animals. We can divide these animal studies into two groups. The first group consists of animals in which NMDAR modulators were shown to normalize both NMDAR dysfunction and ASD-like behaviors, establishing strong association between NMDARs and ASD phenotypes (Fig. 1). In the second group, NMDAR modulators were shown to rescue ASD-like behaviors, but NMDAR dysfunction and its correction have not been demonstrated. Specific examples of each group are described below and summarized in Table 1, sorted based on NMDAR function.

Group 1: ASD models with data showing rescue of both NMDAR dysfunction and ASDlike behaviors Neuroligin-1

Mice lacking neuroligin-1, an excitatory postsynaptic adhesion molecule, show reduced NMDAR function in the hippocampus and striatum, as evidenced by a decrease in NMDA/AMPA ratio and long-term potentiation (LTP) [20–22]. Neuroligin-1 is thought to enhance synaptic NMDAR function, by directly interacting with and promoting synaptic localization of NMDARs [22]. Figure 1 Brain function

Neuroligin-1/ mice show markedly increased selfgrooming, which is rapidly (30 min) attenuated by systemic injection of DCS [20]. Shank2

Shank2 is an excitatory postsynaptic scaffold important for synaptic protein assembly and signaling. Shank2/ mice lacking exons 6 and 7, a mutation found in humans [23], show reduced hippocampal NMDAR function and social deficits that are rapidly normalized by systemic DCS treatment [24]. CDPPB, a positive allosteric modulator of mGluR5 that potentiates NMDARs [25], similarly normalizes NMDAR dysfunction and behavioral deficits, consistent with the idea that indirectly modulating NMDARs through mGluR5 is a viable approach for treating ASDs [26]. Notably, mGluR5-dependent longterm depression (LTD) is unchanged in Shank2/ synapses, indicating that mGluR5 function is normal and suggesting that CDPPB-dependent rescue does not involve normalization of mGluR5 activity per se. Another line of Shank2/ mice that lacks only exon 7, a mutation also found in humans [23], exhibits enhanced rather than reduced NMDAR function, but shows social deficits similar to those observed in exon 6 + 7-deleted mice [27]. Although the mechanistic details are not clear, these results argue that deviation of NMDAR function in either direction can lead to ASD-like phenotypes. IRSp53

Neuroligin-1–/– Shank2 –/– (exon 6+7)

IRSp53 –/–

Tbr1+/– GluD1–/– BTBR Balb/c Selectively bred rats

Normal NMDAR function

Reduced

Group 2

Group 1 Phenotype

Excessive

Rescue

Phenotype

Rescue

ASD-like behaviors

Group 2: ASD models with data showing rescue of ASD-like behaviors but no demonstrated NMDAR dysfunction

NMDAR dysfunction

Current Opinion in Pharmacology

Bidirectional NMDAR dysfunction in animal models of ASD. Animal models of ASD with bidirectional NMDAR dysfunction can be positioned on either side of an NMDAR function curve. Model animals were divided into two groups. Group 1: NMDAR modulators normalize both NMDAR dysfunction and ASD-like behaviors (green). Group 2: NMDAR modulators rescue ASD-like behaviors, but NMDAR dysfunction and its rescue have not been demonstrated (orange). Note that Group 2 animals are tentatively placed on the left-hand side of the slope based on the observed DCS rescue of their ASD-like phenotypes, but the directions of their NMDAR dysfunctions remain to be experimentally determined. www.sciencedirect.com

IRSp53 is a postsynaptic scaffold that regulates F-actin in dendritic spines. IRSp53/ mice show an increased NMDA/AMPA ratio and impaired social interaction [28]. Memantine normalizes both NMDAR dysfunction and social deficits. MPEP, a negative allosteric modulator of mGluR5, similarly rescues the deficits in these animals, which exhibit normal mGluR-dependent LTD. These results suggest that NMDAR hyperfunction is associated with ASD-like phenotypes in IRSp53/ mice. Taken together with the results from neuroligin-1/ and Shank2/ mice, this associates bidirectional NMDAR dysfunction with ASD-like phenotypes.

