Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Aug 1.
Published in final edited form as: Pharmacol Biochem Behav. 2011 Feb 24;99(2):190–210. doi: 10.1016/j.pbb.2011.02.002

Alzheimer’s Disease and Age-Related Memory Decline (Preclinical)

Alvin V Terry Jr 1, Patrick M Callahan 1, Brandon Hall 1, Scott J Webster 1
PMCID: PMC3113643  NIHMSID: NIHMS272769  PMID: 21315756

Abstract

An unfortunate result of the rapid rise in geriatric populations worldwide is the increasing prevalence of age-related cognitive disorders such as Alzheimer’s disease (AD). AD is a devastating neurodegenerative illness that is characterized by a profound impairment of cognitive function, marked physical disability, and an enormous economic burden on the afflicted individual, caregivers, and society in general. The rise in elderly populations is also resulting in an increase in individuals with related (potentially treatable) conditions such as “Mild Cognitive Impairment” (MCI) which is characterized by a less severe (but abnormal) level of cognitive impairment and a high-risk for developing dementia. Even in the absence of a diagnosable disorder of cognition (e.g., AD, MCI), the perception of increased forgetfulness and declining mental function is a clear source of apprehension in the elderly. This is a valid concern given that even a modest impairment of cognitive function is likely to be associated with significant disability in a rapidly evolving, technology-based society. Unfortunately, the currently available therapies designed to improve cognition (i.e., for AD and other forms of dementia) are limited by modest efficacy, adverse side effects, and their effects on cognitive function are not sustained over time. Accordingly, it is incumbent on the scientific community to develop safer and more effective therapies that improve and/or sustain cognitive function in the elderly allowing them to remain mentally active and productive for as long as possible. As diagnostic criteria for memory disorders evolve, the demand for pro-cognitive therapeutic agents is likely to surpass AD and dementia to include MCI and potentially even less severe forms of memory decline. The purpose of this review is to provide an overview of the contemporary therapeutic targets and preclinical pharmacologic approaches (with representative drug examples) designed to enhance memory function.

Keywords: cognition, drug development, pro-cognitive, neurotransmitter receptors, Mild Cognitive Impairment, dementia

1. Introduction

According to the United Nations (UN, 2007) and the United States Centers for Disease Control and Prevention (CDC, 2003) a combination of declining fertility and increasing life expectancies is a phenomenon that is resulting in an unprecedented growth of elderly populations worldwide. An unfortunate result of this trend is the concomitant rise in the number of people suffering from age-related forms of dementia such as Alzheimer’s disease (AD). In the United States alone the number of individuals with AD could be considered epidemic at approximately 5.3 million (Herbert et al., 2003), and unfortunately, this number is expected to increase dramatically in the near future due to the number of aging baby boomers (CDC, 2007; Kinsella et al., 2009). This issue has many dimensions at both the individual level and on societies as a whole. On an individual level, AD significantly shortens life expectancy and it is one of the major causes of physical disability, institutionalization, and decreased quality of life among the elderly (reviewed Qiu et al., 2009). On a societal level, the marked rise in the number of dementia patients is likely to create an extraordinary (potentially unsustainable) economic burden and demand on social systems and health care-related institutions. It is also important to note that the population estimates (for AD) highlighted above do not include individuals with related (potentially treatable) conditions such as “Mild Cognitive Impairment” (MCI) which is characterized by a less severe (but abnormal) level of cognitive impairment (reviewed Petersen et al., 2004) and a high-risk for developing dementia (reviewed, Luck et al., 2010). Even in the absence of a diagnosable disorder of cognition (e.g., AD, MCI), the perception of increased forgetfulness and declining memory function is a clear source of distress, embarrassment, and low self esteem in the elderly (Imhof et al., 2006; Ohman et al., 2008, see also review, Ballard, 2010). Undoubtedly these factors have (at least in part) driven sales of over the counter nutritional supplements (e.g., ginko biloba, phosphatidyl serine) that promise improved memory function (now a billion dollar industry in the United States alone), despite the lack of any clear evidence of their effectiveness (Kennedy, 2004, see also review, Lanni et al., 2008).

Collectively, the information discussed above indicates that it is incumbent on the scientific community to develop safer and more effective pro-cognitive agents that could allow even the oldest in our societies to remain socially active and productive. The purpose of this review is to provide a brief overview of the contemporary therapeutic targets and preclinical pharmacologic approaches (with representative drug examples) designed to enhance memory function. The pharmacologic agents reviewed are not all-inclusive but represent examples from the more actively pursued (i.e., current) areas of investigation. The review does not address the wide variety of potentially exciting disease modifying (mechanism-based) treatments that are in development for AD based on the amyloid cascade hypothesis (vaccines, β and γ secretase inhibitiors), or therapies targeting tau and neurofibrillary tangle formation, neuroinflammation, etc.

2. Current Treatments

Since the FDA approval of the acetylcholinesterase inhibitor (AChEI) tacrine in 1993, the primary therapeutic approach to the cognitive loss associated with AD (and other forms of dementia) has been that of a cholinergic replacement strategy. This approach is based on studies spanning more than 35 years in both humans and lower animals indicating that acetylcholine (cholinergic) pathways in the brain modulate a number of important processes including attention, working memory, and other aspects of cognition (reviewed, Terry and Buccafusco, 2003). Furthermore, the brains of cognitively impaired elderly patients and those who suffered from AD commonly exhibit damage or abnormalities in cholinergic pathways. Both postmortem and antemortem studies in aged humans and AD patients, as well as animal experiments (i.e., in aged animals and AD-related animal models) show decreased high affinity choline uptake; decreased acetylcholine synthesis and release, and decreased numbers of nicotinic and muscarinic acetylcholine receptors (Auld, et al., 2002). Collectively such findings have lead to the so called “Cholinergic Hypothesis of AD” (reviewed, Bartus, 2000). With the exception of the relatively recent FDA approval of the NMDA antagonist, memantine (see below) in the United States, only the clinical data derived from studies based on the cholinergic hypothesis (more specifically, with AChEIs) have provided convincing evidence of an adequate level of efficacy and reliability in AD balanced with an acceptable burden of side effects.

2.1 AChEIs

FDA-approved therapy with donepezil, rivastigmine, and galantamine is based on several large clinical studies in patients diagnosed with mild to moderate AD (Boada-Rovira et al., 2004; Mintzer and Kershaw, 2003; Raskind et al., 2000; Winblad et al., 2001). The compounds produced subtle but statistically significant cognitive improvements in the AD study patients. Further, there are some (albeit controversial) retrospective data to suggest that these agents may enhance activities of daily living and improve behavioral disturbances in AD (reviewed, Standridge, 2004). However, there are a number of limitations associated with AChEI’s. This drug class can be associated with a number of peripheral side effects (e.g., nausea, vomiting, diarrhea, tremors) and it is only effective in a limited number of patients. In addition, few would argue that the compounds in this class have potent neuroprotective activity, the ability to preserve cognition over significant pwerioids of time, or to prevent the inevitable out come of the illness.

2.2 Memantine

Memantine, the other FDA-approved therapeutic agent for the cognitive symptoms of AD is a low to moderate affinity uncompetitive (open-channel) NMDA receptor antagonist which binds preferentially to the NMDA receptor-operated cation channels. These actions are believed support the therapeutic effects of memantine by combating the persistent activation of N-methyl-D-aspartate (NMDA) receptors by the excitatory amino acid glutamate in the brain, a process thought to contribute to the symptomatology of AD. Approval of memantine was based on three randomized, double-blind, placebo-controlled clinical trials in moderate to severe AD (reviewed, Thomas and Grossberg, 2009). The clinical data indicate that memantine is a relatively safe drug with few major side effects and a negligible risk of drug-drug interactions, but only small clinically-relevant effects on cognition, global functioning and activities of daily living have been observed, mainly in patients with moderate AD (reviewed, van Marum, 2009). Memantine is recommended both as monotherapy and in combination with AChEIs, and the later approach is very commonly used, despite a lack of clear evidence that one approach is superior to the other (reviewed, Thomas and Grossberg, 2009).

3. Novel Cholinergic-Based Strategies

Please see Table 1 for a summary of the variety of cholinergic-based strategies for cognitive enhancement.

Table 1.

Cholinergic-Based Compounds

Compound Name Receptor Mechanism References
AChE Inhibitors
TAK-147 AChE Inhibitor Xu, 2002
BZYX AChE Inhibitor Shen, 2009
Phenserine AChE Inhibitor Janas, 2005
CHF2819 AChE Inhibitor Trabace, 2007
Eptastigmine AChE Inhibitor Imbimbo, 2001
Huperzine A AChE Inhibitor Wang, 2010
Loganin AChE Inhibitor Lee, 2009
NP-0361 AChE Inhibitor Buccafusco, 2009
Pseudoberberine AChE Inhibitor Hung, 2008
Memoquin AChE Inhibitor Cavalli, 2007
PMS777 AChE Inhibitor Bate, 2004
Nicotinic
SIB-1508Y α4β2 agonist Schneider et al., 1999
SIB-1553A α2β4 agonist Schneider et al., 2003
ABT-418 α4β2 agonist Prendergast et al., 1998
ABT-594 α4β2 agonist Buccafusco et al., 2007
S-38232 α4β2 agonist Howe et al., 2010
ABT-089 α4β2 partial agonist Decker et al., 1997
Ispronicline/AZD-3480 α4β2 partial agonist Gatto et al., 2004
Varenicline/Chantix® α4β2/α7 partial agonist/full agonist Rollema et al., 2009
GTS-21 α7 partial agonist Buccafusco and Terry, 2009b
MEM-3454/RG-3487 α7 partial agonist Callahan et al., 2006; Rezvani et al., 2009
MEM-63908/RG4996 α7 partial agonist Taly et al., 2009
AZD-0328 α7 partial agonist Sydserff et al., 2009
AR-R17779 α7 agonist Van Kampen et al., 2004
S-24795 α7 partial agonist Lopez-Hernandez et al., 2007
SSR-180711 α7 partial agonist Taly et al., 2009
SEN12333 (WAY-317538) α7 agonist Roncarati et al., 2009
NS-1738 α7 positive allosteric modulator Timmermann et al., 2007
Compound 6 α7 positive allosteric modulator Ng et al., 2007
PNU-120596 α7 positive allosteric modulator Hurst et al., 2005
2087101 α4β2/α7 positive allosteric modulator de Filippi et al., 2010
Cotinine α4β2/α7 nAChR desensitization Buccafusco et al., 2009; Terry et al., 2005a
JWB1-84-1 α4β2/α7 nAChR desensitization Buccafusco et al., 2009; Sood et al., 2007
JAY2-22-33 α4β2/α7 nAChR desensitization Buccafusco et al., 2009; Sood et al., 2007
Muscarinic
Talsaclidine M1 agonist Clader and Wang, 2005
Xanomeline M1 agonist Clader and Wang, 2005
Cevimeline/AF102B M1 agonist Fisher et al, 2002
AF150(S) M1 agonist Fisher et al, 1998;
AF267B M1 agonist Caccomo et al, 2006
CI-1017 M1 agonist Tecle et al, 2000
WAY-132983 M1 agonist Bartolomeo et al, 2000
EUK-1001 M1 agonist Cui et al, 2008a
MCD-386 M1 agonist Buccafusco, 2009a
AC-42 M1 positive allosteric modulator Spalding et al, 2006
77-LH-28-1 M1 positive allosteric modulator Fisher, 2008
TBPB M1 positive allosteric modulator Jones et al, 2008
SCH 57790 M2 antagonist Carey et al., 2001
SCH 72788 M2 antagonist Lachowicz et al., 2001
BIBN-99 M2 antagonist Rowe et al., 2003

3.1 New AChEIs

There is a variety of compounds that have been synthesized as analogs of the currently marketed AChEIs, or as analogs of older AChEIs with well-characterized pharmacological profiles such as physostigmine (originally derived as an alkaloid of the Calabar bean). One such compound, TAK-147, is a derivative of donepezil which has been shown to produce pro-cognitive effects in rats performing a scopolamine reversal task in the Morris water maze (Xu et al., 2002; Chen et al., 2002). The compound has also shown cognitive enhancing effects in a rat based chronic cerebral ischemia cognitive deficit model similar to donepezil (Xu et al., 2002). TAK-147 has also been shown to ameliorate deficits induced by diazepam on passive avoidance and restore performance in a scopolamine deficit model in delayed-matchingto-position task in rats (Miyamoto, et al., 1996). BZYX is another donepezil derivative with a dialkylbenzyl amine moiety from rivastigmine that was designed to function as a dual binding site AChE inhibitor (Sheng et al., 2009). This compound is capable of reversing scopolamine, NaNO2, and ethanol induced deficits in rats in a radial arm maze (Zhang et al., 2009). Further, BZYX seems to have neuroprotective properties by scavenging reactive oxygen species produced by H2O2 and preventing the loss of mitochondria membrane potential (Zhang et al., 2009).

