METHODS OF IDENTIFYING COMPOUNDS FOR TREATING DEPRESSION AND OTHER RELATED DISEASES

Information

  • Patent Application
  • 20150253305
  • Publication Number
    20150253305
  • Date Filed
    October 11, 2013
    10 years ago
  • Date Published
    September 10, 2015
    9 years ago
Abstract
The present disclosure relates in part to methods of identifying a candidate compound suitable for treatment of depression. In some embodiments, a candidate compound may be a NMDAR partial agonist.
Description
BACKGROUND

The N-methyl-D-aspartate (NMDA) receptor (NMDAR) has been implicated in neurodegenerative disorders including stroke-related brain cell death, convulsive disorders, and learning and memory. NMDAR also plays a central role in modulating normal synaptic transmission, synaptic plasticity, and excitotoxicity in the central nervous system. The NMDAR is further involved in Long-term potentiation (LTP).


The NMDAR is activated by the binding of NMDA, glutamate (Glu), and aspartate (Asp). It is competitively antagonized by D-2-amino-5-phosphonovalerate (D-APS; D-APV), and non-competitively antagonized by phenylcyclidine (PCP), and MK-801. Most interestingly, the NMDAR is co-activated by glycine (Gly) (Kozikowski et al., 1990, Journal of Medicinal Chemistry 33:1561-1571). The binding of glycine occurs at an allosteric regulatory site on the NMDAR complex, and this increases both the duration of channel open time, and the frequency of the opening of the NMDAR channel.


Recent human clinical studies have identified NMDAR as a novel target of high interest for treatment of depression. These studies conducted using known NMDAR antagonists CPC-101,606 and ketamine have shown significant reductions in the Hamilton Depression Rating Score in patients suffering with refractory depression. Although, the efficacy was significant, but the side effects of using these NDMAR antagonists were severe.


NMDA-modulating small molecule agonist and antagonist compounds have been developed for potential therapeutic use. However, many of these are associated with very narrow therapeutic indices and undesirable side effects including hallucinations, ataxia, irrational behavior, and significant toxicity, all of which limit their effectiveness and/or safety. Further, 50% or more of patients with depression do not experience an adequate therapeutic response to known administered drugs. There currently is no single effective treatment for depression, anxiety, and other related diseases.


Thus, there remains a need for improved treatments of depression, anxiety and/or other related diseases with compounds that provide increased efficacy and reduced undesirable side effects.


SUMMARY

The present disclosure relates in part to methods of identifying a candidate compound suitable for treatment of depression. In some embodiments, a candidate compound may be a NMDAR partial agonist.


In one aspect, a method for identifying a candidate compound suitable for treatment of depression is provided. The method comprises exposing a cell to a potential compound in a culture medium, or administering a potential compound to an animal; retrieving a sample from the cell and/or culture medium, or from brain or neural tissue of the animal, at one or more predetermined time points; analyzing the sample for increased or decreased expression levels of Wnt1 and/or identifying the candidate compound as suitable for treatment of depression based on the increased expression level of Wnt1.


In another aspect, a method for identifying a candidate compound suitable for treatment of depression is provided. The method comprises exposing a cell to a potential compound in a culture medium, or administering a potential compound to an animal; retrieving a sample from the cell and/or culture medium, or from brain or neural tissue of the animal, at one or more predetermined time points; analyzing the sample for increased expression levels of at least one of the genes listed in Table 1 or 2 indicated with a G, or decreased expression levels or at least one of the genes listed in Table 1 or 2 indicated with a K, and identifying the compound as suitable for treatment of depression based on the increased expression level or decreased expression level.


In some embodiments, the sample has a gene expression pattern as provided in Table 1 or 2 with the indication “G” and the identifying is based on increased expression of those genes.


In some embodiments, a contemplated method further comprises analyzing the candidate compound for NDMA subunit NR2B synaptic plasticity.


In another aspect, a method for identifying a compound suitable for treatment of depression is provided. The method comprises exposing a cell to a potential compound in a culture medium, or administering a potential compound to an animal; retrieving a sample from the cell and/or culture medium, or from brain or neural tissue of the animal, at one or more predetermined time points; analyzing the sample for NMDA receptor NR2B subunit plasticity, and identifying the compound as suitable for treatment of depression based on inducing the NR2B plasticity.


In some embodiments, a candidate compound suitable for treating depression significantly induces NR2B dependent synaptic plasticity as compared to ketamine.


In some embodiments, the tissue is medial prefrontal cortex.


In some embodiments, the animal is a rodent or human, and the cell is a human or rodent cell.


In some embodiments, the compound modulates the NMDA receptor.


In some embodiments, the compound suitable for treating depression has fewer side effects as compared to ketamine.


In some embodiments, the compound does not have substantial addictive sensory motor grating and/or sedative effect.


In some embodiments, the cell is a eukaryotic cell.


In some embodiments, a contemplated method further comprises selecting the candidate compound from a library of compounds.


In yet another aspect, a method of identifying a therapeutic compound capable of treating depression in a patient is provided. The method comprises selecting a compound that significantly induces NR2B dependent synaptic plasticity.





BRIEF DESCRIPTION OF THE FIGURES


FIG. 1 shows the effects of GLYX-13 and Ketamine on gene expression in the medial prefrontal cortex of an adult rat at 1 hour and 24 hours post-IV injection.



FIG. 2 shows the effects of GLYX-13 on Wnt pathway specific gene expression relative to vehicle controls.



FIG. 3 shows a schematic of the Wnt signaling pathway.



FIG. 4 shows the anti-depressant-like effects of GLYX-13 in multiple rat models.



FIG. 5 shows the results from various tests which indicate that GLYX-13 does not show ketamine-like addictive sensory-motor gating, or sedative side effects.



FIG. 6 shows the results of Porsolt tests which demonstrate that GLYX-13 is antidepressant-like compared to ketamine.



FIG. 7 shows results which demonstrate the GLYX-13 induces NR2B-dependent plasticity.



FIG. 8 shows the antidepressant-like effects of GLYX-13 are synaptic-plasticity related.



FIG. 9 shows GLYX-13 increases ex vivo [3H] MK-801 binding in rat medial prefrontal cortex 1 hour after dosing.



FIG. 10 shows plots of phosphoserine 1303 NR2B (pS1303 NR2B) protein levels (left panel) and total NR2B protein levels (middle panel) as a function of post-dosing time with GLYX-13 and shows a bar graph (right panel) of the ratio of pS1303 NR2B protein levels to total NR2B protein levels as a function of post-dosing time with GLYX-13.



FIG. 11 shows plots of phosphoserine 1480 NR2B (pS1480 NR2B) protein levels (left panel) and total NR2B protein levels (middle panel) as a function of post-dosing time with GLYX-13 and shows a bar graph (right panel) of the ratio of pS1480 NR2B protein levels to total NR2B protein levels as a function of post-dosing time with GLYX-13.



FIG. 12 shows a bar graph (left panel) of CK2 kinase activity 15 minutes post-dosing with GLYX-13 as measured by phosphorylation of a CK2 substrate and shows a plot (right panel) of a standard curve for the CK2 kinase activity assay.





DETAILED DESCRIPTION

The present disclosure relates in part to methods of identifying a candidate compound suitable for treatment of depression. In some embodiments, a candidate compound may be a NMDAR partial agonist. In another aspect, the present disclosure relates in part to the use of identified compounds for treatment of clinically relevant depression and/or for general treatment of depression and/or anxiety.


Depression is a common psychological problem and refers to a mental state of low mood and aversion to activity. Various symptoms associated with depression include persistent anxious or sad feelings, feelings of helplessness, hopelessness, pessimism, and/or worthlessness, low energy, restlessness, irritability, fatigue, loss of interest in pleasurable activities or hobbies, excessive sleeping, overeating, appetite loss, insomnia, thoughts of suicide, and suicide attempts. The presence, severity, frequency, and duration of the above mentioned symptoms vary on a case to case basis. In some embodiments, a patient may have at least one, at least two, at least three, at least four, or at least five of these symptoms.


The most common depression conditions include Major Depressive Disorder and Dysthymic Disorder. Other depression conditions develop under unique circumstances. Such depression conditions include but are not limited to Psychotic depression, Postpartum depression, Seasonal affective disorder (SAD), mood disorder, depressions caused by chronic medical conditions such as cancer or chronic pain, chemotherapy, chronic stress, post traumatic stress disorders, and Bipolar disorder (or manic depressive disorder). Refractory depression occurs in patients suffering from depression who are resistant to standard pharmacological treatments, including tricyclic antidepressants, MAOIs, SSRIs, and double and triple uptake inhibitors and/or anxiolytic drugs, as well non-pharmacological treatments such as psychotherapy, electroconvulsive therapy, vagus nerve stimulation and/or transcranial magnetic stimulation. A treatment resistant-patient may be identified as one who fails to experience alleviation of one or more symptoms of depression (e.g., persistent anxious or sad feelings, feelings of helplessness, hopelessness, pessimism) despite undergoing one or more standard pharmacological or non-pharmacological treatment. In certain embodiments, a treatment-resistant patient is one who fails to experience alleviation of one or more symptoms of depression despite undergoing treatment with two different antidepressant drugs. In other embodiments, a treatment-resistant patient is one who fails to experience alleviation of one or more symptoms of depression despite undergoing treatment with four different antidepressant drugs. A treatment-resistant patient may also be identified as one who is unwilling or unable to tolerate the side effects of one or more standard pharmacological or non-pharmacological treatment. In certain embodiments, methods for treating refractory depression by administering an effective amount of an identified compound to a treatment-resistant patient in need thereof are contemplated. In an embodiment, methods of treating depression is contemplated when a patient has suffered depression for e.g. 5, 6, 7, 8 or more weeks, or for a month or more.


In an embodiment, a method for identifying a candidate compound suitable for treatment of depression is provided comprising exposing a cell to a potential compound in a culture medium, or administering a potential compound to an animal; retrieving a sample from the cell and/or culture medium, or from brain or neural tissue of the animal, at one or more predetermined time points; analyzing the sample for increased expression levels of Wnt1, and/or identifying the candidate compound as suitable for treatment of depression based on the increased expression level of Wnt1.