Tbr1

Tbr1 is a transcriptional regulator, one of whose targets is the gene encoding the GluN2B subunit of NMDARs. Mice haploinsufficient for Tbr1 (Tbr1+/) show structural abnormalities in the amygdala and limited GluN2B induction upon behavioral stimulation [29]. Both systemic injection and local amygdalar infusion of DCS rescue social deficits and impaired associative memory in Tbr1+/ mice [29]. However, reduced NMDAR function and its DCS-dependent correction have not been demonstrated. Current Opinion in Pharmacology 2015, 20:8–13

10 Neurosciences

Table 1 NMDAR dysfunctions in animal models of ASD. Animal models of ASD are summarized with specific details of NMDAR dysfunction, ASDlike behavior, and pharmacological rescue. Hp, hippocampus; Str, striatum; NA, not available; DCS, D-cycloserine NMDAR function #

Region of NMDAR dysfunction

Animals

Neuroligin-1

/

mice

Hp, Str

ASD-like behaviors

Ref.

Sociability

Repetitive behavior

Pharmacological rescue

Grooming"

Grooming by DCS

[20–22]

Grooming and jumping"

Sociability by DCS and CDPPB

[24]

Shank2/ mice (exons 6+7)

Hp

Mildly impaired Impaired

"

IRSp53/mice

Hp

Impaired

Not increased (grooming and marble burying)

Sociability by memantine and MPEP

[28]

N/A

Tbr1+/ mice

N/A

Impaired

Not increased (grooming) N/A Grooming"

Sociability by DCS

[29]

Sociability by DCS Sociability and grooming by DCS, MPEP, and GRN-529 Sociability and stereotypy by DCS Sociability by NMDAR agonist, GLYX-13

[30,52] [32,33,34]

GluD1/ mice BTBR inbred mice

Impaired Impaired

Balb/c inbred mice

Impaired

Selectively bred rats

Impaired

GluD1

Mice lacking GluD1, a member of the delta family of ionotropic glutamate receptors, exhibit diminished social interaction and impaired contextual fear conditioning, both of which are rescued by DCS [30]. BTBR mice

BTBR is an inbred mouse model of ASD that shows social interaction deficits, impaired ultrasonic vocalization (USV), and repetitive behavior [31]. DCS improves social interaction and self-grooming in BTBR mice [32], suggesting that NMDAR function is reduced. In addition, negative allosteric modulators of mGluR5 such as MPEP and GRN-529 attenuate repetitive behavior and social deficits in BTBR mice [33,34], although whether this involves NMDAR modulation has not been determined. Balb/c mice

Balb/c is another inbred mouse strain with reduced sociability. DCS improves social interaction and stereotypy in these animals [35,36]. Notably, Balb/c mice show heightened behavioral sensitivity to the NMDAR antagonist, MK801 [37], suggesting that NMDAR function is reduced in these animals. Selectively bred rats

Rats selectively bred for low rates of play-related prosocial USVs show reduced social interaction and increased monotonous (non-frequency modulated) USVs. These Current Opinion in Pharmacology 2015, 20:8–13

Spontaneous stereotypy N/A

[35,36] [38]

USV phenotypes are rescued by GLYX-13, an NMDAR glycine site partial agonist [38].

NMDAR dysfunction in the development of ASDs The results from animal studies mentioned above lead to several fundamental questions that need to be discussed. First, how might mutations in diverse ASD-related genes converge onto NMDAR dysfunctions? Second, how does NMDAR dysfunction alter neuron and circuit functions to cause ASD-like phenotypes? Third, how does NMDAR dysfunction in either direction lead to similar ASD-like phenotypes? NMDARs have been shown to form large synaptic protein complexes together with other receptors, scaffolding proteins, signaling molecules, and cytoskeletal components [39], where a delicate balance between all the components would be required to fine-tune the function of ‘the NMDA receptosome’ in a homeostatic manner. Alterations in the levels or functions in any of these components may disturb the equilibrium and leads to NMDAR dysfunction. Regarding the second question, one straightforward possibility is that NMDAR dysfunction may impact neuronal or synaptic development, or NMDAR-dependent synaptic transmission or plasticity in mature neurons [14]. At molecular and cellular levels, NMDAR dysfunction may alter diverse downstream processes in a neuron, including synaptic signaling, AMPAR trafficking, www.sciencedirect.com