Phenserine, a derivative of physostigmine is a long-acting AChEI and has shown considerable cognitive enhancing abilities. In the Morris water maze, phenserine has displayed pro-cognitive properties in rats by significantly decreasing travel distance and lowering latencies to reach the platform (Janas et al., 2005). In a dog delayed-non-matching-to-sample task, phenserine has demonstrated the ability to reverse scopolamine induced deficits on task accuracies (Araujo et al., 2005; Studzinski et al., 2005). The compound has also been shown to enhance cognition in rats using a 14 unit T-maze by reducing the number of errors it took to complete the maze (Ikari et al., 1995). Interestingly, phenserine is also able to overcome deficits induced by the NMDA receptor antagonist 3-(±) 2-carboxypiperzin-4-yl) propyl phosphonic acid (CPP) in the same 14-unit T-maze (Patel et al., 1998). Similarly, eptastigmine, a derivative of physostigmine, and ganstigmine (CHF2819), a derivative of geneserine (a closely related compound to physostigmine), have both shown cognitive enhancing properties by reversing scopolamine induced deficits (Dawson et al., 1991; Rupniak et al., 1992; Trabace et al., 2007).

Huperzine A, another alkaloid, isolated from the Chinese herb Huperzia serrata, is a potent inhibitor of AChE and has been shown to have a pro-cognitive effect in a wide number of preclinical animal models including the Morris water maze task (Wang et al., 2006; Wang et al., 2010) and the radial arm maze task (Wang and Tang, 1998). The compound has also shown cognitive enhancing properties in a delayed response task in monkeys (Ye et al., 1999; Ou et al., 2001). Further, Huperzine A has been used to reverse scopolamine induced deficits (Wang and Tang, 1998; Gao et al., 2000) and produce pro-cognitive effects in aged rodents and monkeys (Ye et al., 1999; Ye et al., 2000). Two other compounds of interest are loganin, an iridoid glycoside isolated from the Cornus officinalis fruit and pseudoberberine, isolated from Corydalis turtschaninovii. Both are AChE inhibitors and have displayed cognitive enhancing properties (Hung et al., 2008; Lee et al., 2009). Loganin displays cognitive enhancing properties when tested in a scopolamine induced deficit reversal task using the Y-maze and passive avoidance task (Kwon et al., 2009). Loganin has also been shown to improve learning and spatial memory in rats tested in the Morris water maze task (Lee et al., 2009). Pseudoberberine has also been shown to reverse scopolamine induced cognitive impairments in mice in both a passive avoidance task and in a water maze task (Hung et al., 2008).

Recently there has been renewed interest in AChE inhibitors as they have been shown to modulate the processing of APP away from production of the toxic amyloid beta peptide (De Ferrari et al., 2001; Racchi et al., 2005; Peng et al., 2007; Rizzo et al., 2010). This has lead to the synthesis of novel AChE inhibitors many of which hit multiple targets relevant to AD (Fernández-Bachiller et al., 2010; Mohamed and Rao, 2010; Huang et al., 2010; Kikuchi et al., 2010; de Los Ríos et al., 2010; Samadi et al., 2010; Pisani et al., 2010). Many of these compounds are still in their infancies, requiring much more thorough examination before they prove their worth in the treatment of AD. Still, expect to see more interest in this area of research as these AChE inhibitors, as well as their derivatives, continue to be tested in the months and years ahead.

3.2 Muscarinic Acetylcholine Receptor (mAChRs) Ligands

mAChRs are metabotropic receptors that are coupled to G-proteins. Agonist binding to mAChRs results in decoupling of the G-protein prompting a signal transduction cascade that leads to either excitatory or inhibitory actions on the cell. There have been five types of mAChRs identified, labeled M1–M5 (Levey et al., 1991). The M1, M3, and M5 receptor subtypes are coupled to Gq proteins that activate the phospholipase C (PLC) pathway, which results in protein kinase C (PKC) activation leading to intracellular calcium release and excitatory actions on the cell. The M2 and M4 subtypes are coupled to Gi proteins that when activated inhibit adenylyl cyclase activity, resulting in a reduction of cAMP and inhibitory actions on the cell. The M1 receptor subtype has been the dominant target for cognition enhancement therapy, due to its wide expression in the cortex and hippocampus (Levey et al., 1991), and because the receptor subtype seems to be retained longer than other mAChRs in AD (Flynn et al., 1995).

3.2.1 Muscarinic receptor agonists

There have been numerous M1 receptor agonists evaluated in preclinical studies as possible treatments for cognitive impairment associated with AD and age-related memory decline (Clader and Wang, 2005). The first generation of M1 agonists were analogs of the mAChR agonist arecoline, a class of compounds that includes talsaclidine, sabomeline, xanomeline, and cevimeline, among others. Many of these compounds showed positive cognitive effects in preclinical tests. However, a lack of sufficient selectivity for the M1 receptor over the M3 and M5 receptors hindered further development efforts for many of these compounds for AD therapy, due to potential cholinergic side effects. Indeed, this problem has hindered much development of subtype selective mAChR agonists/antagonists, owing to the high amount of sequence homology shared by the orthosteric binding sites of mAChRs subtypes (Hulme et al., 1990). However, the M1 receptor continues to be an attractive target for cognition therapy.

Significant preclinical data supporting M1 receptors as valid targets for cognition enhancement were obtained through studies utilizing the “AF” compounds, a series of M1 partial agonists composed of derivatives of cevimeline. Early studies revealed that treatment with cevimeline, also known as AF102B, produced positive outcomes in a broad range of animal models of AD and cognitive impairment (Clader and Wang, 2005). The analogs developed from AF102B are more efficacious at M1 receptors with greater selectivity. Treatment with AF150(S) abolished cognitive impairments associated with ApoE knockout mice, a strain of mice that shows cognitive deficits resembling AD pathology (Fisher et al., 1998). Other studies with AF150(S) demonstrated memory and cognition improvements in young and old rodents in the delayed matching-to-position task, Morris water maze task, as well as the radial arm maze (Brandeis et al., 1995; Ruske and White, 1999). When administered to 3xTG-AD mice, a mouse model of AD characterized by Aβ plaque buildup, neurofibrillary tangles, and deficits in cognition, the compound AF267B recovered deficits in spatial working memory seen in the Morris water maze test after 2 months of treatment (Caccamo et al., 2006). Treatment with AF267B was also effective in reducing cognitive deficits in animals with lesions in the nucleus basalis (Beach et al., 2003). Other analogs of AF102B include AF125 and AF151 (Fisher et al., 1993). Adding to the appeal of these more selective M1 compounds was an apparent lack of peripheral side effects in preclinical models as well as a longer half life and high bioavailability (Fisher et al., 2002).

Efforts continue to develop newer, more selective M1 agonists for cognition therapy in AD that also have a low potential for undesirable peripheral cholinergic effects. A newly synthesized class of arecoline analogs, N-alkyl/aryl substituted thiazolidinone arecoline derivatives, showed significant affinity for M1 receptors, and improved scopolamine-induced impairments in the passive avoidance step down task and elevated plus maze transfer latency (Sadashiva et al., 2009). Other arecoline derivatives with M1 receptor selectivity are also currently being synthesized and tested in preclinical models of AD (Kumar et al., 2008; Malviya et al., 2008; Malviya et al., 2009). The novel derivative of xanomeline, EUK1001, improved novel object recognition and fear cognition in a passive avoidance task in aged mice (Cui et al., 2008a; Si et al., 2010). EUK1001 was also shown to have higher affinity for M1 receptors than xanomeline coupled with a significantly lower side effect profile (Cui et al., 2008b). It will remain to be seen whether these newer compounds succeed in any clinical setting. However, a great promise still holds for compounds in this pharmacological class, assuming a given new molecular entity carries sufficient selectivity for M1 receptors over other mAChR subtypes.

3.2.2 Muscarinic receptor antagonists

Both in vitro and in vivo (e.g., M2R−/− knockout mice) studies suggests that M2 muscarinic receptors in the mammalian hippocampus and cortex serve as presynaptic autoreceptors mediating the inhibition ACh release. This evidence supports the hypothesis that antagonists at central M2 autoreceptors might enhance cognition by increasing synaptic acetylcholine levels (see Wess et al., 2007 for review). Consistent with this notion are studies indicating that specific M2 receptor-preferring antagonists such as SCH 57790, SCH 72788, and BIBN-99 can improve learning and memory-related tasks in animal models (Carey et al., 2001; Lachowicz et al., 2001; Rowe et al., 2003. However, M2R−/− mice were found to exhibit deficits in some cognitive tasks suggesting that complete pharmacological blockade of central M2 receptors might (in fact) have negative effects on cognition (Tzavara et al., 2003). Several hypotheses have been proposed to explain these discrepant findings. One possibility is that the M2 receptor-preferring antagonists evaluated to date may only serve as partial antagonists of central M2 receptors. Another is that there are both presynaptic and postsynaptic M2 receptor-signalling pathways in the CNS that may have different sensitivities to the antagonists used (see Wess et al., 2007 for review). Thus, additional studies are needed to determine which strategy (M2 antagonism versus enhancement) will be likely to provide a therapeutic benefit in disorders of cognition.

3.2.3 Muscarinic positive allosteric modulators

The lack of sufficient selectivity of orthosteric ligands for targeted muscarinic receptor subtypes has driven research for compounds that act as positive allosteric modulators (PAMs) on the receptor, targeting binding sites away from the orthosteric site (see nicotinic positive allosteric modulator section below for a complete discussion). There have been several mAChR PAMs that have been identified in in vitro studies (Jones et al., 2008; Conn et al., 2009). Many of these compounds have been reported as possessing very high selectivity for the targeted receptor. However, to date only a small number of these compounds have been studied in animal models for cognition enhancement, though promising results have recently been published (Bradley et al., 2010). With the continuing need for new therapies addressing the cognitive decline associated with AD and advanced age, these compounds in this class will surely garner much attention for future preclinical cognition studies.

3.3 Nicotinic Acetylcholine Receptor (nAChR) Ligands

nAChRs are ionotropic receptors that open in response to agonist binding. The result of agonist binding to the receptor is the transfer of cations into, and out of, the cell, notably the influx of sodium and to a lesser degree calcium, and the efflux of potassium. The end result of nAChR activation is membrane depolarization. Nicotinic receptors are composed of five membrane-spanning subunits that form a pentameric arrangement around a water-filled pore (Jones et al., 1999; Karlin 2002; McGehee and Role, 1995). Neuronal nicotinic receptors are formed through a combination of α and β subunits composing heteromeric receptors, or through homomeric α configurations. Although there have been nine α (α2–α10) and three β (β2–β4) subunits currently identified that can form many different combinations, for the purposes of this review, the heteromeric α4β2 and homomeric α7 nAChR subtypes will be primarily discussed. These two subtypes have been the targets for the majority of cognition enhancing drugs for AD and age-related memory decline, as both subtypes are highly expressed in the cerebral cortex and hippocampus. Targeting other nAChR subtypes has been associated with efficacy and side effect issues, owing to their distribution in other brain areas such as the nucleus accumbens (Buccafusco, 2004).

3.3.1 Nicotinic receptor agonists

The ability of nicotine to improve cognition in animal models as well as in humans has been known for over two decades, and has been extensively studied and reviewed (Elrod et al., 1988; Levin, 2002; Murray and Abeles, 2002). However, the use of nicotine clinically for any purpose other than smoking cessation was severely limited by concerns over serious side effects (Benowitz, 1986). The goal for cognition enhancement through nAChRs is to develop receptor subtype selective agonists that do not possess the undesirable side effects accompanied by the use of nicotine, particularly the abuse and addiction potential. Compounds selective for the α4β2 subtype were the first to be developed for cognition enhancement and studied in preclinical studies.