In another embodiment, a method for identifying a candidate compound suitable for treatment of depression, is provided comprising: exposing a cell to a potential compound in a culture medium, and/or or administering a potential compound to an animal; retrieving a sample from the cell and/or culture medium, or from brain or neural tissue of the animal, at one or more predetermined time points; analyzing the sample for increased expression levels of at least one of the genes listed in Table 1 or 2 (as provided below) indicated with a G, or decreased expression levels or at least one of the genes listed in Table 1 or 2 indicated with a K, and identifying the compound as suitable for treatment of depression based on the increased expression level or decreased expression level.


A contemplated sample may have a gene expression pattern as provided in Table 1 or 2 with the indication “G” and the identifying is based on increased expression of those genes.


Contemplated methods may further comprising analyzing the candidate compound for NDMA subunit NR2B synaptic plasticity.


A method for identifying a compound suitable for treatment of depression or other indications is provided herein in an embodiment is provided, wherein the method may include exposing a cell to a potential compound in a culture medium, or administering a potential compound to an animal; retrieving a sample from the cell and/or culture medium, or from brain or neural tissue of the animal, at one or more predetermined time points; analyzing the sample for NMDA receptor NR2B subunit plasticity, and identifying the compound as suitable for treatment of depression based on inducing the NR2B plasticity. A candidate compound suitable for treating depression may significantly induce NR2B dependent synaptic plasticity as compared to ketamine.


Tissues contemplated herein may be tissue of medial prefrontal cortex. Animals contemplated may be a rodent or human; cells may be a human or rodent cell. Contemplated candidate compounds may modulate a NMDA receptor, e.g. a candidate compound may be a NMDA partial agonist.


A candidate compound suitable for treating depression may have fewer side effects as compared to ketamine, for example the compound may not have substantial addictive sensory motor grating and/or sedative effect.


In an embodiment, a method of identifying a therapeutic compound capable of treating depression in a patient, is provided, comprising selecting a compound that significantly induces NR2B dependent synaptic plasticity.


Identified compounds may act predominantly at NR2B-containing NMDARs, and may not display the classic side effects of known NMDAR modulators such as CPC-101,606 and ketamine. For example, identified compounds may have markedly elevated long-term potentiation (LTP) while simultaneously reducing long-term depression (LTD) in rat hippocampal organotypic cultures. In some embodiments, identified compounds may produce an antidepressant effect essentially without dissociative side effects when administered to a subject in therapeutic amounts. In certain embodiments, an antidepressant effect with essentially no sedation may be produced by identified compounds when administered to a subject in therapeutic amounts. In still other embodiments, identified compounds may not have abuse potential (e.g., may not be habit-forming).


In some embodiments, compounds may increase AMPA GluR1 serine-845 phosphorylation or reduce expression in Wnt1 or Wnt signaling, for example as compared to ketamine.


Additionally, identified compounds may have better Blood-Brain Barrier (BBB) penetration as compared to many of the earlier glycine site ligands (Leeson & Iversen, J. Med. Chem. 37:4053-4067, 1994) and may cross the BBB readily. In some embodiments, identified compoudns or a composition comprising same may provide better i.v. in vivo potency and/or brain level concentration, relative to plasma levels, e.g. as compared to ketamine.


A variety of depression conditions are expected to be treated with an identified compound without for example affecting behavior or motor coordination, and without inducing or promoting seizure activity. Exemplary depression conditions that are expected to be treated according to this aspect include, but are not limited to, major depressive disorder, dysthymic disorder, psychotic depression, postpartum depression, premenstrual syndrome, premenstrual dysphoric disorder, seasonal affective disorder (SAD), anxiety, mood disorder, depressions caused by chronic medical conditions such as cancer or chronic pain, chemotherapy, chronic stress, post traumatic stress disorders, risk of suicide, and bipolar disorder (or manic depressive disorder). It should be understood that depression caused by bipolar disorder may be referred to as bipolar depression. In addition, patients suffering from any form of depression often experience anxiety. Various symptoms associated with anxiety include fear, panic, heart palpitations, shortness of breath, fatigue, nausea, and headaches among others. It is expected that the methods of the present condition can be used to treat anxiety or any of the symptoms thereof.


In addition, a variety of other neurological conditions are expected to be treated according to the methods. Exemplary conditions include, but are not limited to, a learning disorder, autistic disorder, attention-deficit hyperactivity disorder, Tourette's syndrome, phobia, post-traumatic stress disorder, dementia, AIDS dementia, Alzheimer's disease, Parkinson's disease, Huntington's disease, spasticity, myoclonus, muscle spasm, bipolar disorder, a substance abuse disorder, urinary incontinence, and schizophrenia.


Also provided herein are methods of treating depression in treatment resistant patients or treating refractory depression, e.g., patients suffering from a depression disorder that does not, and/or has not, responded to adequate courses of at least one, or at least two, other antidepressant compounds or therapeutics. For example, provided herein is a method of treating depression in a treatment resistant patient, comprising a) optionally identifying the patient as treatment resistant and b) administering an effective dose of an identified compound to said patient.


Symptoms of depression, and relief of same, may be ascertained by a physician or psychologist, e.g. by a mental state examination. Symptoms include thoughts of hopelessness, self-harm or suicide and/or an absence of positive thoughts or plans.


Contemplated methods include a method of treating autism and/or an autism spectrum disorder in a patient need thereof, comprising administering an effective amount of an identified to the patient. For example, upon administration, an identified compound may decrease the incidence of one or more symptoms of autism such as eye contact avoidance, failure to socialize, attention deficit, poor mood, hyperactivity, abnormal sound sensitivity, inappropriate speech, disrupted sleep, and perseveration. Such decreased incidence may be measured relative to the incidence in the untreated individual or an untreated individual(s). In some embodiments, patients suffering from autism also suffer from another medical condition, such as Fragile X syndrome, tuberous sclerosis, congenital rubella syndrome, and untreated phenylketonuria.


In another embodiment, methods of treating a disorder in a patient need thereof are contemplated, wherein the disorder is selected from group consisting of: epilepsy, AIDS dementia, multiple system atrophy, progressive supra-nuclear palsy, Friedrich's ataxia, autism, fragile X syndrome, tuberous sclerosis, attention deficit disorder, olivio-ponto-cerebellar atrophy, cerebral palsy, drug-induced optic neuritis, peripheral neuropathy, myelopathy, ischemic retinopathy, glaucoma, cardiac arrest, behavior disorders, and impulse control disorders that includes administering an identified compound.


In an embodiment, contemplated herein are methods of treating attention deficit disorder, ADHD (attention deficit hyperactivity disorder), schizophrenia, anxiety, amelioration of opiate, nicotine and/or ethanol addiction (e.g., method of treating such addiction or ameliorating the side effects of withdrawing from such addiction), spinal cord injury diabetic retinopathy, traumatic brain injury, post-traumatic stress syndrome and/or Huntington's chorea, in a patient in need thereof, that includes administering an identified compound. For example, patients suffering from schizophrenia, addiction (e.g. ethanol or opiate), autism, Huntington's chorea, traumatic brain injury, spinal cord injury, post-traumatic stress syndrome and diabetic retinopathy may all be suffering from altered NMDA receptor expression or functions.


In another embodiment, a method of treating Alzheimer's disease, or e.g., treatment of memory loss that e.g., accompanies early stage Alzheimer's disease, in a patient in need thereof is provided, comprising administering an identified compound.


Toxicity and therapeutic efficacy of subject compounds may be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 and the ED50.


As used herein, the term “GLYX peptide” refers to a peptide having NMDAR glycine-site partial agonist/antagonist activity. GLYX peptides may be obtained by well-known recombinant or synthetic methods such as those described in U.S. Pat. Nos. 5,763,393 and 4,086,196 herein incorporated by reference. In some embodiments, GLYX refers to a tetrapeptide having the amino acid sequence Thr-Pro-Pro-Thr (SEQ ID NO: 13), or L-threonyl-L-prolyl-L-prolyl-L-threonine amide. In some embodiments, candidate compounds have the same microarray results as GLYX-13 and/or the below compounds.


For example, GLYX-13 refers to the compound depicted as:




embedded image


Also contemplated are polymorphs, homologs, hydrates, solvates, free bases, and/or suitable salt forms of GLYX 13 such as, but not limited to, the acetate salt. The peptide may be cyclyzed or non-cyclyzed form as further described in U.S. Pat. No. 5,763,393. In some embodiments, an a GLYX-13 analog may include an insertion or deletion of a moiety on one or more of the Thr or Pro groups such as a deletion of CH2, OH, or NH2 moiety. In other embodiments, GLYX-13 may be optionally substituted with one or more halogens, C1-C3 alkyl (optionally substituted with halogen or amino), hydroxyl, and/or amino Glycine-site partial agonist of the NMDAR are disclosed in U.S. Pat. No. 5,763,393, U.S. Pat. No. 6,107,271, and Wood et al., NeuroReport, 19, 1059-1061, 2008, the entire contents of which are herein incorporated by reference.


Candidate compounds may have substantially the same gene expression effect as one or more of the following compounds:




embedded image


embedded image


embedded image


embedded image


The present disclosure has multiple aspects, illustrated by the following non-limiting examples.


EXAMPLES
Example 1
Gene Expression Patterns After Administration of GLYX-13 or Ketamine

In the present study, gene expression patterns in the medial prefrontal cortex (mPFC) were examined following either GLYX-13 (3 mg/kg, IV; the lowest dose that produces an anti-depressant effect in the Porsolt test) or ketamine (10 mg/kg, IV; a dose that produces a long-lasting anti-depressant effect in the Porsolt test) using a focused microarray platform combined with ontological analyses to identify functionally related gene sets that were differentially effected by GLYX-13 and ketamine Among the most interesting of these was the Wnt signaling pathway. A Wnt pathway-specific qRT-PCR array was used to corroborate these findings. Using this qRT-PCR array, the results showed that at 1 hr after GLYX-13 injections, 5 genes were differentially expressed as compared to saline treated control rats. At 24 hrs after GLYX-13 administration, 4 genes were upregulated. At 1 and 24 hrs following ketamine administration only 1 gene was downregulated. Taken together, these data suggest that although both GLYX-13 and ketamine produce rapid antidepressant-like effects in the Porsolt test, they likely effect changes in different cellular signaling pathways; one such example being the Wnt signaling pathway.