NMDAR dysfunction and ASDs Lee, Choi and Kim 11

neuronal excitability, excitation/inhibition balance (E/I balance), gene expression, and action potential firing. For the last question, one possibility is that NMDAR dysfunction may occur at excitatory synapses on interneuronal dendrites. For instance, reduced NMDAR function in interneurons likely decrease inhibitory input onto target neurons, increasing their excitability, a result that can also be caused by enhanced NMDAR function in the target neurons. In line with this, the impaired social interaction caused by elevated E/I balance in pyramidal neurons of the medial prefrontal cortex by optogenetic stimulation has been shown to be partially rescued by increasing inhibitory input onto these neurons by optogenetic excitation of adjacent parvalbumin-positive interneurons [40]. In addition, NMDAR dysfunction may also occur in modulatory neurons including dopaminergic, adrenergic, and cholinergic neurons, which have diverse influences on excitatory and inhibitory neurons.

Perspectives It should be noted that the hypothesized link between NMDAR dysfunction and ASDs currently relies on a small number of clinical and animal studies. Thus, a larger dataset will clearly be needed to verify the hypothesis. Notably, altered NMDAR functions have been reported in a number of known mouse or rat models of ASD, including Shank3DC/DC mice [41], Neuroligin3R451C knock-in mice [42–45,46], Fmr1/ mice [47], and mice and rats prenatally exposed to valproic acid [48,49], a teratogen known to induce ASDs in humans. However, whether correcting the NMDAR function among the diverse electrophysiological alterations observed in these animals can rescue some of the ASD-like behavioral deficits remain to be determined. An important question regarding the NMDAR dysfunction hypothesis would be to determine how NMDAR dysfunction leads to defects in neurons, circuits, and behaviors. Molecular and cellular mechanisms downstream of NMDAR activation should be carefully analyzed and dissected for specific circuit and behavioral defects. Additionally, experimental approaches and interpretations will require further refinement. First, many behavioral rescue experiments on animal models of ASD have used adult animals (i.e., 8–12-week-old mice), whereas electrophysiological measurements are often made in younger animals (i.e., 3–4 weeks). Though experimentally unavoidable, this approach creates a time gap between the two datasets that should be minimized and carefully interpreted. www.sciencedirect.com

Pharmacological rescue of both NMDAR dysfunction and ASD-like behaviors in experimental animals strongly links the two factors, but does not necessarily establish that NMDAR dysfunction causes the ASD-like behaviors. One way to address this question is to carefully monitor the changes occurring along the developmental time axis and, if possible, attempt conditional gene ablation or pharmacological rescue over specific developmental time windows, as previously reported [50,51]. ASDs involve diverse core and comorbid symptoms. Consistent with this, a single autism-related mutation, neuroligin-3 R451C, causes diverse synaptic phenotypes in different brain regions and circuits. Therefore, synaptic changes should be analyzed in greater detail, ideally using brain region-specific and cell type-specific conditional gene ablation, as recently reported. The NMDAR dysfunction hypothesis likely explains only a part of the behavioral deficits, as already discovered in many rescue studies. Modulators of mGluR5, in addition to NMDARs and AMPARs, have been considered to be a new means of regulating glutamatergic transmission [4,26]. Therefore, pharmacological rescue of animal models of ASD should ideally involve modulation of both NMDARs and mGluR5, or even other NMDA-modulatory approaches, to better facilitate translation to clinical therapy. Lastly, because our hypothesis associates bidirectional NMDAR dysfunction with ASDs, there may be clinical cases, such as where individuals with reduced NMDAR function are treated with NMDAR antagonists, which might aggravate the situation and affect the interpretation.