Two of the first α4β2 agonists tested for cognition enhancement were SIBIA Neuroscience’s SIB-1508Y and Abbott’s ABT-418. These compounds showed great promise in pre-clinical animal models of cognitive impairment (Schneider et al., 1999; Prendergast et al., 1998). However, a lack of selectivity for the α4β2 receptor hindered further development for these compounds. ABT-089, a follow-up compound to ABT-418, was shown to improve Morris water maze performance in aged rats, and when tested in young rats with septal lesions improved acquisition in a spatial discrimination water maze task (Decker et al., 1997). In the same study, ABT-089 improved cognitive performance in young and aged monkeys in the delayed matchingto-sample task. Although ABT-089 has less affinity for α4β2 nAChRs than either nicotine or ABT-418, the compound was shown to be as potent as nicotine in evoking hippocampal ACh release from rat synaptosomes (Sullivan et al., 1997). However, despite ABT-089’s comparable effects to nicotine for ACh release, the compound does not share nicotine’s potency at inducing dopamine release in the ventral tegmental area (VTA), perhaps owing to the compound’s pharmacological profile as a partial agonist. ABT-594, an α4β2 agonist currently being studied for analgesic potential, was shown to improve delayed matching-to-sample task performance when studied in non-human primates (Buccafusco et al., 2007). The α4β2 partial agonist varenicline (Chantix®), already FDA approved for smoking cessation has also been considered a candidate for potential cognition enhancement. In pre-clinical tests, varenicline improved sustained attention task performance in rats, a model which tests top-down control of attention (Rollema et al., 2009). Deficits in top-down attention control are a prominent feature of age-related memory decline. Rats administered varenicline also demonstrated improvements in the novel object recognition task compared to vehicle treated animals in the same study. Ispronicline (AZD-3480), another α4β2 partial agonist, improved working memory in rodents, even after a single dose (Gatto et al., 2004). This improvement was observed for up to 18h despite a half life of only 2h, highlighting prolonged effects present even after elimination of the compound. Finally, a recently published study highlights the positive effects of the agonist S-38232 in a distractor version of the sustained attention task in rodents, though no effect was observed in a standard sustained attention task paradigm (Howe et al., 2010). These recent studies demonstrate the continuing appeal of targeting α4β2 nAChRs for cognition enhancement in preclinical research.

Compounds targeting the homomeric α7 nicotinic receptor subtype have yielded some promising results for cognition enhancement and are a major focus of current preclinical research (Leiser et al., 2009). The distribution of α7 nAChRs is limited primarily to the hippocampus, certain cortical layers, and the hypothalamus, making these receptors intriguing targets for cognition enhancement (Freedman et al., 1995). Activation of α7 nAChRs does not appear to be involved in the rewarding effects of nicotine, suggesting that targeting these receptors has limited abuse potential (Brioni et al., 1996). Also, while not the focus of this review, the extrasynaptic localization of α7 nAChRs suggests a role for the receptor subtype in cell viability. The α7 partial agonist and anabaseine synthetic derivative GTS-21 (3-[(2, 4 dimethoxy) benzylidene]-anabaseine, or DMXBA) is a well studied compound and was poised to perhaps become the first synthetic nicotinic compound to market for cognition enhancement. The compound has been shown in preclinical models to enhance cognitive processes, including studies in rodents and non-human primates (Buccafusco and Terry, 2009; Chen et al., 2010; Rodefer et al., 2007). This cognition enhancement by GTS-21 is accompanied by a low potential for side effects (Kem, 2000). What has made GTS-21 particularly interesting is the compound’s relative specificity for cognition enhancement versus its inactivity in behavioral tasks in which nicotine is effective (Briggs et al, 1997). The most significant effects of GTS-21 reported to date have been observed in tasks of attention, working memory, and episodic secondary memory. The results of preclinical tests using compounds like GTS-21 have driven the development of newer, more selective α7 receptor agonists for use in cognition enhancement. The Memory Pharmaceuticals (acquired by Roche) compound MEM 3454 (known as RG-3487) is an α7 nAChR partial agonist with 5-HT3 receptor antagonist properties that has been shown to enhance task performance in young and aged-cognitively impaired rodents as well as young and aged non-human primates (Callahan et al., 2006; Wallace-Boone et al., 2009; Wallace et al., 2011) across multiple cognition domains (episodic, attention, executive function, spatial reference and working memory). RG-3487 was also shown to improve sustained visual attention in rodents, a property shared by nicotine but not galanthamine (Rezvani et al., 2009). Moreover, the improvements in cognitive performance were shown to be reflective of the observed increases in acetylcholine and dopamine efflux in cortical and hippocampal brain regions (Huang et al., 2006). These positive preclinical observations translated into significant improvements in cognition, as measured by the cognitive drug research (CDR) test battery in healthy human volunteers and mild to moderate AD subjects during phase I and phase IIA clinical testing (Callahan et al., 2006). Another α7 nAChR partial agonist from Memory Pharmaceuticals, MEM 64368, demonstrated robust cognitive enhancing properties in rodents and non-human primates (Rodefer et al., 2007; Wallace-Boone et al., 2007). This α7 nAChR partial agonist was characterized as a low intrinisic activity agonist (~30%) with moderate affinity (Ki ~300 nM) at α7 nAChR receprtors compared to that of RG-3487 (Ki= 6nM; intrinsic activity ~68%) and displayed a 10-fold selectivity for α7 nAChR over 5-HT3 receptors (Wallace-Boone et al., 2007; Wallace et al., 2011). In addition, AstraZeneca’s α7 nAChR compound AZD-0328 improved spatial working memory in mice in a novel object recognition task, an improvement that was partly attributed to enhanced midbrain and coritcal dopamine release (Sydserff et al., 2009). The novel α7 agonist SEN12333 (WAY-317538) has also been shown to improve novel object recognition performance in rats (Roncarati et al., 2009). This improvement was observed even under scopolamine or MK-801-induced amnesia, representing relevant cholinergic and glutamatergic deficits commonly seen in AD. In the same study, SEN12333 recovered deficits in a passive avoidance task induced by scopolamine. Results similar to these have been observed in preclinical cognitive tests using a host of α7 nAChR agonists (Table 1).

3.3.2 Nicotinic Positive Allosteric Modulators

Due to the historical difficulty in developing compounds with sufficient selectivity for the orthosteric ligand binding site of nAChR subtypes, there has been considerable interest directed toward allosteric modulators for targeting nAChRs. Positive allosteric modulators avoid direct receptor activation by utilizing binding sites away from the orthosteric site targeted by traditional agonists. Though allosterically-acting compounds have been more difficult to discover than compounds acting at orthosteric sites, the existence of these compounds has been known for several years and their use has recently been suggested as a favorable strategy for targeting nicotinic receptors (Albuquerque et al., 2009). The emergence of more advanced techniques for investigating molecular site interactions, as well as in silico screening of large compound libraries for receptor targets, has generated interest in this approach in certain drug discovery arenas. However, only a few compounds in this class have been studied in vivo. The drug galantamine, an acetylcholinesterase inhibitor currently in use for AD treatment, has also been demonstrated to act as a positive allosteric modulator of nAChRs (Coyle et al., 2007).

The allosteric potentiating ligands (APLs) of nAChRs that have been tested in preclinical memory and cognition models have demonstrated great potential for this class of compounds. The compound NS-1738, classified as an α7 selective positive allosteric modulator (PAM), recovered scopolamine-induced deficits in the Morris water maze task in rats as well as improving the animals performance in a social recognition model (Timmermann et al., 2007). The PAM known as compound 6 also targets α7 nAChRs, and when administered to rats significantly improved their performance in the radial arm maze task compared to vehicle treated control animals (Ng et al., 2007). Compound 6 also showed positive results in sensory gating deficits, a property shared by another nAChR PAM, PNU-120596 (Hurst et al., 2005).

3.3.3 Desensitization of nicotinic-acetylcholine Receptors (nAChRs)

As is apparent in the preceding paragraphs, drug discovery efforts for pro-cognitive agents in the nicotinic field to date have focused on agonists, partial agonists and allosteric modulators (i.e., all efforts designed to increase activity of nAChRs). These approaches have (for the most part) been based on the idea that enhanced activity at nAChRs promotes the release of neurotransmitters in relevant brain regions which results in the desired behavioral effect (improved cognition). However, in a number of settings, nAChR agonists and antagonists can have very similar physiologic effects (see review, Buccafusco et al., 2009), and further, a large discrepancy often exists between the high concentrations of nicotinic drugs required to activate nicotinic responses in vitro and the lower doses used to induce various behavioral responses in vivo. Nicotine can both activate and desensitize its receptors over a relatively short time course leading to the question of whether (in fact) nAChR desensitization when compared to receptor activation, plays an equal if not more important role in the behavioral effects. Recent studies in our laboratories (Sood et al., 2007; Buccafusco et al., 2009) indicated that nicotine, the major nicotine metabolite, cotinine, and two novel analogs of choline, JWB1-84-1 [2-(4-(pyridin-3-ylmethyl)piperazin-1-yl)ethanol] and JAY2-22-33 [2-(methyl(pyridine-3-ylmethyl)amino)-ethanol], improved working memory in a delayed match to sample task (DMTS) in monkeys. JWB1-84-1 also reduced the number of errors involved in completing a radial arm water maze (RAWM) task in AD-Tg mice that over-express Abeta beginning around 7 months of age (Sood et al., 2007). The effectiveness of the four aforementioned compounds (nicotine, cotinine, JWB1-84-1 and JAY2-22-33) in the DMTS task was linearly related to their effectiveness in producing desensitization of a nAChR agonist response in rats. Only nicotine evoked an agonist-like action in these studies indicating that it is possible to develop new chemical entities (e.g., choline analogs, cotinine analogs) that have the ability to desensitize nAChRs without an antecedent agonist action. Since the side effects of nicotine (e.g., cardiovascular, gastrointestinal) are often associated with its agonist effects, such an approach could offer the advantage of better tolerability.

4. Phosphodiesterase Inhibitors

Please see Table 2 for a summary of the variety of phosphodiesterase-based strategies for cognitive enhancement.

Table 2.