Methods:

Animals:


Adult (2-3 month-old) male Sprague-Dawley rats (Harlan Laboratories, Indianapolis, Ind.) were housed 3 to a cage and injected (IV) with one of the following—GLYX-13 (3 mg/kg), ketamine (10 mg/kg), or saline vehicle (1 ml/kg). At 1 and 24 hours after injection (N=5 per time point for each treatment group), rats were sacrificed, their brains were quickly dissected, frozen, and then stored at −80° C. The medial prefrontal cortex (mPFC) was dissected from frozen tissue on ice. An equal volume of the homogenized tissue was used to extract and purify RNA for microarray analysis and to make cDNA for qRT-PCR analysis. All procedures were approved by the Northwestern University IACUC committee and performed in accordance with the NIH Guide for the Care and Use of Laboratory Animals.


Transcriptomics:


Using an in-house microarray (Kroes et al., 2006), we assayed the expression of 1,178 genes specific to the rat brain and representing more than >90% of the major gene ontological categories in the mPFC of rats at 1 or 24 hrs post-injection (IV) with GLYX-13, ketamine, or saline (N=5 adult male rats at each time-point per treatment group). Equivalent aliquots of rat reference RNA (Stratagene, La Jolla, Ca) were treated concurrently with the tissue samples. Reverse transcription of 5 ug of RNA (primed with an oligo(dT) primer bearing a T7 promoter was followed by in vitro transcription in the presence of amino-allyl dUTP. The aRNA was denatured, hybridized, and washed with high stringency. Fluorescence hybridization was then quantified by a high resolution confocal laser scanner utilizing QuantArray software and analyzed using GeneTraffic (Iobion Informatics, La Jolla, Calif.). Statistical analysis was performed using the permutation-based Significance Analysis of Microarrays (SAM) algorithm using a false discovery rate of <10%. We utilized Database for Annotation Visualization and Integrated Discovery (DAVID) gene functional classification and gene functional annotation tables to examine interrelated genes within the gene list obtained using SAM.


qRT-PCR Array:


Reverse transcription of 1.0 μg of DNAsed, total RNA from 4 rats was primed with oligo(dT) and random hexamers. We utilized SuperScriptIII according to manufacturer's specifications (Invitrogen, Carlsbad, Calif.). A 1:10 dilution of cDNA was used as a template for quantitative real-time PCR, and the analysis was performed with Brilliant SYBR Green qRT-PCR Master Mix (Stratagene) on a Mx3000P Real-Time PCR System. ROX reference dye was included in all reactions. Experiments were performed in triplicate for each data point and transcript abundance was normalized to reference genes included in the rat Wnt PCR array (Qiagen, 330231).


Results:


As shown in FIG. 1, GLYX-13 and ketamine differentially affect gene expression patterns in the mPFC of the adult rat at 1 and 24 hrs post-injection (IV). In FIG. 1, the numbers represent the total number of genes that were shown to be significantly differentially expressed at 1 and 24 hrs following either GLYX-13 or ketamine injections (IV) as compared to vehicle control rats, using SAM analysis (FDR <10%). The sample size for each group in FIG. 1 was 5 adult male rats.


GLYX and ketamine showed a differential effect on the Wnt signaling pathway (Table 1 and FIG. 2). FIG. 2 shows Wnt signaling pathway gene expression in the mPFC following both GLYX-13 and ketamine injections at 1 hr (panel A) and 24 hrs (panel B). Using a commercially available rat Wnt qPCR array (Qiagen, 330231), a greater number of significant gene expression changes (p<0.05) were observed at 1 and 24 hrs post GLYX-13 injection relative to vehicle control rats when compared to the Wnt specific gene expression changes observed following ketamine injections. Fold change values greater than 0.0 indicate an upregulation in gene expression relative to saline vehicle controls, whereas those values less than 0.0 are genes whose expression was downregulated relative to vehicle controls. The sample size for each group in FIG. 2 was 4 rats. The significant reduction in the expression of Wnt11 in the mPFC at 1 hour post GLYX-13 injections suggest that the GLYX-13 mediated effect may, at least in part, involve a reduction in non-canonical Wnt signaling.


Notably, as shown in FIG. 3, GLYX-13 produced a greater number of changes in the Wnt pathway specific gene expression 1 and 24 hours post injection relative to vehicle control rats. Significant gene expression changes observed post-GLYX-13 injection are noted in dark gray, genes present on the qPCR array that did not significantly change are noted in light gray, and genes not present on the array are noted in medium gray. Significant gene expression changes following ketamine injections were observed for Frizzled 2 (Fzd).


Table 1. GLYX-13 and Ketamine differentially affect Wnt signaling pathway gene expression in the mPFC of the adult rat at 1 and 24 hours post injection (IV).









TABLE 1







Microarray Analysis of Wnt Signaling Pathway Gene Expression










GLYX-




13 vs.
Ketamine vs.



Vehicle
Vehicle












Gene Bank Accession No.
Gene Name
1 Hr
24 Hr
1 Hr
24 Hr





NM_024405
axin 1
G





M16112
calcium/calmodulin-dependent

V





protein kinase II beta


NM_001042354
calcium/calmodulin-dependent

G

K



protein kinase II beta


NM_012519
calcium/calmodulin-dependent

V

V



protein kinase II delta


NM_053615
casein kinase 1, alpha 1
V
G
V



NM_053824
casein kinase 2, alpha 1
V
V

V



polypeptide


NM_031021
casein kinase 2, beta subunit
V
G

K


NM_053357
catenin, beta 1
V
G
V
V


NM_153474
frizzled homolog 3



V


X73653
glycogen synthase kinase 3 beta

V




X07286
PKC, alpha

V
V



K03486, X04139
PKC, beta

G

K


L14323, M20636
PLC, beta 1
G
G
K
K


D90035, NM_017041
Protein Phosphatase 2B
G
G




M31809
Protein Phosphatase 3

G




NM_001100922
protein kinase, cAMP-dependent,

G





catalytic, alpha


XM_224987
secreted frizzled-related protein 1
V





AB017912
SMAD family member 2
G
V

V


XM_235639
Wnt 1
G
V




XM_237296
Wnt 10A


V
K


NM_053402
Wnt 4


V



XM_226051
Wnt 8A
V
V




NM_053738
Wnt inhibitory factor 1

V







Significance Analysis of Microarray Data (FDR < 10%).


G: indicative of higher levels of gene expression in GLYX-13 treated rats;


K: indicative of higher levels of gene expression in ketamine treated rats;


V: indicative of a higher level of expression in saline vehicle treated rats (downregulated in GLYX-13 or ketamine treated rats as indicated).


Genes present on the array that comprise the Wnt signaling pathway were defined using DAVID analysis.


N = 5 rats per group.






Data in Table 2 indicate the genes that were significantly differentially expressed in GLYX-13 and ketamine treated rats relative to either vehicle (GLYX-13 v Vehicle or Ketamine v Vehicle), or relative to each other (GLYX-13 v Ketamine) using significance analysis of microarrays (False Discovery Rate <10%). G: indicative of higher levels of gene expression in GLYX-13 treated rats; K: indicative of higher levels of gene expression in ketamine treated rats; V: indicative of a higher level of expression in saline vehicle treated rats (downregulated in GLYX-13 or ketamine treated rats as indicated). N=5 rats per group.









TABLE 2







Significance Analysis of Microarray Data for GLYX-13, Ketamine,


and vehicle control rats.


















Ketamine v
Glyx v













Glyx v Vehicle
Vehicle
Ketamine

















Gene Bank

1
24
2
1
24
2
1
24
2


Accession No.
Gene Name
Hr
Hr
wks
Hr
Hr
wks
Hr
Hr
wks





A03913
glia-derived neurite-
V


V








promoting factor











A17753
D3 receptor

V





K



AB000280
solute carrier family 15,




V


G




member 4











AB002801
cyclic nucleotide gated

V





K




channel alpha 3











AB003478
beta 1,3-
V

V
V

V






galactosyltransferase,












polypeptide 4











AB003991,
SNAP 25
G
G



K
G
G
K


AB003992












AB006137
galactoside 2-alpha-L-


G









fucosyltransferase











AB008682
FGF 17




V






AB010963,
calcium-activated potassium

G


K






AF020712
channel beta subunit











AB011679
tubulin, beta 5

V

K
K

K




AB013130
myozenin 3; synaptopodin

V


V

K
G



AB015946
tubulin, gamma 1

V

V

V





AB016160
GABA B receptor 1

V


V
K

G



AB017656,
mAChR 3

V


V

K
G



M16407,












M18088












AB017912
SMAD family member 2
G
V


V

G




AB018049
ST3 beta-galactoside alpha-

G


K
K






2,3-sialyltransferase 5











AB018546
stress-associated
V
V
V
V








endoplasmic reticulum












protein 1











AB020978
growth arrest and DNA-
V











damage-inducible, gamma











AF000423
synaptotagmin XI

V









AF000973
potassium

V
V


V






intermediate/small












conductance calcium-












activated channel, subfamily












N, member 1











AF001423
NMDAR 2A
V
V


V

K




AF003598
integrin, beta 7
V





K




AF003825,
GDNF family receptor alpha
V



V


G



U97143
2











AF005720
chloride channel 2

V


V






AF007583
acetylcholinesterase

V









AF007758,
synuclein, alpha

G
G


K

G



S73007












AF012347
SMAD family member 9

G
V








AF013144
dual specificity phosphatase
V
V
V
V
V







5











AF015728
cyclic nucleotide gated






K





channel beta 1











AF017637
carboxypeptidase Z
V





K




AF019973
enolase 2, gamma, neuronal

G
G

K






AF021935
CDC42 binding protein




V


G
G



kinase alpha











AF022819
potassium channel,




V


G




subfamily K, member 1











AF022935
prolactin

V









AF025670
caspase 6


G


K
G




AF025671
caspase 2

G


K


G



U77933












AF027984
calcium channel, voltage-

G


K



G



dependent, T type, alpha 1G












subunit











AF028784
glial fibrillary acidic protein






G




AF030086
neurotransmitter-induced

V
G

V
K

K




early gene 1 (ania-1)