Conflict of interest Nothing declared.

Acknowledgement This work was supported by the Institute for Basic Science (IBS-R002-D1 to E.K.).

References and recommended reading Papers of particular interest, published within the period of review, have been highlighted as:  of special interest  of outstanding interest 1.

Huguet G, Ey E, Bourgeron T: The genetic landscapes of autism spectrum disorders. Annu Rev Genomics Hum Genet 2013, 14:191-213.

2.

Jeste SS, Geschwind DH: Disentangling the heterogeneity of autism spectrum disorder through genetic findings. Nat Rev Neurol 2014, 10:74-81.

3.

Krumm N, O’Roak BJ, Shendure J, Eichler EE: A de novo convergence of autism genetics and molecular neuroscience. Trends Neurosci 2014, 37:95-105. Current Opinion in Pharmacology 2015, 20:8–13

12 Neurosciences

4.

Spooren W, Lindemann L, Ghosh A, Santarelli L: Synapse dysfunction in autism: a molecular medicine approach to drug discovery in neurodevelopmental disorders. Trends Pharmacol Sci 2012, 33:669-684.

21. Chubykin AA, Atasoy D, Etherton MR, Brose N, Kavalali ET, Gibson JR, Sudhof TC: Activity-dependent validation of excitatory versus inhibitory synapses by neuroligin-1 versus neuroligin-2. Neuron 2007, 54:919-931.

5.

Bhakar AL, Dolen G, Bear MF: The pathophysiology of fragile X (and what it teaches us about synapses). Annu Rev Neurosci 2012, 35:417-443.

6.

Zoghbi HY, Bear MF: Synaptic dysfunction in neurodevelopmental disorders associated with autism and intellectual disabilities. Cold Spring Harb Perspect Biol 2012:4.

22. Budreck EC, Kwon OB, Jung JH, Baudouin S, Thommen A, Kim HS, Fukazawa Y, Harada H, Tabuchi K, Shigemoto R et al.: Neuroligin-1 controls synaptic abundance of NMDA-type glutamate receptors through extracellular coupling. Proc Natl Acad Sci U S A 2013, 110:725-730.

7.

8.

Uzunova G, Hollander E, Shepherd J: The role of ionotropic glutamate receptors in childhood neurodevelopmental disorders: autism spectrum disorders and fragile X syndrome. Curr Neuropharmacol 2014, 12:71-98. Kenny EM, Cormican P, Furlong S, Heron E, Kenny G, Fahey C, Kelleher E, Ennis S, Tropea D, Anney R et al.: Excess of rare novel loss-of-function variants in synaptic genes in schizophrenia and autism spectrum disorders. Mol Psychiatry 2014, 19:872-879.

O’Roak BJ, Vives L, Fu W, Egertson JD, Stanaway IB, Phelps IG, Carvill G, Kumar A, Lee C, Ankenman K et al.: Multiplex targeted sequencing identifies recurrently mutated genes in autism spectrum disorders. Science 2012, 338:1619-1622. This human genetics study identifies GRIN2B, which encodes the GluN2B subunit of NMDARs, as one of the key ASD-releated genes.

9. 