Phosphodiesterase Inhibitors

Initially, phosphodiesterase (PDE) inhibitors (e.g., papaverine) were examined for their ability to increase cerebral blood flow in an attempt to improve cerebral metabolism and thereby, improve cognitive function in aged patients (for review see Yesavage et al., 1979). More recently, research as concentrated on the ability of PDE inhibitors to modulate the second messenger molecules cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP). Eleven subclasses of PDE inhibitors (PDE1–PDE11) have been indentified with most having one or more isoforms (e.g., PDE4A, PDE4B, PDE4C and PDE4D) and each isoform may have multiple splice variants (e.g., PDE4D1–PDE4D9). Overall, more than 100 human PDEs exist (Bender and Beavo, 2006). All PDEs have a common catalytic domain, but PDE4, PDE7 and PDE8 hydrolyze cAMP whereas PDE5, PDE6 and PDE9 hydrolyze cGMP. PDE1, PDE2, PDE3, PDE10 and PDE11 are capable of hydrolyzing both cAMP and cGMP. PDE inhibitors (PDE-I) increase intracellular levels of cAMP and/or cGMP which can lead to gene transcription by activating the CREP signaling pathways (Impey et al., 1996; Lu and Hawkins, 2002; Tully et al., 2003). Activation of the cAMP/PKA/CREB and cGMP/PKG/CREB pathway has been implicated in synaptic plasticity such as long-term potentiation (LTP) and cognitive function (Frey et al., 1993; Son et al., 1998; Blokland et al., 2006). Despite 11 subclasses of PDEs, research has focused on PDE2, PDE4, PDE5, PDE9 and PDE10 and their potential involvement in learning and memory processes (see Rose et al., 2005 and Reneerkens et al., 2009 for a comprehensive review). To date, BAY 60–7550 is the only known selective PDE2-I that has been tested in animal models of cognition. BAY 60–7550 increased CA1 hippocampal LTP without affecting basal synaptic transmission (Boess et al., 2004). This PDE2-I improved object recognition acquisition and consolidation memory in rats and mice as well as social recognition memory performance in rats (Boess et al., 2004; Domek-Lopacinska and Strosznajder, 2008; Rutten et al., 2007). Cognitive enhancing effects of BAY 60–7550 was also observed in aged-impaired 24 month old rats in the object recognition task (Domek-Lopacinska and Strosznajder, 2008) and was shown to reverse the object recognition memory deficits of acute tryptophan depletion in rats (van Donkelaar et al., 2008). Moreover, BAY 60–7550 reversed the NMDA antagonist MK 801-induced working memory deficit in the spontaneous alternation T-maze task in mice (Boess et al., 2004). Of the PDE subtypes, PDE4 has been extensively characterized. PDE4-Is selectively inhibit the hydrolysis of cAMP and strong evidence exists for the cAMP/PKA/CREB pathway in mediating the cognitive effects of PDE4-I (Rose et al., 2005; Tully et al., 2003). Rolipram has been the most extensively studied PDE4-I. Rolipram has been shown to facilitate hippocampal LTP function in young and aged rodents (Barad et al., 1998; Navakkode et al., 2004), reverse β-amyloid (Aβ1-42) peptide impairment (Vitolo et al., 2002) and counteract the inhibitory effects of high levels of Aβ on synaptic plasticity in the transgenic APP/PS1 mouse model of AD (Gong et al., 2004). Behaviorally, rolipram has shown improvements in object recognition, spatial reference, working and associative memory behavioral tasks in young and aged rodents (Bach et al., 1999; Barad et al., 1998; Monti et al., 2006; Ramos et al., 2003; Rutten et al., 2006. Rutten et al., 2007) as well as improvements in working and executive memory function in young (Ramos et al., 2003; Rutten et al., 2008) but not aged adult monkeys (Ramos et al., 2003). Rolipram also restores working and reference memory in the radial arm maze task and associative memory in the passive avoidance task following administration of scopolamine (Egawa et al., 1997; Ghelardini et al., 2002; Imanishi et al., 1997; Zhang and O’Donnell, 2000), MK-801 (Zhang et al., 2000, 2005), MAPK/ERK kinase (MEK) inhibition (Zhang et al., 2004) or anisomycin (Randt et al., 1982). Further, rolipram is capable of reversing cognitive impairments in genetic mouse models of human disorders such as Rubinstein-Taybi syndrome (Bourtchouladze et al., 2003) and AD (Gong et al., 2004; Comery et al., 2005). In addition to rolipram, several newly developed PDE4-Is have been evaluated for their potential involvement in learning and memory processes. HT-0712 has been shown to increase cAMP thereby enhancing the CREB signaling pathway leading to improved memory performance in young mice in the object recognition and fear conditioning tasks as well as aged mice in a trace conditioning behavioral task (Bourtchouladze et al., 2003; Helicon Therapeutics.com). Moreover, HT-0712, like rolipram, was shown to reverse the object recognition deficit in a genetic mouse model of Rubinstein-Taybi syndrome (Bourtchouladze et al., 2003). L-454,560 improved cognitive performance in the delayed matching to position (DMTP) version of the Morris water maze task (Huang et al., 2007). MEM 1018 and MEM 1091 were efficacious in reversing the MK-801-induced working and reference memory deficits in the rat radial arm maze and each compound antagonized the amnesic effect of MK-801 on passive avoidance behavior (Zhang et al., 2005). Although PDE5-Is were originally developed for the treatment of erectile dysfunction (e.g., sildenafil (Viagra), vardenafil (Levitra) and tadalafil (Cialis); Setter et al., 2005), recent preclinical evidence has suggested a potential role in cognition. PDE5-Is selectively inhibit the enzymatic activity of PDE5 leading to increase intracellular levels of cGMP (Bender and Beavo, 2006). PDE5-Is have been shown to enhance memory function in the object recognition task in rats (Domek-Lopacinska and Strosznajder 2008; Prickaerts et al., 2002; 2005; Rutten et al., 2007) and mice (Rutten et al., 2005). The PDE5-I sildenafil also improved executive memory performance in monkeys using the prefrontal cognition object retrieval paradigm (Rutten et al., 2008). In pharmacological deficit models, PDE5-Is, sildenafil and vardenafil, have reversed recognition, spatial and associative memory performance impairments produced by acute tryptophan depletion (van Donkelaar et al., 2008), nitric oxide synthase (NOS) inhibition (Devan et al., 2006; Prickaerts et al., 1997), scopolamine (Devan et al., 2004; Shafiei et al., 2006) and diabetic neuropathy (Patil et al., 2004). Currently, only two PDE9-Is, SCH 81566 and BAY 73–6691, have been reported and only BAY 73–6691 has been characterized in animal models of cognition (van der Staay et al., 2008). PDE9-Is, like PDE5-Is, inhibit the degradation of the second messenger cGMP. BAY 73–6691 treatment increased hippocampal LTP function in young and aged rats and produced pro-cognitive effects in the object recognition and social recognition tasks in rats and mice. Further, BAY 73–6691 reversed the scopolamine-induced passive avoidance deficit as well as the MK-801-induced deficit in the working memory T-maze task (van der Staay et al., 2008). Lastly, PDE10A-Is have been suggested as a target for cognition particularly in the treatment of cognitive dysfunction associated with schizophrenia (Rodefer et al., 2005). PDE10A-Is regulate the activity of both cAMP and cGMP and while PDE10A-Is like papaverine, TP-10 (Schmidt et al., 2008) and MP-10 (Grauer et al., 2009) possess greater pharmacological similarity to that of the traditional antipsychotics (Grauer et al., 2009; Schmidt et al., 2008), there is evidence for a role in modulating cognitive function. Papaverine has been shown to reverse the executive memory deficits of subchronic phencyclidine following washout in the attentional set-shifting paradigm which is a rodent model analogous to the Wisconsin Card Sorting task in humans (Rodefer et al., 2005). Further, papaverine, but not MP-10 enhanced object recognition memory following a 48 hr delay interval in rats (Grauer et al., 2009) and both compounds attenuated the MK-801-induced social odor recognition deficits in mice (Grauer et al., 2009). Despite these encouraging results following acute administration of PDE10A-I, chronic administration of papaverine to mice disrupted spatial reference memory and reversal learning using the Morris water maze task (Hebb et al., 2008).

Overall, the preclinical evidence clearly demonstrates that PDE-Is for the PDE2, PDE4, PDE5, PDE9 and PDE10 subclasses are involved in learning and memory processes. It is now up to the pharmaceutical industry to develop safe and viable compounds to assess these preclinical effects on cognition in clinical trials in humans.

5. Serotonin Receptors

Please see Table 3 for a summary of the variety of serotoninergic-based strategies for cognitive enhancement.

Table 3.

Serotoninergic-Based Compounds

Compound Name Receptor Reference
5-HT1A
8-OH-DPAT agonist Meneses and Perez-Garcia, 2007; Orgen et al., 2008
Buspirone partial agonist Meneses, 1999; Orgen et al., 2008
MDL 73005 agonist Bertrand et al., 2001
S15535 agonist Millan et al., 2004
lecozotan/SRA-333 antagonist Schechter et al., 2005
NAD-299 antagonist Luttgen et al., 2005; Misane and Ogren, 2003
WAY 100635 antagonist Harder and Ridley, 2000; Misane and Ogren, 2003; Pitsikas et al., 2003
WAY 101405 antagonist Hirst et al., 2008
5-HT2A
MDL 100,907 antagonist Meneses, 1999; Roth et al., 2004; Williams et al., 2002; Winstanley et al., 2003
EMD 281014 antagonist Terry et al., 2005b
Ketanserin antagonist Levin and Rezvani, 2007
Mianserin antagonist Roth et al., 2004
5-HT3
DAU 6215 antagonist Pitsikas et al., 1996
Granisetron antagonist Naghdi and Harooni, 2005; Pitsikas and Borsini, 1997;
Ondansetron antagonist Arnsten et al., 1997; Barnes et al., 1999; Carli et al., 1997; Domeney et al., 1991; Pitsikas and Borsini, 1997
RS-56812 antagonist Terry et al., 1996
SEC 579 antagonist Meneses, 1999
WAY 100289 antagonist Hodges et al., 1995
5-HT4
RS 67333 partial agonist Chaillan et al., 2004; Fontana et al., 1997; Kulla and Manahan-Vaughan, 2002; Levallet et al., 2009
SC 53116 partial agonist Matsumoto et al., 2001
SL 65.0155 partial agonist Hille et al., 2008; Moser et al., 2002; Spencer et al., 2004
BIMU1 partial agonist Galeotti et al., 1998
VRX 03011 partial agonist Mohler et al., 2007
RS 17017 agonist Terry et al., 1998
GR 125487 antagonist Galeotti et al., 1998; Lamirault and Simon, 2001; Spencer et al., 2004
RS 67532 antagonist Chaillan et al., 2004; Fontana et al., 1997
SDZ 205,557 antagonist Galeotti et al., 1998
5-HT6
Ro-04-6790 antagonist King et al., 2004, 2005, 2009; Meneses, 2001; Woolley et al., 2004
Ro-4368554 antagonist Lschreiber et al., 2007
SB 271046 antagonist Callahan et al., 2004; Foley et al., 2004; King et al., 2004; Loiseau et al., 2008; Marcos et al., 2008; Rodefer et al., 2008; Routledge et al., 2000
SB 357134 antagonist Rogers and Hagan, 2001; Stean et al., 2002
SB 399885 antagonist Hatcher et al., 2005; Hirst et al., 2006; Rowe et al., 2008
SB 742457 antagonist Callahan et al., 2009; Maher-Edwards et al., 2010; Rowe et al., 2009; Upton et al., 2008
MEM 68626 antagonist Rowe et al., 2008
MEM 34551 antagonist Rowe et al., 2008
PRX-07034 antagonist Gannon et al., 2006
SB 742457 antagonist Callahan et al., 2009; Maher-Edwards et al., 2010; Rowe et al., 2009; Upton et al., 2008
EMD 386088 agonist Kendall et al., 2010; Meneses et al., 2008
E-6801 agonist Kendall et al., 2010; Romero et al., 2006
R-13c agonist Fone, 2008
WAY 181187 agonist Burnham et al., 2010; Callahan et al., 2009; Loiseau et al., 2008; Rowe et al., 2009; Schechter et al., 2008; West et al., 2009
WAY 208466 agonist Schechter et al., 2008
SB 399885 antagonist Hatcher et al., 2005; Hirst et al., 2006; Rowe et al., 2008

Serotonin (5-hydroxytryptamine; 5-HT) receptor systems are expressed throughout the peripheral and central nervous systems and are involved in a multitude of physiological functions such as cardiovascular and gastrointestinal regulation, thermoregulation, affective disorders as well as learning and memory (Barnes and Sharp, 1999; Cassel and Jeltsch, 1995). Within the 7 classes of 5-HT receptors (5-HT1–7) there are at least 13 distinct G protein-couple receptors (GPCRs) and one ligand-gated ion channel (5-HT3; Barnes and Sharp, 1999; Hannon and Hoyer, 2008). Several of the 5-HT receptor subtypes (i.e, 5-HT1A, 5-HT2A, 5-HT3, 5-HT4 and 5-HT6) have been implicated in learning and memory processes (for review see King et al., 2008; Terry et al., 2008) and are expressed in key brain regions (e.g., amygdala, cortex, hippocampus and striatum) where they modulate the neurotransmitter release of acetylcholine, dopamine, GABA and glutamate and noradrenaline (Barnes and Sharp, 1999).

5.1 5-HT1A receptors

The 5-HT1A receptor functions as both a somatodendritic inhibitory autoreceptor involved in raphe nuclei regulation as well as a postsynaptic heteroreceptor modulating neuronal release (Barnes and Sharp, 1999). For the most part, 5-HT1A receptor agonists (e.g., 8-OH-DPAT and buspirone) have been shown to disrupt learning and memory processes (Meneses and Perez-Garcia, 2007; Orgen et al., 2008), although there are reports of 5-HT1A receptor agonists (MDL 73005 and S15535) showing improvements in cognitive function (Bertrand et al., 2001; Millan et al., 2004). The real interest within 5-HT1A receptor pharmacology stems from the observations that selective “silent” 5-HT1A receptor antagonists (e.g., lecozotan (SRA-333), NAD-299, WAY 100635 and WAY 101405) reverse anatomical lesions (e.g., fornix and hippocampal) and pharmacological impairments (e.g., scopolamine and MK 801) as well as possess stand-alone improvements across a broad spectrum of behavioral models of cognition in rodents and non-human primates (Harder et al., 1996; Harder and Ridley, 2000; Hirst et al., 2008; Luttgen et al., 2005; Misane and Ogren, 2003; Pitsikas et al., 2003; Schechter et al., 2005). Although the exact mechanism underlying the observed pro-cognitive effects of 5-HT1A receptor antagonists is not completely known, it is thought that modulation of cholinergic and/or glutamatergic transmission is involved. Interestingly, the 5-HT1A receptor antagonist lecozotan (SRA-333) was shown to be safe and well tolerated in healthy young and elderly subjects (Patat et al., 2009) and a clinical phase II trial in mild to moderate AD patients was recently completed, though the outcome hasn’t been reported (Sabbagh, 2009).