AF030087
activity and




K


K




neurotransmitter-induced












early gene 2 (ania-2) mRNA











AF030088
homer homolog 1



K







AF030089
doublecortin-like kinase 1




K






AF030091
cyclin L1
V
V





K



AF030253
GABA vesicular transporter


G


K





AF030358
chemokine (C-X3-C motif)

G



K






ligand 1











AF031384
potassium channel,


G





G



subfamily K, member 3











AF031522
galanin receptor 3


G








AF032872
protein inhibitor of activated




V


G




STAT, 3











AF035632
syntaxin 12




V


G



AF037067
TNF superfamily, member 4

V


V






AF037071
NOS 1, neuronal
G


K
V
K
G
G
K


X59949












AF038571
solute carrier family 1

V





K




(neuronal/epithelial high












affinity glutamate












transporter, system Xag),












member 1











AF041244
hypocretin (orexin) receptor




K







1











AF041373
phosphatidylinositol binding






K





clathrin assembly protein











AF042499
SMAD family member 7




V






AF042713
neurexophilin 3
G



V






AF042714
neurexophilin 4
V










AF044581
syntaxin 12

G


K






AF044910
survival motor neuron 1

V


V






AF048828
voltage-dependent anion
V


V








channel 1











AF049239
sodium channel, voltage

V
G

V



G



gated, type VIII, alpha












subunit











AF049344
UDP-N-acetyl-alpha-D-

V


V

K





galactosamine:polypeptide












N-












acetylgalactosaminyl-












transferase 5 (GalNAc-T5)











AF049882
Cd82 molecule

V

V
V






AF050659
activity and

V




K





neurotransmitter-induced












early gene 7 (ania-7) mRNA











AF050660
activity and

V





K




neurotransmitter-induced












early gene 8 (ania-8) mRNA











AF050661
activity and




V







neurotransmitter-induced












early gene 9 (ania-9) mRNA











AF050663
activity and
G

G





G



neurotransmitter-induced












early gene 11 (ania-11)











AF052596
SNAP 23

V









AF054586
ring finger protein 112V
V




K
K




AF057308
hypoxia-inducible factor 1,

G
V

K







alpha subunit











AF058795
GABA B receptor 2

G


K
K


K


AF060879
neurocan

V









AF064541,
arginine vasopressin

V


V






D45400,
receptor 1B











U27322












AF076167
UDP-N-acetyl-alpha-D-





V


G



galactosamine:polypeptide












N-












acetylgalactosaminyltransfer-












ase 7 (GalNAc-T7)











AF078779
sodium leak channel, non-
G
G

K
K


G




selective











AF081366
potassium inwardly-rectifying







G




channel, subfamily J,












member 1











AF081557
glial cells missing homolog 1


G





G



(Drosophila)











AF087431
glucosidase 1




K






AF087453
potassium voltage-gated

V


V







channel, subfamily Q,












member 2











AF087454
potassium voltage-gated

V





K




channel, subfamily Q,












member 3











AF087839
ATP-binding cassette, sub-

V










family C (CFTR/MRP),












member 9











AF089730
potassium channel,

G










subfamily T, member 1











AF090113
glutamate receptor




V







interacting protein 2











AF096291
Bcl2-like 2; poly(A) binding






G





protein, nuclear 1











AF159803
Non-specific lipid-transfer




K







protein 6











AF191028
Xylem cysteine proteinase 2




V


G



AF205717
transmembrane 4 L six

V










family member 4











AF247559
Triosephosphate isomerase,
V
V










chloroplastic;












Triosephosphate isomerase











AF264018
transferase

V









AF459021
tubulin, beta 3
G
G
G
V
K
K
G




AJ000515
cyclic nucleotide gated
G
V




G





channel beta 1











AJ000556
Janus kinase 1
V



V

K




AJ001029
SRY (sex determining region
V


V








Y)-box 10











AJ002942
retinoic acid receptor, beta




V


G



AJ003065
potassium inwardly-rectifying




V







channel, subfamily J,












member 14











AJ006519
amiloride-sensitive cation




V







channel 2, neuronal











AJ006710
phosphoinositide-3-kinase,




V


G




class 3











AJ006855
synaptojanin 1
G

G


K





AJ007627,
potassium voltage-gated

V
V




K
K


AJ007628
channel, subfamily H (eag-












related), member 4











AJ007632
potassium voltage-gated

V


V







channel, subfamily H (eag-












related), member 8











AJ012603
ADAM metallopeptidase


V


V






domain 17











AJ222813,
interleukin 18




K


K



U77776












AJ277828
endothelial PAS domain

V


V







protein 1











AJ295749
xylosyltransferase II




V






D00634
adrenergic, beta-1-, receptor

G






G


D00698,
IGF 1
V
V

V

V
K
K
G


M15480,












M15481,












X06107












D00833,
glycine receptor, alpha 1

V


V






NM_013133












D10106
platelet-derived growth

G
V

K
V






factor alpha polypeptide











D12519
syntaxin 1A

G



K

G
K


D12524
v-kit Hardy-Zuckerman 4

V


V







feline sarcoma viral












oncogene homolog











D12573
hippocalcin
G



K






D13212,
NMDAR 2C

G


K
K
G




NM_012575,












U08259












D13417
hairy and enhancer of split 1





V

K
G


D13418
hairy and enhancer of split 3
G





G




D13871
solute carrier family 2

V


V







(facilitated glucose/fructose












transporter), member 5











D13985
chloride channel, nucleotide-
V


V








sensitive, 1A











D14048
heterogeneous nuclear

G
V
V
K

G





ribonucleoprotein U











D14480
calpain 8




V

K




D14869,
prostaglandin E receptor 3
V
V

V
V






D16443
(subtype EP3)











D17469
thyrotropin releasing




V







hormone receptor











D17521
chloride channel 3

V


V

G




D17764
synuclein, beta

G
G


K

G



D25224
similar to 40S ribosomal
V
V
G


K
K





protein SA











D25233
retinoblastoma 1

G
G

K

G




D25290
cadherin 6
V
V


V






D26111
chloride channel Kb

V









D26154
RB109



V


G




D28560
ectonucleotide


G


K






pyrophosphatase/phospho-












diesterase 2











D28562
solute carrier family 2

V


V







(facilitated glucose/fructose












transporter), member 5











D30666
acyl-CoA synthetase long-

G



K






chain family member 3











D32045,
adrenergic, alpha-1B-,
G



V






M60655
receptor











D32249
praja 2, RING-H2 motif

G
G

K

G

G



containing











D38380
signal recognition particle

G
V
V


G
G




receptor, B subunit;












transferrin











D44495
APEX nuclease


V
V
V
V
G
G




(multifunctional DNA repair












enzyme) 1











D45187
cathepsin E

V









D50093
prion protein

G





G



D50497
chloride channel 5




V


G



D63772
solute carrier family 1


V





K



(neuronal/epithelial high












affinity glutamate












transporter, system Xag),












member 1











D84450
ATPase, Na+/K+


G


K






transporting, beta 3












polypeptide











D86039
potassium inwardly rectifying

V


V







channel, subfamily J,












member 11











D87839
4-aminobutyrate

V


V

G





aminotransferase











D88672
phospholipase D2



K







D89655
scavenger receptor class B,


G





G



member 1












protein phosphatase 3











D90035,
(formerly 2B), catalytic
G
G
G


K
G
G
G


NM_017041
subunit, alpha isoform











D90048,
ATPase, Na+/K+
V
V
G

V

K
G



J04629
transporting, beta 2












polypeptide











D90258
proteasome subunit, alpha

G










type 3











E00988
IGF II

V


V






E01789
rat C-kinase type-II (beta-2)

G





G



E01884
rat IL-1-beta(interleukin-1







G




beta).











E05646
rat hippocampal cholinergic
V


V
V







neurostimulating











E12625
novel protein which is



V

V






expressed with nerve injury











E12746
Rat cholecystokinin-A

V










receptor gene











E13644
Neurodap-1
G
G




G

G


E13732
CC chemokine receptor

V


V







protein











J02722
heme oxygenase (decycling)
V











1











J03624
galanin prepropeptide

G

K


K




J03754
ATPase, Ca++ transporting,
V


V




K



plasma membrane 2











J03933
thyroid hormone receptor



V








beta











J04024
ATPase, Ca++ transporting,

G


K







cardiac muscle, slow twitch












2











J04532
PKC, zeta

G





G



J04563,
phosphodiesterase 4B,

V


V






M25350
cAMP specific











J04625
carboxypeptidase E

G


K

G




J04636
nACh receptor, beta 3

G





G



J04731
potassium voltage-gated
G

G



G

G



channel, shaker-related












subfamily, member 2











J04811
growth hormone receptor

V


V






J04963
carcinoembryonic antigen-
V
V










related cell adhesion












molecule 1 (biliary












glycoprotein)