10. O’Roak BJ, Vives L, Girirajan S, Karakoc E, Krumm N, Coe BP, Levy R, Ko A, Lee C, Smith JD et al.: Sporadic autism exomes reveal a highly interconnected protein network of de novo mutations. Nature 2012, 485:246-250. 11. O’Roak BJ, Deriziotis P, Lee C, Vives L, Schwartz JJ, Girirajan S, Karakoc E, Mackenzie AP, Ng SB, Baker C et al.: Exome sequencing in sporadic autism spectrum disorders identifies severe de novo mutations. Nat Genet 2011, 43:585-589. 12. Tarabeux J, Kebir O, Gauthier J, Hamdan FF, Xiong L, Piton A, Spiegelman D, Henrion E, Millet B, team SD et al.: Rare mutations in N-methyl-D-aspartate glutamate receptors in autism spectrum disorders and schizophrenia. Transl Psychiatry 2011, 1:e55. 13. Yoo HJ, Cho IH, Park M, Yang SY, Kim SA: Family based association of GRIN2A and GRIN2B with Korean autism spectrum disorders. Neurosci Lett 2012, 512:89-93. 14. Paoletti P, Bellone C, Zhou Q: NMDA receptor subunit diversity: impact on receptor properties, synaptic plasticity and disease. Nat Rev Neurosci 2013, 14:383-400. 15. Posey DJ, Kem DL, Swiezy NB, Sweeten TL, Wiegand RE, McDougle CJ: A pilot study of D-cycloserine in subjects with autistic disorder. Am J Psychiatry 2004, 161:2115-2117. 16. Urbano M, Okwara L, Manser P, Hartmann K, Herndon A, Deutsch SI: A trial of D-cycloserine to treat stereotypies in older  adolescents and young adults with autism spectrum disorder. Clin Neuropharmacol 2014, 37:69-72. This clinical study, together with another study [15], demonstrates that DCS can ameliorate social interaction and repetitive behavior in individuals with ASD. 17. Hosenbocus S, Chahal R: Memantine: a review of possible uses in child and adolescent psychiatry. J Can Acad Child Adolesc Psychiatry 2013, 22:166-171. 18. Hosenbocus S, Chahal R: Amantadine: a review of use in child and adolescent psychiatry. J Can Acad Child Adolesc Psychiatry 2013, 22:55-60. 19. Auerbach BD, Osterweil EK, Bear MF: Mutations causing  syndromic autism define an axis of synaptic pathophysiology. Nature 2011, 480:63-68. This study demonstrates that deviation of mGluR5 function in either direction causes cognitive deficits in mice, and correcing mGluR5 dysfunction by pharmacological or mouse genetic approaches rescues these phenotypes. 20. Blundell J, Blaiss CA, Etherton MR, Espinosa F, Tabuchi K, Walz C, Bolliger MF, Sudhof TC, Powell CM: Neuroligin-1 deletion results in impaired spatial memory and increased repetitive behavior. J Neurosci 2010, 30:2115-2129. Current Opinion in Pharmacology 2015, 20:8–13