5.2 5-HT2A receptors

5-HT2A receptors are expressed in brain regions (e.g., cortex, striatum and hippocampus) known to be involved in cognition and perception and 5HT2A receptor antagonists appear to be involved in cognitive processes (Terry et al., 2005b; Williams et al., 2002; Winstanley et al., 2003). For example, medial prefrontal cortex infusions with the selective 5HT2A receptor antagonist MDL 100,907 improved visuospatial attention and decreased impulsivity in the rat 5-choice serial reaction time task (Winstanley et al., 2003) and oral administration of the 5HT2A receptor antagonist EMD 281014 improved medium and long retention interval performance in the delayed matching-to-sample task in young and aged rhesus monkeys (Terry et al., 2005b). However, the 5HT2A receptor antagonist ketanserin failed to show improvements in the radial arm maze or operant signal detection task in rats and co-administration of ketanserin with either acute or chronic administration of nicotine attenuated the enhanced attentional performance produced by nicotine (Levin and Rezvani, 2007). Interestingly, the 5HT2A receptor antagonists mianserin and MDL 100,907 have shown modest, but promising effects on cognition in schizophrenic patients (Roth et al., 2004). Additional research and interest from the pharmaceutical community is required to truly assess the potential of this drug target.

5.3 5-HT3 receptors

The 5-HT3 receptor and in particular, 5-HT3 receptor antagonists have also generated interest as putative cognitive enhancers given their ability to increase acetylcholine release (Diez-Ariza et al., 2002; Ramirez et al., 1996) and improve certain aspects of cognitive function (Meneses, 1999; Terry et al., 2008). Overall, 5-HT3 receptor antagonists (e.g., DAU 6215, granisetron, ondansetron, RS-56812, SEC 579 and WAY 100579) appear to be efficacious in reversing pharmacologically-induced impairments (e.g., atropine, scopolamine and ibotenate acid forebrain lesions) as well as produce stand-alone improvements in cognitive function across multiple cognition behavioral models in young and aged rats (Barnes et al., 1990; Carli et al., 1997; Fontana et al., 1995; Hodges et al., 1995; Pitsikas and Borsini 1996; 1997; Pitsikas et al., 1993) and non-human primates (Arnsten et al., 1997; Barnes et al., 1990; Carey et al., 1992; Domeney et al., 1991; Terry et al., 1996). Despite these encouraging results, several studies have failed to support the pro-cognitive effects of 5-HT3 receptor antagonists (Bratt et al., 1994; Naghdi and Harooni, 2005; Pitsikas et al., 1993; Pitsikas and Borsini, 1997). Whereas ondansetron was found to be inactive in an AD clinical trial (Dysken et al., 2002), a renewed interest has been initiated for the cognitive dysfunction associated with schizophrenia (Akhondzadeh et al., 2009; Zhang et al., 2006).

5.4 5-HT4 receptors

The concentration of 5-HT4 receptors in limbic structures (e.g., cortex, amygdala, septum and hippocampus), along with modulation of cholinergic neuronal activity (Bockaert et al., 2004; Bonaventure et al., 2000; Vilaro et al., 2005) and neurochemical release of acetylcholine (Consolo et al., 2004; Matsumoto et al., 2001; Mohler et al., 2007) within these brain regions make it an attractive target for restoration of cognitive dysfunction. 5-HT4 receptors are positively linked to adenylate cyclase and activation leads to increased intracellular levels of cAMP, a fundamental component in synaptic long-term potentiation (LTP; Frey et al., 1993). Indeed, 5-HT4 receptor partial agonists (e.g., RS 67333, SC 53116 and SL 65.0155) enhance the activity of cAMP (Eglen et al., 1995) facilitate hippocampal (Kulla and Manahan-Vaughan, 2002; Marchetti et al., 2004; Matsumoto et al., 2001) and amygdala LTP function (Huang and Kandel, 2007). Moreover, 5-HT4 receptor agonists were capable of reversing hippocampal deficient LTP synaptic transmission in transgenic mice overexpressing the amyloid β peptide (Spencer et al., 2004). Behaviorally, 5-HT4 receptor agonists produce pro-cognitive effects in animal models of learning and memory (King et al., 2008; Meneses, 1999). The 5-HT4 receptor partial agonists (e.g., BIMU 1, SC 53116, RS 67333, SL 65.0155 and VRX 03011) have been shown to reverse the amnesic effects of scopolamine and atropine on passive avoidance and spatial navigation performance in rats and mice (Fontana et al., 1997; Galeotti et al., 1998; Matsumoto et al., 2001; Moser et al., 2002), enhance place and object recognition memory in young and aged rats (Lamirault and Simon, 2001; Levallet et al., 2009; Moser et al., 2002), enhance delayed spontaneous alternation behavior (Mohler et al., 2007) and reverse the aged related deficit observed in the linear maze in rats (Moser et al., 2002). Additionally, SL 65.0155 improved attentional performance in the rat 5-choice serial reaction time task (Hille et al., 2008) and RS 17017 enhanced attentional and working memory performance in young and aged monkeys in the delayed matching-to-sample task (Terry et al., 1998). The ability of selective 5-HT4 receptor antagonists (e.g., GR 125487, RS 67532 and SDZ 205,557) to block the pro-cognitive actions of 5-HT4 receptor agonists across several behavioral models strengthens the role of 5-HT4 receptors in cognitive processes (Fontana et al., 1997; Lamirault and Simon, 2001; Moser et al., 2002). Lastly, synergistic effects with cholinesterase inhibitors (e.g., donepezil, galanthamine and rivastigmine) and 5-HT4 receptor agonists have been observed in animal models of cognition (Cachard-Chastel et al., 2008; Mohler et al., 2007; Moser et al., 2002) suggesting therapeutic utility for AD. In addition to providing potential symptomatic relief, activation of the 5-HT4 receptor in vitro stimulates the secretion of the soluble form of amyloid precursor protein (sAPPα), a neuroprotective protein in the brain (Lezoualc’h 2007; Robert et al., 2001) and in vivo, 5-HT4 receptor agonists have been shown to increase sAPPα levels in cortex and hippocampus in young adult and transgenic APP-overexpressing mice (Cachard-Chastel et al., 2007; 2008). Taken together, these data provide strong preclinical support for 5-HT4 receptor involvement in the treatment of AD symptomatology and pathology.

5.5 5-HT6 receptors

The 5-HT6 G-protein coupled receptor was first cloned from rat striatum using RT-PCR techniques in the early 1990’s and its activation stimulates cAMP production and protein kinase A (Kohen et al., 1996; Monsma et al., 1993; Ruat et al., 1993). 5-HT6 receptor expression is almost exclusively within the central nervous system (CNS) thereby limiting any potential peripheral side effects. Receptor distribution resides within brain areas (e.g., striatum, cortex, hippocampus and hypothalamus) responsible for mediating many of its observed preclinical effects on anxiety and depression (Svenningsson et al., 2007; Wesolowska and Nikiforuk 2007), epilepsy (Routledge et al., 2000), obesity (Heal et al., 2008) and the current topic, learning and memory (Fone, 2008; King et al., 2008). Initial evidence supporting the involvement of 5-HT6 receptors in cognitive processes was derived from the finding that receptor knockdown after intracerebroventricular treatment with 5-HT6 receptor antisense oligonucleotides (AO) improved retention of the learned hidden platform position during probe trials in the water maze task in normal rats (Bentley et al., 1997; Woolley et al., 2001). The significance of this receptor blockade was later confirmed when administration of 5-HT6 receptor antagonists Ro-04-6790, SB 271046 and SB 357134 led to improved probe trial, but not acquisition learning performance in normal adult rats (Marcos et al., 2008; Rogers and Hagan, 2001; Stean et al., 2002; Woolley et al., 2001). Conversely, in aged rats 5-HT6 receptor antagonists are capable of enhancing both acquisition learning and retention probe trial performance (Foley et al., 2004; Hirst et al., 2006; Stean et al., 2002) suggesting that within the water maze task 5-HT6 receptor antagonists may have a greater influence on declining cognitive function especially as it relates to cholinergic activity. Indeed, these aged-related findings were extended by classifying the aged rat population as either being aged cognitively-impaired or aged cognitively-unimpaired based upon their acquisition water maze performance to that of young adult rats (Rowe et al., 2007). In these cognitively-impaired aged rats, hippocampal acetylcholine levels are significantly decreased compared to their cognitively-unimpaired cohort and therefore, represent a model of “natural” aged-related cognitive decline, akin to that observed in humans suffering from mild cognitive impairment (MCI) or AD (Quirion et al., 1995). Administration of 5-HT6 receptor antagonists (e.g., MEM 68626, MEM 34551, SB 271046 and SB 742457) completely reversed the acquisition learning and probe retention trial water maze performance deficits observed in this aged cognitively-impaired cohort (Callahan et al., 2004; Rowe et al., 2008, 2009). SB 742457 and MEM 68626 were also shown to reverse the spatial working memory deficits of aged cognitively-impaired rats in the 8-arm radial water maze task (Rowe et al., 2008). Moreover, synergistic effects were demonstrated with a sub-threshold dose of the acetylcholinesterase inhibitor donepezil in combination with SB 271046 on spatial memory performance (Callahan et al., 2004), whereby the drug combination enhanced acquisition and probe trial performance of the aged cognitively-impaired animal over that performance observed for either drug given alone. These results suggest that 5-HT6 receptor antagonists administered along with cholinesterase inhibitors may provided added beneficial effects in the treatment of cognitive dysfunction in humans. In addition to spatial reference and working memory improvements, 5-HT6 receptor antagonists are active across multiple behavioral models reflecting various cognitive domains (e.g., episodic, associative, executive function, and working memory) and brain regions (e.g., cortical, hippocampal, striatal and amygdalar). 5-HT6 receptor antagonists reverse pharmacologically-induced deficits (e.g., scopolamine, MK 801 and phencyclidine) as well as possess stand-alone effects in young and aged rats in the following tasks, object recognition (Callahan et al., 2004, 2009; Gannon et al., 2007; Hirst et al., 2006; King et al., 2004,2005, 2009; Schreiber et al., 2007; Wolff et al., 2002), social recognition (Loiseau et al., 2008; Mitchell et al., 2006; Schreiber et al., 2007), social discrimination (Schreiber et al., 2007), operant autoshaping (Meneses 2001; Perez-Garcia and Meneses, 2005; Schreiber et al., 2007), passive avoidance (Bos et al., 2001; Callahan et al., 2004; Foley et al., 2004; Riemer et al., 2003; Schreiber et al., 2007), attentional set-shifting (Hatcher et al., 2005; Rodefer et al., 2008) and radial arm maze (Wolff et al., 2002). Moreover, the pro-cognitive effects observed in rodents have been extended to include non-human primates, though a limited number of studies of been published to date. SB 271046 was shown to improve accuracy in aged rhesus monkeys in the delayed matching-to-sample task and in marmosets, SB 271046 reversed the MK 801-induced deficits on both perceptual visual and visuospatial discrimination tasks (Upton et al., 2008). Many of the behavioral effects on cognition in young and aged animals can be attributed to the ability of 5-HT6 receptor antagonists to increase the release of acetylcholine (Hirst et al., 2006; Riemer et al., 2003; Shirazi-Southall et al., 2002; Zhang et al., 2007), glutamate (Dawson et al., 2000; Dawson and Nguyen, 2001) and the monoamines (Lacroix et al., 2004; Li et al., 2007) in brain regions involved in learning and memory processes. Collectively, these preclinical findings along with the recent positive phase II clinical trail effects with SB 742457 in mild to moderate AD subjects (Maher-Edwards et al., 2010; Upton et al., 2008) suggest that 5-HT6 receptor antagonists may be viable drug candidates for the symptomatic relief of cognitive dysfunction associated with age-related cognitive decline, AD and possibly, schizophrenia.