J05107
hydroxysteroid 11-beta
V

V





K



dehydrogenase 1











J05122
translocator protein
V
V




K
K



J05510
IP3 receptor, type 1

G
G





G


K00512
myelin basic protein
G





G




K01701
oxytocin, prepropeptide
G


K







K03486
PKC, beta

G
G

K
K





X04139












L02926
interleukin 10

V


V






L04535
somatostatin receptor 5



V

V





L04684
calcium channel, voltage-




V







dependent, L type, alpha 1S












subunit











L04739
ATPase, Ca++ transporting,
G
G



K
G

K



plasma membrane 1











L04796
glucagon receptor

V


V

G
K



L05435
synaptic vesicle glycoprotein

G



K

G




2a











L05596
serotonin receptor 1F
G





G




L06894
platelet-derived growth

V


V







factor alpha polypeptide











L08228,
NMDAR 1
G
G
V
K


G




X63255












L08492
GABA A receptor, alpha 3
V


V



G



L08493
GABA A receptor, alpha 4







G



L08497
GABA A receptor, gamma 2

G



K

G



L09119
neurogranin

G



K


K


L09120
calpain 2

V









L10072
serotonin receptor 5A

V


V
K





L10326
GNAS complex locus





K
G




L14323,
PLC, beta 1
G
G
G
K
K






M20636












L14851
neurexin 3
V


V
V


G



L16532
2′,3′-cyclic nucleotide 3′



V


G





phosphodiesterase











L16764
heat shock 70 kD protein 1B



K


K




L18889
calnexin




V






L20821
syntaxin 4

V









L20822
syntaxin 5
V
V

V
V


G



L21192,
GAP 43
G
G
G

K
K
G
G
K


NM_017195












L22339
sulfotransferase family,





V






cytosolic, 1C, member 3











L23088
selectin, platelet



V







L24907,
calcium/calmodulin

G


V
K

G
K


NM_134468
dependent protein kinase I











L25925
prostaglandin-endoperoxide

V





K




synthase 2











L27059
phosphodiesterase 4D,




V







cAMP-specific











L27487
calcitonin receptor-like V
V


V

K





L31546
serotonin receptor 2A
G
G


K
K





L31620
nAChR, alpha 4



K







L31622
nAChR, beta 2




K






L31771
adrenergic, alpha-1D-,

V





G



M60654
receptor











L36884
protein tyrosine

V


V







phosphatase, receptor type,












V











L41623
platelet-derived growth

V










factor beta polypeptide











M10088
prodynorphin



K
V

K




M10244
tyrosine hydroxylase




K






M11794
metallothionein 2A
V
V

V







M12492
protein kinase, cAMP

G


K



K



dependent regulatory, type II












beta











M15191
tachykinin 1

G









M15880
neuropeptide Y

G


K






M16112
calcium/calmodulin-

V










dependent protein kinase II












beta











M16406
mAChR 1

G




G




M16409
mAChR 4





K





M17069,
calmodulin

G


K
V

G



X13817,












X14265












M17526
G protein alpha activating
G




K
G

K



activity polypeptide O











M18416
early growth response 1
G


K
K


K



M19257
retinol binding protein 1,
G











cellular











M19533
peptidylprolyl isomerase A
V
G


K






M20133
androgen receptor
G

G



K

G


M20636
PLC, beta 1
V










M21060,
SOD 1
V
G
V

K
V





M25157,












Y00404












M22253
sodium channel, voltage-

G

K



G




gated, type I, alpha











M22254
sodium channel, voltage-

G
G


K

G




gated, type II, alpha 1











M22357
myelin-associated
G
G



K






glycoprotein











M22412
potassium voltage-gated

V





K




channel, Isk-related












subfamily, member 1











M23601
monoamine oxidase B


V





K


M23697
plasminogen activator,








K



tissue











M24104
vesicle-associated

V



K






membrane protein 1











M24852
Purkinje cell protein 4

G


K

G




M25638
neurofilament, light

G





G




polypeptide











M25646
arginine vasopressin




V
V





M25888
proteolipid protein 1
G
G



K
G
G
K


M25890
somatostatin

G


K



K


M26161
potassium voltage-gated
G
G




G





channel, shaker-related












subfamily, member 1











M26715
phosphodiesterase 4A,

G





G




cAMP-specific











M26744,
interleukin 6

V

V
V






M26745












M27293
IGF 1 receptor




V






M27925
synapsin II

G





G



M30312
potassium voltage-gated

V
G

V
K






channel, shaker-related












subfamily, member 3











M31174,
thyroid hormone receptor

G


K

G




X12744
alpha











M31176
gastrin releasing peptide




V


G



M31178
calbindin 1

G

K
K






M31433





V


G



M31809
protein phosphatase 3,

G
G


K

G




catalytic subunit, beta












isoform











M32061
adrenergic, alpha-2B-,

V


V







receptor











M34253
interferon regulatory factor 1

V


V






M34445
glutamate decarboxylase 1

G

K
K
K





M35162,
GABA A receptor, delta


G



K

G


NM_017289












M36074
nuclear receptor subfamily 3,




V


G




group C, member 2











M36831,
D2 receptor
V

V
V


G




X56065,












NM_012547












M37394
epidermal growth factor

V


V







receptor











M38061
AMPA 2 receptor
G
G
G

K
K
G
G



M55291
neurotrophic tyrosine kinase,
G

G

V
K
G
G




receptor, type 2











M57664
creatine kinase, brain

G


K






M58040
transferrin receptor

G


K

G




M58316
adrenergic, alpha-2C-,
G






G



X57659
receptor











M58634
IGF binding protein 1

V




K




M59313,
potassium voltage gated

G
V

V
K


K


X62839
channel, Shaw-related












subfamily, member 2











M59980
potassium voltage gated

V










channel, Shal-related family,












member 2











M60525,
VGF nerve growth factor
G
G

K
K



K


M74223
inducible











M60753
catechol-O-
V


V








methyltransferase











M61099
mGluR 1

G









M61177,
MAPK 3
V



K






NM_017347












M62781
IGF binding protein 5

V


V






M63101
IL 1 receptor antagonist




V


G



M63122
tumor necrosis factor

V





K




receptor superfamily,












member 1a











M64300
MAPK 1




V


G



M68971
hexokinase 2





K

K



M69055
IGF binding protein 6
V
V





K



M77482
potassium voltage gated

V










channel, Shab-related












subfamily, member 2











M77809
transforming growth factor,




V

G
G




beta receptor III











M80545
calcium channel, voltage-
V











dependent, beta 2 subunit











M81783
potassium voltage-gated




V


G




channel, subfamily F,












member 1











M82824
NF1



K
V






M84009,
D4 receptor
G
V
G
K
V



G


NM_012944












M84725
transgelin 3

G



K

G



M85035
AMPA 2
V
V

V
V






M85183
NLR family, pyrin domain






G





containing 6











M86389,
heat shock protein 1
V
V
V

K

K
K



S45392,












X54793












M86621
calcium channel, voltage-
G
G



K
G
G
K



dependent, alpha2/delta












subunit 1











M86742
neurotrophin 4

V









M86835
vasoactive intestinal peptide



V

K
G

K



receptor 1











M88096
cholecystokinin A receptor

V









M88751
calcium channel, voltage-




V







dependent, beta 3 subunit











M89924
calcium channel, voltage-


G


K






dependent, L type, alpha 1C












subunit











M89953
serotonin receptor 1D

V


V






M89954,
serotonin receptor 1B




K
K





X62944












M91466
adenosine A2B receptor

V









M91599
FGFR 4

V









M91652
glutamate-ammonia ligase

G


K
K


K



(glutamine synthetase)











M91808
sodium channel, voltage-




V


G




gated, type I, beta











M92075
mGluR 2
G
G


K






M92076
mGluR 3




V






M92905
calcium channel, voltage-
G
G




G





dependent, N type, alpha 1B












subunit











M95735
syntaxin 1B



K


K




M95762
solute carrier family 6



K








(neurotransmitter












transporter, GABA), member












13











M96375
neurexin 1



K
K
K





M96376
neurexin 2

G


K
K





M96853
discs, large homolog 4





K






(Drosophila)











M98820
interleukin 1 beta

V


V






NM_001003
DNA (cytosine-5-)-
V

V
V

V





964
methyltransferase 3-like











NM_001007
catenin (cadherin associated
V

V

K

K
K



145
protein), alpha 1











NM_001008
eukaryotic translation

G
G

K
K





335
initiation factor 4A2











NM_001017
eukaryotic translation

G


K


G



374
initiation factor 4, gamma 2,












pseudogene 1











NM_001024
catenin, beta like 1




K


K



870












NM_001024
exocyst complex component

G
V





K


964
3











NM_001033
eukaryotic translation

V


V
V





069
initiation factor 4E binding












protein 2











NM_001033
casein kinase 1, gamma 2


V








870












NM_001042
calcium/calmodulin-

G


K






354
dependent protein kinase II












beta











NM_001100
eukaryotic translation





V

G
G


158
initiation factor 4A, isoform 3











NM_001100
protein kinase, cAMP-

G









922
dependent, catalytic, alpha











NM_001104
Eph receptor B1
G
G



K

G
K


528












NM_001106
AKT1 substrate 1 (proline-
G





G




259
rich)











NM_001106
catenin (cadherin associated

G


K
K





598
protein), alpha 2











NM_001106
eukaryotic translation

G


K
K





693
initiation factor 4 gamma, 3











NM_001106
chromobox homolog 5 (HP1
G










797
alpha homolog, Drosophila)











NM_001107
adaptor-related protein

G


K
K





511
complex 2, alpha 1 subunit











NM_001108
SHC (Src homology 2

G


K






065
domain containing)












transforming protein 2











NM_001108
protein phosphatase 2A

G
V

K



K


577
activator, regulatory subunit












4











NM_001108
eukaryotic translation







K



808
initiation factor 4E member 2











NM_001109
c-fos serum response

G









302
element-binding












transcription factor











NM_001130
Transforming protein p21

G


K


G



441












NM_001134
regulatory associated




K
K


K


499
protein of MTOR, complex 1











NM_001135
son of sevenless homolog 2

G
V

K
V





561












NM_012512
beta-2 microglobulin
V

V

K
V
K




NM_012519
calcium/calmodulin-

V


V







dependent protein kinase II












delta











NM_012528
nAChR, beta 1 (muscle)

V









NM_012734
hexokinase 1

G


K
K
G




NM_012752
CD24 molecule




K



K


NM_012813
ST8 alpha-N-acetyl-




V


G




neuraminide alpha-2,8-












sialyltransferase 1











NM_012821
adenylate cyclase 6





K





NM_012957,
GABA A receptor, beta 2
G
G




G




X15467












NM_013058
inhibitor of DNA binding 3
V





K




NM_013060
inhibitor of DNA binding 2

G


K
K





NM_013180
integrin beta 4

V


V






NM_013216
Ras homolog enriched in
G
G
V

K
K


K



brain











NM_017022
integrin beta 1 (fibronectin
V



K

K





receptor beta)











NM_017035
PLC, delta 1
G





G




NM_017066
pleiotrophin

G

K
K
K
K

K


NM_017078
nAChR, alpha 5



K
V






NM_017093
v-akt murine thymoma viral




V







oncogene homolog 2











NM_017125
Cd63 molecule
V

V



K
K



NM_017291
GABA receptor, rho 1




V






NM_019218
neurogenic differentiation 1

G



K

G



NM_019220
amino-terminal enhancer of

G


K







split











NM_019248
neurotrophic tyrosine kinase,








K



receptor, type 3











NM_019361
activity-regulated

V


V
K


K



cytoskeleton-associated












protein











NM_021576
5′ nucleotidase, ecto

V


V



G


NM_021835,
Jun oncogene





V





X17163












NM_021836
jun B proto-oncogene
V
V




K
K



NM_022282
discs, large homolog 2







K




(Drosophila)