23. Berkel S, Marshall CR, Weiss B, Howe J, Roeth R, Moog U, Endris V, Roberts W, Szatmari P, Pinto D et al.: Mutations in the SHANK2 synaptic scaffolding gene in autism spectrum disorder and mental retardation. Nat Genet 2010, 42:489-491. 24. Won H, Lee HR, Gee HY, Mah W, Kim JI, Lee J, Ha S, Chung C,  Jung ES, Cho YS et al.: Autistic-like social behaviour in Shank2mutant mice improved by restoring NMDA receptor function. Nature 2012, 486:261-265. This article associates reduced NMDAR function with ASD-like social deficits in Shank2/ mice by rescuing both phenotypes with DCS. 25. Awad H, Hubert GW, Smith Y, Levey AI, Conn PJ: Activation of metabotropic glutamate receptor 5 has direct excitatory effects and potentiates NMDA receptor currents in neurons of the subthalamic nucleus. J Neurosci 2000, 20:7871-7879. 26. Gasparini F, Bilbe G, Gomez-Mancilla B, Spooren W: mGluR5 antagonists: discovery, characterization and drug development. Curr Opin Drug Discov Dev 2008, 11:655-665. 27. Schmeisser MJ, Ey E, Wegener S, Bockmann J, Stempel AV,  Kuebler A, Janssen AL, Udvardi PT, Shiban E, Spilker C et al.: Autistic-like behaviours and hyperactivity in mice lacking ProSAP1/Shank2. Nature 2012, 486:256-260. This article demonstrates Shank2 deficiency leads to NMDAR hyperfunction and ASD-like phenotypes, suggesting, together with Ref. [24], that deviation of NMDAR function in either direction is associated with ASDlike phenotypes. 28. Chung W, Choi SY, Lee E, Park H, Kang J, Park H, Choi Y, Lee D, Park SG, Kim R et al.: Social deficits in IRSp53 mutant mice improved by NMDAR and mGluR5 suppression. Nat Neurosci, in press. http://dx.doi.org/10.1038/nn.3927 29. Huang TN, Chuang HC, Chou WH, Chen CY, Wang HF, Chou SJ,  Hsueh YP: Tbr1 haploinsufficiency impairs amygdalar axonal projections and results in cognitive abnormality. Nat Neurosci 2014, 17:240-247. The authors demonstrate that haploinsufficiency of the transcription factor Tbr1 leads to ASD-like behaviors in mice that can be rescued by systemic or amygdalar administration of DCS. 30. Yadav R, Hillman BG, Gupta SC, Suryavanshi P, Bhatt JM, Pavuluri R, Stairs DJ, Dravid SM: Deletion of glutamate delta-1 receptor in mouse leads to enhanced working memory and deficit in fear conditioning. PLoS ONE 2013, 8:e60785. 31. Silverman JL, Yang M, Lord C, Crawley JN: Behavioural phenotyping assays for mouse models of autism. Nat Rev Neurosci 2010, 11:490-502. 32. Burket JA, Benson AD, Tang AH, Deutsch SI: D-Cycloserine improves sociability in the BTBR T+ Itpr3tf/J mouse model of autism spectrum disorders with altered Ras/Raf/ERK1/2 signaling. Brain Res Bull 2013, 96:62-70. 33. Silverman JL, Tolu SS, Barkan CL, Crawley JN: Repetitive selfgrooming behavior in the BTBR mouse model of autism is blocked by the mGluR5 antagonist MPEP. Neuropsychopharmacology 2010, 35:976-989. 34. Silverman JL, Smith DG, Rizzo SJ, Karras MN, Turner SM, Tolu SS, Bryce DK, Smith DL, Fonseca K, Ring RH et al.: Negative  allosteric modulation of the mGluR5 receptor reduces repetitive behaviors and rescues social deficits in mouse models of autism. Sci Transl Med 2012, 4:131ra151. This study demonstrates that mGluR5 modulators can rescue ASD-like deficits in animal models, suggesting, together with [24,33], that mGluR5 modulation has therapeutic potential for ASD. 35. Benson AD, Burket JA, Deutsch SI: Balb/c mice treated with Dcycloserine arouse increased social interest in conspecifics. Brain Res Bull 2013, 99:95-99. www.sciencedirect.com

NMDAR dysfunction and ASDs Lee, Choi and Kim 13

36. Deutsch SI, Pepe GJ, Burket JA, Winebarger EE, Herndon AL, Benson AD: D-Cycloserine improves sociability and spontaneous stereotypic behaviors in 4-week old mice. Brain Res 2012, 1439:96-107. 37. Perera PY, Lichy JH, Mastropaolo J, Rosse RB, Deutsch SI: Expression of NR1, NR2A and NR2B NMDA receptor subunits is not altered in the genetically-inbred Balb/c mouse strain with heightened behavioral sensitivity to MK-801, a noncompetitive NMDA receptor antagonist. Eur Neuropsychopharmacol 2008, 18:814-819. 38. Burgdorf J, Moskal JR, Brudzynski SM, Panksepp J: Rats selectively bred for low levels of play-induced 50 kHz vocalizations as a model for autism spectrum disorders: a role for NMDA receptors. Behav Brain Res 2013, 251:18-24. 39. Husi H, Ward MA, Choudhary JS, Blackstock WP, Grant SG: Proteomic analysis of NMDA receptor-adhesion protein signaling complexes. Nat Neurosci 2000, 3:661-669. 40. Yizhar O, Fenno LE, Prigge M, Schneider F, Davidson TJ, O’Shea DJ, Sohal VS, Goshen I, Finkelstein J, Paz JT et al.: Neocortical excitation/inhibition balance in information processing and social dysfunction. Nature 2011, 477:171-178.