More recently, 5-HT6 receptor agonists (e.g., EMD 386088, E-6801, R-13c, WAY 181187 and WAY 208466) have been identified (Cole et al., 2005; Mattsson et al., 2005; Romero et al., 2006; Schechter et al., 2008) and their potential involvement on cognitive processes assessed (Burnham et al., 2010; Callahan et al., 2009; Fone, 2008; Kendall et al., 2010; Loiseau et al., 2008; Meneses et al., 2008; Rowe et al., 2009). Initial studies have yielded controversy with reports showing pro-cognitive effects on attentional set-shifting (Burnham et al., 2010) and object recognition (Fone, 2008; Kendall et al., 2010) while others have demonstrated either cognitive impairment on social recognition (Loiseau et al., 2008) and autoshaping (Meneses et al., 2008) or no effects on cognition in object recognition, passive avoidance and water maze (Callahan et al., 2009; Rowe et al., 2009). In many cases, co-administration of a selective 5-HT6 receptor antagonist has been used to either reverse the agonist-induced deficit (Loiseau et al., 2008) or agonist-induced improvement (Burnham et al., 2010). In the object recognition task, Kendall and colleagues (2010) showed that both 5-HT6 receptor agonists and antagonists enhanced recognition memory at a 4 hr delay and that combining sub-optimal doses of each led to enhanced memory performance, whereas Callahan and colleagues (2009) showed that object recognition memory was neither affected at a short 1 hr delay or enhanced at a 48 hr delay by the 5-HT6 receptor agonist WAY 181187. Moreover, the pro-cognitive effect observed at the 48 hr delay interval by the antagonist SB 742457 was dose-dependently blocked by co-administration of WAY 181187. Further, whereas SB 742457 dose-dependently reversed the age-related cognitive impairment in Fischer rats, WAY 181187 was ineffective in reversing the natural age-related cognitive deficit (Rowe et al., 2009). Given that these investigators have shown pro-cognitive effects with 5-HT6 receptor antagonists (as well as other drug classes) in the same behavioral models used to characterize 5-HT6 receptor agonists, simply stating methodological differences across laboratories appears to be an unlikely reason for the present disparity. Additionally, one might argue that the differences stems from the use of non-selective 5-HT6 receptor agonists, however many of the investigators have used the same 5-HT6 receptor agonist, WAY 181187 (Burnham et al., 2010; Callahan et al., 2009; Loiseau et al., 2008; Rowe et al., 2009), although a few have reported on less well characterized compounds (e.g., EMD 386088, E-6801 and R-13c; Fone, 2008; Kendall et al., 2010; Meneses et al., 2008) especially with respects to neurochemical profiling. For example, the high affinity and receptor selective 5-HT6 agonist WAY 181187 has been showed to increase GABA release in rat frontal cortex, hippocampus, striatum and amygdala. Effects that are blocked by SB 271046 administration (Schechter et al., 2008), thus supporting the proposed disinhibitory action by 5-HT6 receptor antagonists on cholinergic and glutamatergic activity. WAY 181187 has also been shown to decrease dopamine and serotonin release in rat cortex and striatum and inhibit potassium-stimulated glutamate release from hippocampal slices (Schechter et al., 2008), effects opposite to 5-HT6 receptor antagonists. Additionally, WAY 181187 has been shown to attenuate CA1 hippocampal LTP function, an effect blocked by SB 399885 (West et al., 2009). Generally, pro-cognitive drugs (e.g., 5-HT4 receptor agonists, Matsumoto et al., 2001) enhance LTP activity which represents the synaptic basis for learning and memory (Bliss and Collingridge, 1993). Taken together, these data suggest that 5-HT6 receptor agonists would either have no effect on cognition or impair cognitive performance. Conversely, it’s been argued that the cognitive enhancing properties of 5-HT6 receptor agonists occur by direct activation of 5-HT6 receptors located on cholinergic and/or glutamatergic neurons, however autoradiography (Roberts et al., 2002), immunohistochemical (Woolley et al., 2004) and cholinergic lesion studies (Marcos et al., 2006) suggest little 5-HT6 receptor expression on cholinergic neurons (as well as glutamate neurons) and that the observed increase in acetylcholine and glutamate release by 5-HT6 receptor antagonists is due to an indirect disinhibitory effect on GABAergic neurons. If 5-HT6 receptor agonists directly modulate cholinergic and/or glutamatergic release then microdialysis studies will provide the much needed proof of concept for the proposed mechanism of action. Alternatively, 5-HT6 receptor agonists could be activating second messenger intraneuronal signaling cascades such as cAMP and/or cGMP which are widely known to be involved in learning and memory processes (Reneerkens et al., 2009; Rose et al., 2005). 5-HT6 receptors are functionally coupled to cAMP and agonists would be expected to facilitate cAMP production triggering downstream protein kinase A (PKA) or mitogen-activated protein (MAP) kinase activity thereby leading to activation of transcription factor cAMP-response element binding protein (CREB) and subsequently gene expression and protein synthesis which is necessary for long-term memory formation (Tully et al., 2003). 5-HT4 receptors, like 5-HT6 receptors, trigger this cAMP/PKA signaling pathway which is regulated by phosphodiesterases (PDEs) and it was recently demonstrated that the cognitive improvement in object recognition memory by the 5-HT4 receptor agonist RS 67333 was due to a direct stimulation of prefrontal cortex and hippocampal PDE activity (Levallet et al., 2009). It is therefore plausible to suggest that the observed pro-cognitive effects associated with 5-HT6 agonists may involve modification of this second messenger system and in particular, PDE activity. Similar studies will need to be executed with 5-HT6 agonists to verify this potential mechanism of action.

6. Histamine H3 Receptor Antagonists

Histamine is an important neurotransmitter involved in many physiological functions (e.g., allergies, gastric acid secretion, immunomodulation and neurotransmitter release; Leurs et al., 2005). Histamine exerts its effects through four G-protein-coupled histamine receptor subtypes (H1, H2, H3 and H4).The histamine H3 receptor (H3R) and, in particular, H3R antagonists are being pursued as potential drug targets for the treatment of cognitive dysfunction associated with age-related cognitive decline, AD, attentional deficit hyperactivity disorder (ADHD) and schizophrenia (Esbenshade et al., 2008; Witkin and Nelson, 2004). H3Rs are expressed almost exclusively in mammalian brain with high densities in cortex, hippocampus, striatum and hypothalamus, areas involved in wakefulness, attention, vigilance and cognition (Martinez-Mir et al., 1990; Pollard et al., 1993). H3Rs are presynaptic autoreceptors that regulate the synthesis and release of histamine from histaminergic neurons (Arrang et al., 1983; 1987). H3Rs also function as heteroreceptors and mediate the release of not only histamine but other neurotransmitters such as acetylcholine, dopamine, noradrenaline and serotonin (Leurs et al., 2005; Schlicker et al., 1994) and inhibition of H3Rs with selective antagonists can augment the release of neurotransmitters involved in cognition. (Fox et al., 2005; Galici et al., 2009; Medhurst et al., 2007a). Due to the high constitutive activity of native H3Rs and the ability of H3R antagonists to inhibit G-protein-coupled receptor signaling, some H3R antagonists have been termed “inverse agonist” (e.g., ciproxifan, thioperamide, ABT-239, BF 2.649, GSK 189254; Fox et al., 2005; Leurs et al., 2005; Ligneau et al., 2007; Medhurst et al., 2007a), however, not all H3R antagonists show this intrinsic activity and are considered to be “neutral” antagonists (e.g., JNJ- 5207852; JNJ-10181457; Barbier et al., 2004; Galici et al., 2009).

In preclinical behavioral models of cognition, a variety of H3R antagonists have proven to be efficacious in restoring and/or enhancing cognitive performance (for comprehensive reviews see Esbenshade et al., 2008; Leurs et al., 2005; Witkin and Nelson 2004). For example, studies with the first generation H3R antagonists such as thioperamide, ciproxifan or clobenpropit were shown to reverse passive avoidance (Giovannini et al., 1999; Miyazaki et al., 1997), object recognition (Giovannini et al., 1999; Pascoli et al., 2009), two-choice discrimination water maze, Barnes maze (Komater et al., 2005) and radial arm maze (Chen, 2000) impairments produced by scopolamine treatment. Additional support for cholinergic modulation came from the observation that newly developed H3R antagonists like BF 2.649, GSK 207040, GSK 334429, GSK 189254 and JNJ-10181457 were also efficacious in reversing scopolamine-induced passive avoidance (Medhurst et al., 2007a,b), object recognition (Ligneau et al., 2007) and delayed non-matching to position (DNMTP; Galici et al., 2009) performance deficits. Working and reference memory deficits produced by MK-801 administration have also been attenuated by clobenpropit in the rat radial arm maze task suggesting an interaction of histamine on NMDA function (Huang et al., 2004). H3R antagonists are also capable of enhancing cognitive performance of young and aged animals in the absence of pharmacological impairment. Administration of ciproxifan has been shown to improve attention and decrease impulsivity in the rat 5-choice serial reaction time test (5-CSRTT) which is analogous to the continuous performance test (CPT) widely used to assess attention in humans (Day et al., 2007; Ligneau et al., 1998). Thioperamide, BF 2.649 and GSK 189254 have been shown to enhance object recognition memory in young rats and mice when drug is given prior to training (Ligneau et al., 2007; Medhurst et al., 2007a; Rowe et al., 2006) but a recent report by Pascoli and colleagues (2009) using ciproxifan and thioperamide showed that recognition memory was only enhanced when drug was given immediately before the retention test session. The authors indicated that the H3R antagonists were unable to enhance recognition memory when the drugs were given prior to or immediately after the training trial. Additional work is required to clarify these observed differences. The H3R antagonists A-304121, A-317920, ABT-239 and JNJ-10181457 improved attention in a repeated acquisition inhibitory avoidance task in spontaneously hypertensive (SHR) rat pups (Bonaventure et al., 2007; Fox et al., 2003; 2005) and A-304121, A-317920 and ABT-239 also improved short-term memory recall in young adult (Fox et al., 2003; 2005) and aged rats (Fox et al., 2005) with a potency similar to that of ciproxifan. ABT-239 was also able to reverse the scopolamine-induced spatial memory deficit in a two-choice discrimination water maze task (Fox et al., 2005). Similarly, GSK 189254 improved water-maze acquisition latency and probe trial performance in aged rats (Medhurst, 2007a). Moreover, when aged rats are screened and classified as either aged cognitively-impaired or aged cognitively-unimpaired based upon their water-maze acquisition latency performance to that of young rats and testing occurs in the aged cognitively-impaired cohort, the H3R antagonist thioperamide is capable of completely reversing the acquisition latency and probe trial performance deficits (Rowe et al., 2006). Rowe and colleagues (2006) extended this positive effect of thioperamide on cognitive restoration of spatial memory in aged cognitively-impaired mice and thioperamide also improved avoidance learning in senescence-accelerated mice (Meguro et al., 1995). In addition to the positive behavioral effects of H3R antagonists across multiple cognitive domains and the therapeutic possibilities to treat ADHD, age-related cognitive decline, AD and various sleep disorders, H3R antagonists may also play an important role in the treatment of schizophrenia. H3R antagonists have been demonstrated to reduce the hyperlocomotor effects of dopamine agonists (e.g., amphetamine, apomorphine and methamphetamine; Akhtar et al., 2006; Clapham and Kilpatrick, 1994; Fox et al., 2005), reduce apomorphine-induced climbing behavior (Akhtar et al., 2006) and in some cases (Akhtar et al., 2006; Pillot et al., 2002) but not all (Zhang et al., 2005) potentiate the cataleptic effects of antipsychotics. H3R antagonists have also been shown to reverse the inherent sensorimotor gating deficits of DBA/2J mice in prepulse inhibition of startle (PPI; Browman et al., 2004) and hippocampal N40 sensory gating (Fox et al. 2005). Additionally, the H3R antagonist GSK 207040 attenuated the isolation rearing-induced deficit in PPI in rats (Southam et al., 2009) and GSK 189254 enhanced cognitive flexibility and executive memory function in the attentional set-shifting paradigm (Medhurst et al., 2007a). Taken together, these data strongly support a role for H3R antagonists in modulating the neurophysiological attributes associated with schizophrenia.

7. Multiple Drug Targets and Multifunctional Compounds

In complex illnesses such as AD, it could be argued that a multiple drug target approach to therapy is necessary to address the complex pathophysiology of the disease and its diverse symptoms. However, even if the strategy of combining drugs with different therapeutic targets is feasible, the development of multi-functional compounds would circumvent the challenge of administering multiple drugs with potentially different degrees of bioavailability, pharmacokinetics, and metabolism (reviewed, Youdim and Buccafusco, 2005). An additional advantage to single drugs with multiple actions is the simplification of the therapeutic regimen and improved compliance (an important consideration, especially for individuals who suffer from memory-related disorders). Another advantage is the potential to treat multiple symptoms including both cognitive deficits and non-cognitive behavioral symptoms. This is especially important given that the currently available AD therapeutic agents (in addition to modest efficacy and adverse side effects), do not effectively treat the very problematic non-cognitive behavioral symptoms (e.g., agitation) of the illness. Such behavioral symptoms are thus; often managed clinically by potent antipsychotic drugs which are particularly problematic in older patients since they are associated with movement disorders and/or a wide variety of metabolic abnormalities (e.g., weight gain, hyperglycemia, hyperlipidemia, reviewed, Myamoto et al., 2005) as well as an increase mortality (Ballard et al., 2009). Thus, a compound exhibiting pro-cognitive and antipsychotic properties might be specialty valuable in AD as well as conditions like schizophrenia which is characterized by abnormal behavioral symptoms as well as cognitive deficits. There would also be a number of advantages to an agent that exhibits both pro-cognitive and neuroprotective activity or other disease modifying action. The obvious advantages would be enhanced cognition (and improved day to day quality of life) while at the same time delaying or preventing the progression of the illness (properties not observed in the currently available therapies). One potential advantage of a multifunctional compound over a disease-modifying agent that has no acute pro-cognitive actions pertain to the design and length of clinical trials (reviewed, Buccafusco, 2009). Clinical trials of disease modifying agents (by definition) require long evaluation periods which are often cost-prohibitive. A pro-cognitive compound (with disease-modifying actions) could be approved for cognition enhancement in shorter clinical trials, providing a return on the manufacturer’s investment while the more lengthy evaluations of disease-modifying actions could be carried out.