NM_022855
casein kinase 1, gamma 3

G


K
K





NM_022952
adaptor-related protein


V









complex 2, sigma 1 subunit











NM_024405
axin 1
G

V








NM_031007
adenylate cyclase 2
G


K



G



NM_031008
adaptor-related protein
G

V



G





complex 2, alpha 2 subunit











NM_031021
casein kinase 2, beta
V
G


K







subunit











NM_031338
calcium/calmodulin-





K






dependent protein kinase












kinase 2, beta











NM_031515,
v-Ki-ras2 Kirsten rat
V
V


V






U09793
sarcoma viral oncogene












homolog











NM_031527
protein phosphatase 1,

G
V

K







catalytic subunit, alpha












isoform











NM_031575
v-akt murine thymoma viral
V
V


V

K





oncogene homolog 3












(protein kinase B, gamma)











NM_031590
WNT1 inducible signaling

V





K




pathway protein 2











NM_031622
MAPK 6
V
G









NM_031639
discs, large homolog 3


V

K


K
K



(Drosophila)











NM_031662
calcium/calmodulin-

G



K






dependent protein kinase












kinase 1, alpha











NM_031985
ribosomal protein S6 kinase,




K







70 kDa, polypeptide 1











NM_032058
eukaryotic translation
V
G
V


V

G




initiation factor 2B, subunit 2












beta











NM_033230
v-akt murine thymoma viral


V

K


K




oncogene homolog 1











NM_053346,
neuritin 1
G
G


K
K
G

K


U88958












NM_053351
calcium channel, voltage-
G
G

K

K

G
K



dependent, gamma subunit












2











NM_053354
DNA (cytosine-5-)-
G



K
K

K




methyltransferase 1











NM_053357
catenin (cadherin associated
V
G
V
V
V

K
G




protein), beta 1











NM_053400
transducin-like enhancer of
V





K





split 3 (E(sp1) homolog,












Drosophila)











NM_053402
wingless-type MMTV



V


G





integration site family,












member 4











NM_053615
casein kinase 1, alpha 1
V
G

V



G



NM_053649
kringle containing


V

V
V

G




transmembrane protein 1











NM_053698
Cbp/p300-interacting



K
K
V
K
K




transactivator, with Glu/Asp-












rich carboxy-terminal












domain, 2











NM_053738
Wnt inhibitory factor 1

V









NM_053824
casein kinase 2, alpha 1
V
V


V







polypeptide











NM_053837
adaptor-related protein

G
V









complex 2, mu 1 subunit











NM_053857
eukaryotic translation
V

V


V

K
G



initiation factor 4E binding












protein 1











NM_053861
tenascin C





V





NM_080394
reelin

G


K






NM_080583
adaptor-related protein

G


K







complex 2, beta 1 subunit











NM_130779
adenylate cyclase 3
G



K






NM_133605
calcium/calmodulin-





K


K



dependent protein kinase II












gamma











NM_133609
eukaryotic translation
G











initiation factor 2B, subunit 3












gamma











NM_138905
phosphatidic acid
G
G


K

G





phosphatase type 2B











NM_139060
casein kinase 1, delta

G
G

K
K





NM_153474
frizzled homolog 3


V

V



K


NM_172029
eukaryotic translation

V


V

G
G




initiation factor 2B, subunit 1












alpha











NM_173331
MAPK 15

V









NM_173337
calcium/calmodulin-

G
G


K
G
G




dependent protein kinase II












inhibitor 1











NM_182842
calcium/calmodulin-

G





G




dependent protein kinase IG











NM_199372
eukaryotic translation




K







initiation factor 4A1











S42358
neurotransmitter transporter,

G










GABA, member 11











S44606
beta-integrin
V

V



K




S48813
adrenergic, beta, receptor

G


K







kinase 1











S49491
proenkephalin 1
G
G



V





S53527
S100 calcium binding
G
G


K

G
G




protein B











S54008
FGFR 1





K





S59158,
glial high affinity glutamate
G
G


K
K
G




X63744
transporter, member 3











S62043
serotonin receptor 6

V









S67770
transforming growth factor,

V


V







beta receptor II











S68944
solute carrier family 6








G



(neurotransmitter












transporter), member 17











S70690,
cholecystokinin

G


K
K


K


X01032












S71570
calcium/calmodulin-





K


K



dependent protein kinase II












gamma











S72505,
glutathione S-transferase A3




V

G




X78848












S75952
glucagon-like peptide 1




V







receptor











S76145
neurotransmitter transporter,


G


K






dopamine, member 3











S76779
apolipoprotein E
G
G



K
G




S77528,
CCAAT/enhancer binding
V
V
V
K


K
K



X60769
protein (C/EBP), beta











S79263
colony stimulating factor 2




V

K
G




receptor, beta, low-affinity












(granulocyte-macrophage)











S82649,
neuronal pentraxin 2

V


V
K





XM_221901












S83194
calcium/calmodulin-

G



K

G
K



dependent protein kinase












kinase 1, alpha











U03390
G protein, beta polypeptide
V
V
V


V
K





2 like 1











U04738
somatostatin receptor 4




V


G



U05784
microtubule-associated

G


K







protein 1 light chain 3 beta











U08141
Ferritin light chain 2




V


G



U08255
glutamate receptor,
V
G
V









ionotropic, delta 1











U08256
glutamate receptor,
V
V





K




ionotropic, delta 2











U08260
NMDAR 2D

G









U08290
neuronatin

G


K
K





U10071
CART prepropeptide



V







U11031
contactin 3

V


V


K



U12336
nAChR, alpha 9





K


K


U13368
adrenergic, alpha-1A-,

V



K






receptor











U14533
nuclear receptor subfamily 1,
V



K







group H, member 2











U15211
retinoic acid receptor, alpha

V









U17254
nuclear receptor subfamily 4,



K
K

K
K




group A, member 1











U17607
nuclear transcription factor-Y





V






gamma











U18650
huntingtin

V


V



G


U18982
FBJ osteosarcoma





K






oncogene B











U20105
synaptotagmin VI

V





K



U20283
syntaxin binding protein 2
V










U21662
mannosyl (alpha-l,6-)-
V
G


K







glycoprotein beta-1,2-N-












acetylglucosaminyl-












transferase











U22414
chemokine (C—C motif)
V











ligand 3











U26402
synaptotagmin V

V


V






U26541
PDGFA associated protein 1

G

V







U29701
4-aminobutyrate

G



K

G




aminotransferase











U30290
galanin receptor 1
V
V


V

K




U30938,
microtubule-associated

G


K
K


K


X53455
protein 2











U31203
noggin







G



U31554
limbic system-associated

G


V


G




membrane protein











U33472
serine/threonine kinase 10

V

V



K



U35365
FYN oncogene related to




V







SRC, FGR, YES











U37058
neuromedin B receptor




V






U37142
brevican
V
V




K




U38306
arylalkylamine N-

V





K




acetyltransferase











U38653
IP3 receptor, type 1

V


V






U48829,
NOS 2

V
V

V

K




S71597,












U03699,












U16359












U49729
Bcl2-associated X protein
V










U49953
p21 protein (Cdc42/Rac)-


G


K






activated kinase 1











U50147
discs, large homolog 3




V


G




(Drosophila)











U50194
tripeptidyl peptidase II

G

K







U52948
complement component 9



V







U53927
solute carrier family 7
V




K
K





(cationic amino acid












transporter, y+ system),












member 2











U57715
FGF receptor activating
V
V

V
V







protein 1











U59809
IGF 2 receptor
G

V


V
G




U61696
arginyl aminopeptidase



V
V

G
G




(aminopeptidase B)











U63740
fasciculation and elongation
V
G



K

G




protein zeta 1 (zygin I)











U67140
discs, large homolog-

G
V




G




associated protein 4











U72350
similar to Bcl2-like 1 isoform



V








3; Bcl2-like 1











U72353
lamin B1

V









U73142
MAPK 14
G
V




G




U73859
hexokinase 3

V




G




U75899
heat shock protein 2

V


V






U81492
interleukin 3
V
V

V
V






U83112
forkhead box M1
V
G
V




G



U88036
solute carrier organic anion

G





G




transporter family, member












1a4











U88324,
G protein, beta polypeptide
V
G
V
V
K
K


K


AF022083
1











U92469
gonadotropin releasing
V





K





hormone receptor











U92655
potassium voltage-gated
G
G

K
K







channel, subfamily Q,












member 1











V01217
actin, beta
V
G


K

K




V01227
tubulin, alpha
V
G
V
V
K

G
G



X00336
interferon alpha family

G



K


K


X00911,
IGF 2
G
G


K
K
G

K


X16703,












X17012












X01454
thyroid stimulating hormone,




V
K






beta











X02231
GAPDH
V
V
G
V
V

K
G
G


X03347
FBR-murine osteosarcoma
V











provirus genome











X03475
ribosomal protein L35a
V





K




X04979
apolipoprotein E


V
K


K




X06554
myelin-associated

V










glycoprotein











X06769
FBJ osteosarcoma


V

K
V
K
K




oncogene











X06827
hydroxymethylbilane
V
V
V




K




synthase











X06942
v-raf murine sarcoma 3611


V









viral oncogene homolog











X07286
PKC, alpha

V

V







X07467
glucose-6-phosphate




K







dehydrogenase











X07729
enolase 2, gamma, neuronal

V









X13016
Cd48 molecule
V
G
V

K
V
K
K



X13804
neurofilament, heavy

G










polypeptide











X15013
ribosomal protein L7a
V




V


G


X17184
AMPAR 1

V


V
K


K


X17621
potassium voltage gated

V


V







channel, shaker related












subfamily, member 6











X51992
GABA A receptor, alpha 5



K


K
G



X55812
CB1 receptor
G
G


K
K


K


X56917
IP3 3-kinase A

G



K

G
K


X57514
GABA A receptor, gamma 1
V





K




X58149
Phosphoribulokinase,







G




chloroplastic











X61159
glycine receptor, alpha 2

G


K

K




X62085
hypoxanthine

G

V
K


G




phosphoribosyltransferase 1











X62295
angiotensin II receptor, type

V










1a











X62314
somatostatin receptor 1



K



G



X62840
potassium voltage gated
V

G



K





channel, Shaw-related












subfamily, member 1











X62841
potassium voltage gated


G





G



channel, Shaw-related












subfamily, member 4











X62952
vimentin
V
V
G



K
K
G


X63143
syndecan 3





V





X63995
solute carrier family 6




V



K



(neurotransmitter












transporter, serotonin),












member 4











X66842
serotonin receptor 2B

V









X66870
lamin A
V
V

V
V
V
G
G



X69903
interleukin 4 receptor, alpha




V






X70521
sodium channel, nonvoltage-





K






gated, type I, alpha











X70662
potassium voltage-gated







G




channel, shaker-related












subfamily, beta member 1











X73653
glycogen synthase kinase 3

V










beta











X73683
similar to H3 histone, family
V
V
V



K
K




3B











X74833
cholinergic receptor,

V










nicotinic, beta 1 (muscle)