45. Foldy C, Malenka RC, Sudhof TC: Autism-associated neuroligin-3 mutations commonly disrupt tonic endocannabinoid signaling. Neuron 2013, 78:498-509. 46. Rothwell PE, Fuccillo MV, Maxeiner S, Hayton SJ, Gokce O,  Lim BK, Fowler SC, Malenka RC, Sudhof TC: Autism-associated neuroligin-3 mutations commonly impair striatal circuits to boost repetitive behaviors. Cell 2014, 158:198-212. This study, together with other studies on mice carrying a neuroligin-3 mutation (R451C) [40,41,42,44], demonstrates that a single gene defect can lead to diverse synaptic deficits, including NMDAR dysfunction and impaired GABAergic transmission. 47. Pilpel Y, Kolleker A, Berberich S, Ginger M, Frick A, Mientjes E, Oostra BA, Seeburg PH: Synaptic ionotropic glutamate receptors and plasticity are developmentally altered in the CA1 field of Fmr1 knockout mice. J Physiol 2009, 587:787-804. 48. Rinaldi T, Kulangara K, Antoniello K, Markram H: Elevated NMDA receptor levels and enhanced postsynaptic long-term potentiation induced by prenatal exposure to valproic acid. Proc Natl Acad Sci U S A 2007, 104:13501-13506. 49. Mehta MV, Gandal MJ, Siegel SJ: mGluR5-antagonist mediated reversal of elevated stereotyped, repetitive behaviors in the VPA model of autism. PLoS ONE 2011, 6:e26077.

41. Kouser M, Speed HE, Dewey CM, Reimers JM, Widman AJ, Gupta N, Liu S, Jaramillo TC, Bangash M, Xiao B et al.: Loss of predominant Shank3 isoforms results in hippocampusdependent impairments in behavior and synaptic transmission. J Neurosci 2013, 33:18448-18468.

50. Baudouin SJ, Gaudias J, Gerharz S, Hatstatt L, Zhou K, Punnakkal P, Tanaka KF, Spooren W, Hen R, De Zeeuw CI et al.: Shared synaptic pathophysiology in syndromic and nonsyndromic rodent models of autism. Science 2012, 338:128-132.

42. Tabuchi K, Blundell J, Etherton MR, Hammer RE, Liu X, Powell CM, Sudhof TC: A neuroligin-3 mutation implicated in autism increases inhibitory synaptic transmission in mice. Science 2007, 318:71-76.

51. Clement JP, Aceti M, Creson TK, Ozkan ED, Shi Y, Reish NJ,  Almonte AG, Miller BH, Wiltgen BJ, Miller CA et al.: Pathogenic SYNGAP1 mutations impair cognitive development by disrupting maturation of dendritic spine synapses. Cell 2012, 151:709-723. The authors demonstrate that haploinsufficiency of SYNGAP1 leads to premature spine development and neuronal hyperactivity, which requires the mutation to be present during a critical developmental time window (first 3 weeks).

43. Jaramillo TC, Liu S, Pettersen A, Birnbaum SG, Powell CM: Autism-related neuroligin-3 mutation alters social behavior and spatial learning. Autism Res 2014, 7:264-272. 44. Etherton M, Foldy C, Sharma M, Tabuchi K, Liu X, Shamloo M, Malenka RC, Sudhof TC: Autism-linked neuroligin-3 R451C mutation differentially alters hippocampal and cortical synaptic function. Proc Natl Acad Sci U S A 2011, 108:13764-13769.

www.sciencedirect.com

52. Yadav R, Gupta SC, Hillman BG, Bhatt JM, Stairs DJ, Dravid SM: Deletion of glutamate delta-1 receptor in mouse leads to aberrant emotional and social behaviors. PLoS ONE 2012, 7:e32969.

Current Opinion in Pharmacology 2015, 20:8–13

NMDA receptor dysfunction in autism spectrum disorders.

Abnormalities and imbalances in neuronal excitatory and inhibitory synapses have been implicated in diverse neuropsychiatric disorders including autis...
416KB Sizes 0 Downloads 20 Views