7.1 Ladostigil

Ladostigil is a multi-functional compound designed to incorporate the cognitive enhancing properties of rivastigmine (Ohara et al., 1997; Ballard and McAllister, 1999; Wesnes et al., 2002) with the neuroprotection properties of rasagiline (Maruyama et al., 2001; Weinreb et al., 2004). As such, the carbamate moiety of rivastigmine was introduced in the 6 position of rasagiline and a compound conveying inhibition at both acetylcholinesterase and monoamine oxidase (MAO) was born (Weinstock et al., 2000; Sterling et al., 2002; Buccafusco et al., 2003). MAO is a family of enzymes which catalyze the oxidation of biogenic amine neurotransmitters such as norepinephrine, dopamine and 5-HT. Ladostigil is a brain specific inhibitor of MAO-A and MAO-B and has thus been linked to an increase norepinephrine, dopamine and 5-HT levels in the brain after administration (Buccafusco et al., 2003; Sagi et al., 2003; Weinstock et al., 2003). Because of this MAO inhibition, ladostigil has been shown to be effective in animal models to assess antidepressant activity. (Weinstock et al., 2002) More importantly to the treatment of AD, ladostigil has been shown to be neuroprotective similar to other MAO inhibitors (Weinstock et al., 2001; Yogev-Falach et al., 2002; Sagi et al., 2003). Another interesting neuroprotective property of ladostigil centers on its ability to modulate APP processing through mitochondrial membrane potential stabilization and regulation of PKC and mitogen-activeated protein kinase (MAPK) dependant signaling pathways (Yogev-Falach et al., 2002; Weinreb et al., 2008; Bar-Am et al., 2009) to reduce the production of neurotoxic amyloid beta (Yogev-Falach et al., 2006). Ladostigil also increases catalase activity and has thus been shown to protect against oxidative stress related apoptosis (Weinreb et al., 2008). Further, ladostigil can convey neuroprotection by increasing brain-derived nerve factor (BDNF) mRNA expression and production (Van der Schyf et al., 2007). Ladostigil has also shown pro-cognitive effects in a wide range of preclinical animal models (Buccafusco et al., 2003; Shoham et al., 2007; Luques et al., 2007; Weinstock et al., 2009) Administration of ladostigil was capable of preventing glial activation and cognitive deficits in object and place recognition tasks in a streptozotocin (STZ) model of AD (Shoham et al., 2007). More recently, ladostigil has been shown to enhance spatial memory in the Morris water maze by increasing proNGF, prevent age related increases in activation of astrocytes and microglia, and prevent age related reductions in cortical AChE activity in the hippocampus of aged rats. (Weinstock et al., 2009) Ladostigil has also shown pro-cognitive effects in a delayed matching-to-sample (DMTS) distracter task, as well as a DMTS titrating task in monkeys (Buccafusco et al., 2003). Similarly, ladostigil has shown positive cognitive effects in a cytochrome oxidase model of AD by preventing the decrease in choline acetyltransferase (CHaT) and restoring memory deficits in a object recognition task (Luques et al., 2007). Due to ladostigil’s positive effects on such a wide range of Alzheimer’s related targets (i.e. neuroprotective activity, regulatory effects on APP processing, stimulatory effects on PKC and MAPK, effects on BDNF production), it is easy to see why taking a more encompassing approach in future drug design could be beneficial in AD drug development.

7.2 Dimebolin

Dimebolin originally developed and used for its anti-histaminergic properties since the early 1980’s in Russia, has recently enjoyed a new found interest as a potential treatment for AD. Dimebolin is a multifunctional drug enjoying a broad range activity profile on many targets relating to the treatment of AD. Dimebolin is a weak acetylcholinesterase (IC50 = 7.9 µM) and butyrylcholinesterases (IC50 = 42 µM) inhibitor (Bachurin et al., 2001). The compound also displays antagonistic properties at the glutamatergic NMDA (Bachurin et al., 2001; Wu and Li, 2008) receptor and has shown positive modulation of glutamate AMPA receptors (Grigorev et al., 2003). Further, dimebolin administration is able to protect neurons from the damaging effects of excessive cell stimulation through antagonistic action (IC50 = 50 µM) at L-type Ca2+ channels (Lermontova et al., 2001). Dimebolin is also an antagonist to the brain 5-HT6 receptor and has been shown to improve social recognition memory similar to other 5-HT6 receptor antagonists (Schaffhauser et al., 2009). Dimebolin has also shown to be neuroprotective through attenuating amyloid beta induced neurotoxicity in rat cortical neuron cultures (Lermontova et al., 2001) and through its regulation of mitochondrial pores after neurotoxic insult (Bachurin et al., 2003). Dimebolin has also shown pro-cognitive effects on behavior in a senescence-accelerated mouse prone 10 (SAMP 10) model of aging by enhancing short-term memory assessed by a passive avoidance task (Grigorev et al., 2003). Similarly, the compound has been shown to improve cognition in a rat novel object recognition task by improving retention punitively through 5-HT6 receptor antagonism. However, the exact mechanism through which dimebolin produces its cognitive enhancing effects is not know. Still dimebolin is active at many targets (AChE inhibition, NMDA antagonism, H3 receptor antagonism, and 5-HT6 receptor antagonism) that related to cognitive enhancement and has shown positive results in several preclinical models of cognition as well as in AD patients (Doody et al., 2008.) Other reports have failed to see this improvement in AD patients treated with dimebolin (Miller, 2010). Weather or not dimebolin will prove itself as a viable treatment for AD is still uncertain. However, there remains an active interest in finding multi-functional drugs to treat AD.

7.3 JWS-USC-75-IX

Several years ago, our collaborators (Valli et al., 1992) synthesized a series of analogs of the ranitidine (an H2 antagonist commonly used to treat duodenal ulcers) for the purpose of creating non-toxic AChEIs. The impetus for this work was the prior observation that ranitidine (a histamine H2 antagonist commonly used to treat duodenal ulcers) had mild AChEI properties in vitro (Gwee and Cheah, 1986). Several of the compounds from this series of ranitidine analogs were subsequently found to possess potent AChEI properties as well as low toxicity profiles. Of this series, one compound, JWS-USC-75-IX (3-[[[2-[[(5-dimethylaminomethyl)-2-furanyl]methyl]thio]ethyl]amino]-4-nitropyridazine) was found to possess AChEI activity in vitro (IC50 ~ 0.47 µM) as well as potent M2 receptor antagonist activity in mouse cerebral cortex (IC50 ~ 60 nM). As had been suggested previously (Quirion, 1993; Mash et al., 1985), an M2 (autoreceptor) antagonist might be very useful given in conjunction with an AChEI in AD to prevent the acetylcholine from decreasing its own release. JWS-USC-75-IX, thus, combined both features in a single molecule and offered a significant potential for improved reliability as a treatment of diseases where cholinergic function is impaired (e.g., AD). Later we (Terry et al., 1999) evaluated JWS-USC-75-IX for effects in memory-related tasks in rodents. The compound was found to enhance spatial learning, inhibitory avoidance, and working memory. More recent studies in our laboratories indicate that JWS-USC-75-IX has the potential to improve other domains of cognition in rodents (e.g., recognition memory) as well as attention and working memory in non-human primates (Terry et al., 2011).

7.4 Additional Multifunctional Compounds

The compound memoquin resulted from a multi-targeted approach, combining the radical scavenger moiety of coenzyme Q (CoQ) with the polyamine backbone of caproctamine conferring AChE inhibition properties. The rationale was to produce a single molecular entity that combined the neuroprotective qualities of CoQ with the cognitive enhancing properties of AChE inhibition. The result was a molecule that has been shown to simultaneously reduce the neurodegenerative pathology and rescue the behavioral impairment in an object recognition task in a mouse (AD11 mice) model of AD (Cavalli, 2007). Similarly, PMS777 is a multiple target AChE inhibitor that displays dual inhibition of AChE and platelet-activating factor (PAF). PAF is thought to participate in the neurodegeneration related AD (Bazan, 2002; Bate, 2004). Thus, the rationale for PMS777 is to act through neuroprotectention as well as cognitive enhancement. PMS777 has been shown to alleviate scopolamine induced impairment in mice (Li, 2006).

8. Other Targets

For several years chromatin modifications, especially histone-tail acetylation, have been implicated in modulating synaptic plasticity and long-lasting changes of neural circuits that contribute to memory formation. Moreover, increased histone-tail acetylation induced by inhibitors of histone deacetylases (HDACis) has been observed to facilitate learning and memory in wild-type mice as well as in mouse models of neurodegeneration (Guan et al., 2009). Accordingly, selective inhibitors HDAC (e.g., Bayer’s compound EVP 0334) have been suggested as treatments for neurodegenerative diseases, including AD (Hannen et al., 2008).

Both angiotensin converting enzyme (ACE) inhibitors and angiotensin receptor blocker (ARB) drugs have been shown to improve cognitive function in animal models (reviewed, Gard, 2004) and as a result have been in various stages of development for AD over the last 15 years. While no such agent has progressed very far in clinical trials to date, recent observations that ARBs are associated with a significant reduction in the incidence and progression of AD and dementia (Li et al., 2010) may suggest that such a strategy remains viable.

While psychostimulent drugs (e.g., amphetamine, methylphenidate) have been employed for many years as effective treatments for ADHD and narcolpepsy, their potential as pro-cognitive agents in other disorders has been equiviocal. Interest in newer ADHD and narcopsy-related treatments such as the norepinephrine transporter (NET) inhibitor atomoxetine and the psychostimulant modafinil, respectively, is accruing and based on recent published evidence. For example atomoxetine was observed to improve reversal learning in rats and monkeys (Seu et al., 2009) while modafinil has been overved to improve gognition in several domains including working memory and episodic memory in both rodent models and humans (reviewed, Minzenberg and Carter, 2008).

There is a host of additional therapeutic targets that have been investigated or are in various stages of investigation for the symptomatic treatment of AD, MCI and cognitive deficits associated with schizophrenia. Unfortunately, a full overview of these targets is beyond the scope of the present review and we refer the reader to additional articles (Gallagher et al., 1985; Minzenberg and Carter, 2008; Buccafusco, 2009; Lynch et al., 2011;) that discuss examples such as glycine transporter related drugs, mGluR2/3 agonists, somatostatin receptor ligands, estrogen receptor ligands, D2 agonists, calcium channel modulators (e.g., MEM 1003, nimodipine), adrenergic compounds (e.g., nicergoline), opioid antagonists (e.g., naloxone) and GABA-related compounds.

9. Animal Models and Their Translational Value

In the last several years there has been a growing concern over the discrepancies between preclinical results in AD-related animal models and clinical evaluations of novel AD-related compounds (Carlsson, 2008). A more general concern over the translation of preclinical results to clinical trial data is now evident in multiple health-related disciplines (see Lowenstein, 2009; Simon, 2008). It is clear that new and more rigorous efforts to develop animal models that meet the demands of face, construct, and predictive validity be undertaken so that preclinical endeavors become more translational. It is also our opinion that specific batteries of behavioral tasks that more closely map onto the domains of cognition that are evaluated in clinical trials be developed and that some type of composite score approach to the animal tests be created that is analagous to those used in clinical studies (e.g., Mini-Mental Status Exam) so that an “overall” drug effect on cognition can be estimated. It is also very important that the full results of failed clinical trials be published in peer reviewed journals (a practice that is uncommon at present) so that the human and animal data can be carefully compared. Currently, such clinical trial results are often found only in abstract form or as summary data in reviews or lay publications, a practice that often makes it difficult for the science community to understand the reason for the so called “failure” of an investigational new drug (see review, McArthur et al., 2010).