X76489
CD9 molecule
V


V


K




X78606
RAB28, member RAS

G


K







oncogene family











X79321
microtubule-associated

G


K







protein tau











X83094
heat shock transcription
G




K






factor 1











X83580,
potassium inwardly-rectifying
G
G


K
K
G
G
K


X87635
channel, subfamily J,












member 4











X83585
potassium inwardly-rectifying





K






channel, subfamily J,












member 10











X86789
synuclein, gamma (breast





V






cancer-specific protein 1)











X91810
signal transducer and

V

V
V







activator of transcription 3











X92070
purinergic receptor P2X,




V


G




ligand-gated ion channel, 6











X95096
Macrophage stimulating 1





V


G



(hepatocyte growth factor-












like)











X95466
spectrin repeat containing,
G
G


K
K
G

K



nuclear envelope 1











X95579
GABA receptor, rho 1

V


V
K





X96488
mitogen-activated protein




V


G




kinase 12











X97121
neurotensin receptor 2

V





K



X97374
prepronociceptin






G




X98564
potassium channel,





K






subfamily V, member 1











XM_001060
eukaryotic translation





K


K


756
initiation factor 4 gamma, 1











XM_001066
eukaryotic translation
V










554
initiation factor 4E family












member 1B











XM_217346
PTK7 protein tyrosine kinase
V
G




K
G




7











XM_217785
Rattus norvegicus TATA box

V










binding protein (Tbp),











XM_224987
secreted frizzled-related
V

V









protein 1











XM_226051
wingless-type MMTV
V
V










integration site family,












member 8A











XM_234422
c-fos
V
V
V
V

V





XM_235454
forkhead box H1

V


V


K



XM_235639
wingless-type MMTV
G
V




G





integration site family,












member 1











XM_237295
wingless-type MMTV








K



integration site family,












member 6











XM_237296
wingless-type MMTV



V
K


K




integration site family,












member 10A











XM_575489
neurogenic differentiation 6

G





G



Y11433
pyrimidinergic receptor P2Y,

V










G-protein coupled, 4











Y14635
amiloride-sensitive cation








K



channel 1, neuronal











Y16563
bassoon




V


G



Z11558
glia maturation factor, beta
G
V




G




Z12152
neurofilament, medium

V


V
K






polypeptide











Z24721
SOD 3, extracellular


V

V
V












Example 2
GLYX-13 Induces Rapid Antidepressant-Like Effects without Dissociative Side Effects

The present study examined GLYX-13 for its potential as a clinically relevant antidepressant using multiple rat models of depression, and tested for ketamine-like side effects in rats. The study also examined whether the antidepressant-like effects of GLYX-13 required AMPA glutamate receptor activation, and whether GLYX-13 could facilitate metaplasticity.


Methods:

Behavioral Pharmacology: Male Sprague-Dawley (SD) rats (2-3 Months old) were given injections of GLYX-13 (1-56 mg/kg IV; 1-100 mg/kg SC; 0.1-10 μg MPFC), ketamine (10 mg/kg IV; 0.1-10 μg MPFC), fluoxetine positive control (three doses at 10 mg/kg SC) or sterile 0.9% saline vehicle, either 20-60 min or 24 hrs before Porsolt testing. Pretreatment with NBQX (10 mg/kg IP) was used to test the role of AMPAR in the antidepressant-like effect of GLYX-13 (3 mg/kg IV) in the Porsolt test. Antidepressant-like drug effects were measured by decrease in floating time in the Porsolt test, decreased feeding latency in a novel but not familiar environment for the novelty-induced hypophagia (NIH) test, and decreased number of escape failures in the learned helplessness (LH) test. Ketamine like abuse potential and reward was measured by ketamine-like responding in drug discrimination testing and time spent in the drug paired side in the conditioned place preference assay. Ketamine-like disruptions in sensory-motor gating were measured by decreased pre-pulse inhibition. Ketamine-like sedation was measured by decreases in open field locomotor activity and operant response rate in a drug discrimination study. Molecular Pharmacology: Adult male SD rats were dosed with GLYX-13 (3 mg/kg IV), ketamine (10 mg/kg IV) or saline vehicle and sacrificed 24 hrs post dosing. MPFC and hippocampal slices were prepared, and cell surface expressing proteins were cross-liked by biotinylation. Cell surface expression of GluR1 and NR2B were measured by Western blot. Electrophysiology: Hippocampal slices were prepared from adult male SD rats 24 hours after a single injection of GLYX-13 (3 mg/kg IV), ketamine (10 mg/kg IV) or vehicle. LTP at Schaffer collateral-CA1 synapses was measured in response to three submaximal bouts of high-frequency Schaffer collateral stimulation (2×100 Hz/800 ms). The percent contribution of NR2B and NR2A-containing NMDARs to pharmacologically isolated total NMDAR conductance were measured in Schaffer collateral-evoked EPSCs of CA1 pyramidal neurons by using the NR2B-selective NMDAR antagonist ifenprodil (10 μM), and the NR2A-NMDAR selective antagonist NVP-AM077 (100 nM).


Results:

As shown in FIG. 4, GLYX-13 produces antidepressant-like effects in multiple rat models. The data were collected as described in the Methods and below.


Porsolt Test: 2-3 month old Sprague Dawley (SD) rats treated with a single dose of GLYX-13 (TPPT-NH3; 1-56 mg/kg, IV), scrambled GLYX-13 (PTTP-NH3; 3 mg/kg, IV), ketamine (10 mg/kg, IP), 3 doses of fluoxetine (20 mg/kg SC; 24, 5, and 1 hr before testing; (Detke et al., 1995)), or sterile saline vehicle (1 ml/kg, IV) 30-60 min before testing, or a single dose of GLYX-13 (3 mg/kg, IV), ketamine (10 mg/kg, IV) or 3 doses of fluoxetine (20 mg/kg SC; last dose 24 hrs before testing) or saline vehicle treated rats tested 24 hrs post dosing. NIH test: latency to eat in the novelty induced hypophagia (NIH) test in SD rats dosed with GLYX-13 (3 mg/kg, IV), ketamine (10 mg/kg, IV) or saline and tested 1 hr post dosing. LH test: escape failures in the footshock induced learned helplessness (LH) test in SD rats dosed with single dose of GLYX-13 (3 mg/kg IV; 24 hrs before testing), 3 doses Fluoxetine (20 mg/kg SC; last dose 1 hr before testing), or sterile saline vehicle (1 ml/kg IV; tail vein) 24 hrs before testing. Naïve control animals did not receive pre-shock or injection before LH testing. USVs test: Hedonic and Aversive USVs in adult SD rats receiving 2 min of heterospecific play (alternating blocks of 15 sec stimulation followed by 15 sec no stimulation). Data expressed as Mean (±SEM). N=7-21 per group. P<0.05 Fishers PLSD post hoc test vs. vehicle.


Results of the various tests presented in FIG. 5 demonstrate that GLYX-13 does not show ketamine-like addictive sensory motor gating, or sedative side effects. The data were collected as described in the Methods and below.


Drug discrimination: Percentage ketamine-lever responding and for different doses of ketamine (IP and SC) and GLYX-13 (SC) in SD rats trained to discriminate 10 mg/kg ketamine (Ket), IP, from saline (Sal). Values above Sal and Ket are the results of control tests conducted before testing each dose response curve. Place Preference: Ketamine (10 mg/kg IV) but not GLYX-13 (10 mg/kg IV) induced conditioned place preference as measured by % time in drug paired chamber. Prepulse Inhibition: Ketamine (10 mg/kg IP) but not GLYX-13 (10 mg/kg IV) decreased sensory-motor gating as measured by prepulse inhibition. Open field: A sedating dose of ketamine (10 mg/kg SC) but not GLYX-13 (10 mg/kg IV) reduced locomotor activity in the open field as measured by line crosses. N=8-11 per group. Data are expressed as Mean (±SEM). *P<0.05 Fishers PLSD post hoc test vs. vehicle.


As indicated in FIG. 6, injection of GLYX-13 into the prefrontal cortex shows antidepressant-like effects in the Porsolt test. The data were collected as described below.


Mean (±SEM) time (sec) spent immobile in the Porsolt test in 2-3 month old male rats implanted with (a) medial prefrontal or motor cortex (dorsal control) cannulae and injected with GLYX-13 (0.1, 1, 10 μg/side) or sterile saline vehicle (0.5 μL/1 min) and tested 1 hr post dosing or rats given MPFC injections of ketamine (0.1, 1, 10 μg), GLYX-13 (1 μg), or saline and tested 20 min and 24 hrs post dosing. Animals received a 15 min training swim session one day before dosing. Mean (±SEM) line crosses in the open field 20 min following MPFC infusion of GLYX-13 (1 μg), ketamine (0.1 μg) or sterile saline vehicle. Given that 0.1 μg dose of ketamine increased locomotor activity, the Porsolt data for that dose were not included in the analysis given that increasing locomotor activity produces a false positive antidepressant-like response. A representative H&E stained section depicting MPFC cannulae placemen, arrow indicates injection site. N=5-10 per group. *P<0.05, Fisher PLSD vs. vehicle


The data in FIG. 7 demonstrate that GLYX-13 induces NR2B-dependent synaptic plasticity. The data were collected as described in the Methods and below.


ex vivo cell surface protein levels: Biotinylated cell surface GluR1 protein levels in the medial prefrontal cortex (MPFC) or hippocampus as measured by western blot in SD rats treated with GLYX-13 (3 mg/kg IV) ketamine (10 mg/kg IV) or sterile saline vehicle 24 hours prior to sacrifice. ex vivo NMDAR current: NMDA receptor-dependent single shock-evoked EPSCs in the presence of the NR2B-selective NMDA receptor antagonist ifenprodil (10 μM), in CA1 pharmacological isolated NMDA current in rats that were dosed with GLYX-13 (3 mg/kg IV) ketamine (10 mg/kg IV) or sterile saline vehicle (IV) 24 hrs before ex-vivo NMDA current measurement. ex vivo LTP: GLYX-13 (3 mg/kg IV) or ketamine (10 mg/kg IV) 24 hrs post dosing enhances the magnitude of ex vivo long-term potentiation (LTP) of synaptic transmission at Schaffer collateral-CA1 synapses. Data are expressed as Mean (±SEM). N=5-11 per group. *P<0.05, **P<0.01 Fishers PLSD post hoc test vs. vehicle.


In FIG. 8, the data show that the antidepressant-like effects of GLYX-13 are synaptic-plasticity related. The data were collected as described in the Methods and below.


Ex vivo cell surface protein levels: Biotinylated cell surface GluR1 protein levels in the medial prefrontal cortex (MPFC) or hippocampus as measured by western blot in SD rats treated with GLYX-13 (3 mg/kg IV) or sterile saline vehicle 24 hours prior to sacrifice. AMPAR antagonism: Mean (±SEM) Floating time in the Porsolt test in animals pretreated with the AMPA receptor antagonist NBQX (10 mg/kg IP) before GLYX-13 (3 mg/kg IV) dosing and tested 1 hr post dosing.


Summary

In total, the data show that (i) GLYX-13 produces a robust antidepressant-like effect without dissociative side effects; and (ii) GLYX-13 produce an antidepressant-like effect by facilitating synaptic plasticity in the MPFC.


Example 3
GLYX-13 Increases Ex Vivo [3H] MK-801 Binding, a Non-Competitive Antagonist of the NMDA Receptor, in the Rat Medial Prefrontal Cortex 1 Hour Post Dosing


FIG. 9 shows that ex vivo [3H] MK-801 binding in the rat medial prefrontal cortex increases one hour after dosing with GLYX-13. The data were collected as described below.


Mean±SEM specific [3H] MK-801 binding (5 nM; 22.5 Ci/mmol) to well washed rat MPFC membranes (200 μg) in 2-3 month old Male SD rats treated with GLYX-13 (3 mg/kg IV) or sterile saline vehicle (1 ml/kg tail vein) and decapitated without anesthesia 1 hr post dosing, and brain rapidly removed (60 sec), frozen on dry ice, and stored at −80° C. until assay. [3H]MK-801 binding for was measured under equilibrium conditions (2 hrs) in the presence 1 mM glycine. Non-specific binding was were determined in the absence of any glycine ligand and in the presence of 30 μM 5,7 DCKA. Maximal stimulation was measured in the presence of 1 mM glycine. 50 μM glutamate was present in all reactions. n=5-6 per group. *P<0.05 vs. respective vehicle.


Example 4
Rapid Antidepressant Effects of GLYX-13 May be Mediated by an E-LTP-Like Mechanism

To examine the rapid-acting effects of GLYX-13, the biochemical processes that underlie the induction of early stage long term potentiation (E-LTP) were studied.


Without wishing to be bound by any theory, E-LTP is dependent upon the persistent activation of protein kinases, including Ca2+/calmodulin-dependent protein kinase (CAMKII), protein kinase C (PKC), and casein kinase II (CK2). GLYX-13 (3 mg/kg, IV), or vehicle, were administered to adult (2-3 months old) male Sprague-Dawley rats, and medial prefrontal cortex samples were collected at 15, 30, 60, and 120 min post-dosing (n=7-9 per group). Total cellular proteins were subjected to 7.5% SDS-PAGE and probed with antibodies directed against GluN2B (4207S, Cell Signaling, MA), pS-1303 GluN2B (Millipore, Mass.), or pS-1480 GluN2B (ab73014, Abcam, Mass.). Enhanced chemiluminescence was used to quantitate individual bands. CK2 activity was measured by phosphorylation of a CK2 substrate peptide using the transfer of the gamma-phosphate of [gamma-32P]-ATP (Millipore, Mass.). Total protein (7.5 micrograms) was incubated with CK2 substrate peptide for 10 min in the presence of 0.1 microliters of stock [gamma-32P]-ATP (100 nCi/reaction).


GLYX-13 led to a significant increase in total GluN2B protein within 15 min (1.53 fold vs. vehicle, P<0.05) of administration that peaked at 30 min (1.71 fold, P<0.05) and returned to control levels by 60 min (60 min, 1.13 fold, P >0.05; 120 min, 1.16 fold, P >0.05) (FIGS. 10 and 11). CAMKII/PKC-mediated serine-1303 phosphorylation of GluN2B levels were increased at the 30 min (1.93 fold, P<0.05), 60 min (2.23 fold, P<0.05), and 120 min (2.67 fold, P<0.05) timepoints but did not change at the 15 min timepoint (1.02 fold, P >0.05) (FIG. 10). CK2-mediated serine-1480 phosphorylation of GluN2B levels peaked within 15 min (2.01 fold, P<0.05) and remained significantly elevated up to 120 min (30 min, 1.80 fold, P<0.05; 60 min, 1.48 fold, P<0.05; 120 min 1.50 fold, P<. 05) after administration of GLYX-13 (FIG. 11). A significant increase in CK-2 specific activity was also observed at 15 min (3.08 fold, P<0.05) (FIG. 12). CK2 and CAMKII activity has been shown to be rapidly increased at the onset of LTP (Charriaut-Marlangue et al., 1991, PNAS, 88, 10232; Fukunaga et al., 1993, JBC, 268, 7863). This observation, along with the results reported here, suggest that the rapid onset of the antidepressant effect of GLYX-13 is mediated, at least in part, by the same mechanisms that regulate E-LTP.


EQUIVALENTS

While specific embodiments of the subject disclosure have been discussed, the above specification is illustrative and not restrictive. Many variations of the disclosure will become apparent to those skilled in the art upon review of this specification. The full scope of the disclosure should be determined by reference to the claims, along with their full scope of equivalents, and the specification, along with such variations.


Unless otherwise indicated, all numbers expressing quantities of ingredients, reaction conditions, parameters, descriptive features and so forth used in the specification and claims are to be understood as being modified in all instances by the term “about.” Accordingly, unless indicated to the contrary, the numerical parameters set forth in this specification and attached claims are approximations that may vary depending upon the desired properties sought to be obtained by the present invention.


INCORPORATION BY REFERENCE

All publications and patents mentioned herein, including those items listed below, are hereby incorporated by reference in their entirety as if each individual publication or patent was specifically and individually indicated to be incorporated by reference. In case of conflict, the present application, including any definitions herein, will control.

Claims
  • 1. A method for identifying a candidate compound suitable for treatment of depression, comprising: exposing a cell to a potential compound in a culture medium, or administering a potential compound to an animal;retrieving a sample from the cell and/or culture medium, or from brain or neural tissue of the animal, at one or more predetermined time points;analyzing the sample for increased expression levels of Wnt1, andidentifying the candidate compound as suitable for treatment of depression based on the increased expression level of Wnt1.
  • 2. A method for identifying a candidate compound suitable for treatment of depression, comprising: exposing a cell to a potential compound in a culture medium, or administering a potential compound to an animal;retrieving a sample from the cell and/or culture medium, or from brain or neural tissue of the animal, at one or more predetermined time points;analyzing the sample for increased expression levels of at least one of the genes listed in Table 1 or 2 indicated with a G, or decreased expression levels or at least one of the genes listed in Table 1 or 2 indicated with a K, andidentifying the compound as suitable for treatment of depression based on the increased expression level or decreased expression level.
  • 3. The method of claim 2, wherein the sample has a gene expression pattern as provided in Table 1 or 2 with the indication “G” and the identifying is based on increased expression of those genes.
  • 4. The method of any one of claims 1-3, further comprising analyzing the candidate compound for NDMA subunit NR2B synaptic plasticity.
  • 5. A method for identifying a compound suitable for treatment of depression, comprising: exposing a cell to a potential compound in a culture medium, or administering a potential compound to an animal;retrieving a sample from the cell and/or culture medium, or from brain or neural tissue of the animal, at one or more predetermined time points;analyzing the sample for NMDA receptor NR2B subunit plasticity, andidentifying the compound as suitable for treatment of depression based on inducing the NR2B plasticity.
  • 6. The method of claim 5, wherein a candidate compound suitable for treating depression significantly induces NR2B dependent synaptic plasticity as compared to ketamine.
  • 7. The method of any one of claims 1-6, wherein the tissue is medial prefrontal cortex.
  • 8. The method of any one of claims 1-7, wherein the animal is a rodent or human, and the cell is a human or rodent cell.
  • 9. The method of any one of claims 1-7, wherein the compound modulates the NMDA receptor.
  • 10. The method of any one of claims 1-8, wherein the compound suitable for treating depression has fewer side effects as compared to ketamine.
  • 11. The method of claim 10, wherein the compound does not have substantial addictive sensory motor grating and/or sedative effect.
  • 12. The method of any one of claims 1-11, wherein the cell is a eukaryotic cell.
  • 13. The method of any one of claims 1-12, further comprising selecting the candidate compound from a library of compounds.
  • 14. A method of identifying a therapeutic compound capable of treating depression in a patient, comprising selecting a compound that significantly induces NR2B dependent synaptic plasticity.
CROSS REFERENCE TO RELATED APPLICATIONS

This application claims the benefit of and priority to U.S. Provisional Patent Application Ser. No. 61/824,667, filed May 17, 2013, and U.S. Provisional Patent Application Ser. No. 61/713,085, filed Oct. 12, 2012, each of which is hereby incorporated by reference in its entirety.

PCT Information
Filing Document Filing Date Country Kind
PCT/US13/64625 10/11/2013 WO 00
Provisional Applications (2)
Number Date Country
61713085 Oct 2012 US
61824667 May 2013 US