10. Summary and Conclusions

Given the rapid growth of our elderly population and their concerns about memory loss and dementia it is incumbent on the scientific community to develop more effective pro-cognitive drugs. As diagnostic criteria for memory disorders evolve, the demand for pro-cognitive therapeutic agents is likely to move beyond AD and dementia to include MCI and potentially even less severe forms of memory decline. While there are a considerable number of compounds in various stages of preclinical development as pro-cognitive agents, due to the complexity of human cognition (and illness like AD that devastate cognition), the development of compounds that are both safe and effective (with sustained pro-cognitive effects over time) will likely require the diligent efforts of academia and private industry and the full armamentarium of a variety of scientific methods. Such methods include modern drug discovery techniques (e.g., molecular modeling, combinatorial chemistry, high throughout biological assays), the latest advances in molecular biology and transgenics, and more traditional research methods such as medicinal chemistry, behavioral neuroscience, and classical pharmacology.

Table 4.

Histamine (H3) Receptor Ligands and Miscellaneous Categories

Compound Name Receptor/Mechanism Reference
H3 Receptor Ligands
ciproxifan inverse agonist/antagonist Day et al., 2007; Ligneau et al., 1998; Pascoli et al., 2009
thioperamide inverse agonist/antagonist Giovannini et al., 1999; Meguro et al., 1995; Rowe et al., 2006
ABT-239 inverse agonist/antagonist Esbenshade et al., 2005, 2008; Fox et al., 2005
BF 2.649 inverse agonist/antagonist Ligneau et al., 2007
GSK 189254 inverse agonist/antagonist Medhurst et al., 2007a
GSK 207040 Antagonist/antagonist Medhurst et al., 2007b; Southam et al., 2009
GSK 334429 Antagonist/antagonist Medhurst et al., 2007b
JNJ- 5207852 neutral antagonist Barbier et al., 2004; Bonaventure et al., 2007
JNJ-10181457 neutral antagonist Bonaventure et al., 2007
clobenpropit inverse agonist/antagonist Giovannini et al., 1999; Huang et al., 2004
A-304121 inverse agonist/antagonist Esbenshade et al., 2008; Fox et al., 2003
A-317920 inverse agonist/antagonist Esbenshade et al., 2008; Fox et al., 2003
ABT-239 inverse agonist/antagonist Esbenshade et al., 2005, 2008; Fox et al., 2005
Multi-Functional Compounds
Ladostigil AChEI/MAOI Reviewed, Buccafusco, 2003; Reviewed, Youdim, 2006
Dimebolin AChEI/ NMDA, H3, and 5HT6 antagonist Bachurin, 2001; Grigorev, 2003; Schaffhauser, 2009
JWS-USC-75IX AChEI/M2 antagonist Terry et al., 1999; Terry et al., 2011
memoquin AChEI/Coenzyme Q activity Cavalli, 2007; Bolognesi, 2009
PM5777 AChEI/Platelet activating factor inhibitors Li, 2007
Other Agents and Targets
EVP 0334 HDAC Inhibitor Hannen et al., 2008
Angiotensin converting enzyme inhibitiors and receptor blockers Reviewed, Gard, 2004
glycine transporter, mGluR2/3, somatostatin receptor, estrogen receptor, D2 receptor, calcium channels receptors, GABA Reviewed, Buccafusco, 2009

Acknowledgments

The authors would like to thank Ms. Ashley Davis for her administrative assistance in preparing this article. This corresponding author’s laboratories are supported by the National Institute of Aging (AG032140 and AG029617), the National Institute of Environmental Health Sciences (ES012241), and the National Institute on Drug Abuse (DA029127). The corresponding author has also provided consultation or performed research (relevant to this article) in the last three years either contractually or in collaboration with a several pharmaceutical companies including Abbott Laboratories, Helicon, Merck, Neuralstem, Neurosearch, Roche Palo Alto, and Servier. This review is dedicated to our close personal friend, mentor, and colleague, the late Dr. Jerry J. Buccafusco whose contributions to the science of drug discovery and development for disorders of cognition spanned more than 25 years.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  1. Akhondzadeh S, Mohammadi N, Noroozian M, Karamghadiri N, Ghoreishi A, Jamshidi AH, et al. Added ondansetron for stable schizophrenia: a double blind, placebo controlled trial. Schizophr Res. 2009;107:206–212. doi: 10.1016/j.schres.2008.08.004. [DOI] [PubMed] [Google Scholar]
  2. Akhtar M, Uma Devi P, Ali A, Pillai KK, Vohora D. Antipsychotic-like profile of thioperamide, a selective H3-receptor antagonist in mice. Fundam Clin Pharmacol. 2006;20:373–378. doi: 10.1111/j.1472-8206.2006.00411.x. [DOI] [PubMed] [Google Scholar]
  3. Albuquerque EX, Pereira EF, Alkondon M, Rogers SW. Mammalian nicotinic acetylcholine receptors: from structure to function. Physiol Rev. 2009;89:73–120. doi: 10.1152/physrev.00015.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Araujo JA, Studzinski CM, Milgram NW. Further evidence for the cholinergic hypothesis of aging and dementia from the canine model of aging. Prog Neuropsychopharmacol Biol Psychiatry. 2005;29(3):411–422. doi: 10.1016/j.pnpbp.2004.12.008. [DOI] [PubMed] [Google Scholar]
  5. Arnsten AF, Lin CH, Van Dyck CH, Stanhope KJ. The effects of 5-HT3 receptor antagonists on cognitive performance in aged monkeys. Neurobiol Aging. 1997;18:21–28. doi: 10.1016/s0197-4580(96)00162-5. [DOI] [PubMed] [Google Scholar]
  6. Arrang JM, Garbarg M, Schwartz JC. Auto-inhibition of brain histamine release mediated by a novel class (H3) of histamine receptor. Nature. 1983;302:832–837. doi: 10.1038/302832a0. [DOI] [PubMed] [Google Scholar]
  7. Arrang JM, Garbarg M, Schwartz JC. Autoinhibition of histamine synthesis mediated by presynaptic H3-receptors. Neuroscience. 1987;23:149–157. doi: 10.1016/0306-4522(87)90279-x. [DOI] [PubMed] [Google Scholar]
  8. Auld DS, Kornecook TJ, Bastianetto S, Quirion R. Alzheimer's disease and the basal forebrain cholinergic system: relations to beta-amyloid peptides, cognition, and treatment strategies. Prog Neurobiol. 2002;68:209–245. doi: 10.1016/s0301-0082(02)00079-5. [DOI] [PubMed] [Google Scholar]
  9. Bach ME, Barad M, Son H, Zhuo M, Lu YF, Shih R, et al. Age-related deficits in spatial memory are correlated with deficits in the late phase of hippocampal long-term potentiationin vitro and are attenuated by drugs that enhance the cAMP signaling pathway. Proc Natl Acad Sci USA. 1999;96:5280–5285. doi: 10.1073/pnas.96.9.5280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Bachurin S, Bukatina E, Lermontova N, Tkachenko S, Afanasiev A, Grigoriev V, et al. Antihistamine agent Dimebon as a novel neuroprotector and a cognition enhancer. Ann N Y Acad Sci. 2001;939:425–435. doi: 10.1111/j.1749-6632.2001.tb03654.x. [DOI] [PubMed] [Google Scholar]
  11. Ballard C, Hanney ML, Theodoulou M, Douglas S, McShane R, Kossakowski K, et al. The dementia antipsychotic withdrawal trial (DART-AD): long-term follow-up of a randomised placebo-controlled trial. Lancet Neurol. 2009 Feb;8(2):151–157. doi: 10.1016/S1474-4422(08)70295-3. [DOI] [PubMed] [Google Scholar]
  12. Ballard J. Forgetfulness and older adults: concept analysis. J Adv Nurs. 2010 Jun;66(6):1409–1419. doi: 10.1111/j.1365-2648.2010.05279.x. [DOI] [PubMed] [Google Scholar]
  13. Ballard TM, McAllister KH. The acetylcholinesterase inhibitor, ENA 713 (Exelon), attenuates the working memory impairment induced by scopolamine in an operant DNMTP task in rats. Psychopharmacology (Berl) 1999;146:10–18. doi: 10.1007/s002130051082. [DOI] [PubMed] [Google Scholar]
  14. Barad M, Bourtchouladze R, Winder DG, Golan H, Kandel E. Rolipram, a type IV-specific phosphodiesterase inhibitor, facilitates the establishment of long-lasting long-term potentiation and improves memory. Proc Natl Acad Sci USA. 1998;95:15020–15025. doi: 10.1073/pnas.95.25.15020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Bar-Am O, Weinreb O, Amit T, Youdim MB. The novel cholinesterase-monoamine oxidase inhibitor and antioxidant, ladostigil, confers neuroprotection in neuroblastoma cells and aged rats. J Mol Neurosci. 2009;37:135–145. doi: 10.1007/s12031-008-9139-6. [DOI] [PubMed] [Google Scholar]
  16. Barbier AJ, Berridge C, Dugovic C, Laposky AD, Wison SJ, Boggs J, et al. Acute wake-promoting actions of JNJ-5207852, a novel, diamine-based H3 antagonist. Brit J Pharmacol. 2004;143:649–661. doi: 10.1038/sj.bjp.0705964. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Barnes JM, Costall B, Coughlan J, Domeney AM, Gerrard PA, Kelly ME, et al. The effects of ondansetron, a 5-HT3 receptor antagonist, on cognition in rodents and primates. Pharmacol Biochem Behav. 1990;35:955–962. doi: 10.1016/0091-3057(90)90385-u. [DOI] [PubMed] [Google Scholar]
  18. Barnes NM, Sharp T. A review of central 5-HT receptors and their function. Neuropharmacology. 1999;38:1083–1152. doi: 10.1016/s0028-3908(99)00010-6. [DOI] [PubMed] [Google Scholar]
  19. Bartolomeo AC, Morris H, Buccafusco JJ, Kille N, Rosenzweig-Lipson S, Husbands MG, et al. The preclinical pharmacological profile of WAY-132983, a potent M1 preferring agonist. J Pharmacol Exp Ther. 2000;292(2):584–596. [PubMed] [Google Scholar]
  20. Bartus RT, Dean RL, 3rd, Beer B, Lippa AS. The cholinergic hypothesis of geriatric memory dysfunction. Science. 1983;217(4558):408–414. doi: 10.1126/science.7046051. [DOI] [PubMed] [Google Scholar]
  21. Bartus RT. On neurodegenerative diseases, models, and treatment strategies: lessons learned and lessons forgotten a generation following the cholinergic hypothesis. Exp Neurol. 2000 Jun;163(2):495–529. doi: 10.1006/exnr.2000.7397. [DOI] [PubMed] [Google Scholar]
  22. Bate C, Salmona M, Williams A. The role of platelet activating factor in prion and amyloid-b neurotoxicity. Neuroreport. 2004;15:509–513. doi: 10.1097/00001756-200403010-00025. [DOI] [PubMed] [Google Scholar]
  23. Bazan NG, Colangelo V, Lukiw WJ. Prostaglandins and other lipid mediators in Alzheimer’s disease. Prostaglandins & Other Lipid Mediators. 2002:68–69. 197–210. doi: 10.1016/s0090-6980(02)00031-x. [DOI] [PubMed] [Google Scholar]
  24. Beach T, Walker D, Potter L, Sue L, Scott S, Layne K, et al. Immunotoxin lesion of the cholinergic nucleus basalis causes Abeta deposition: towards a physiologic animal model of Alzheimer’s disease. Curr Med Chem. 2003;3:233–243. [Google Scholar]
  25. Bender AT, Beavo JA. Cyclic nucleotide phosphodiesterases: molecular regulation to clinical use. Pharmacol Rev. 2006;58:488–520. doi: 10.1124/pr.58.3.5. [DOI] [PubMed] [Google Scholar]
  26. Benowitz NL. Clinical pharmacology of nicotine. Ann Rev Med. 1986;37:21–32. doi: 10.1146/annurev.me.37.020186.000321. [DOI] [PubMed] [Google Scholar]
  27. Bentley J, Sleight AJ, Marsden CA. Fone KCF. 5-HT6 antisense oligonucleotide i.c.v. affects rat performance in the water maze and feeding. J Psychopharmacol. 1997;11:A64. [Google Scholar]
  28. Benzi G, Moretti A. Is there a rationale for the use of acetylcholinesterase inhibitors in the therapy of Alzheimer's disease? Eur J Pharmacol. 1998;346(1):1–13. doi: 10.1016/s0014-2999(98)00093-4. [DOI] [PubMed] [Google Scholar]
  29. Bertrand F, Lehmann O, Galani R, Lazarus C, Jeltsch H, Cassel JC. Effects of MDL 73005 on water-maze performances and locomotor activity in scopolamine-treated rats. Pharmacol Biochem Behav. 2001;68:647–660. doi: 10.1016/s0091-3057(01)00448-8. [DOI] [PubMed] [Google Scholar]
  30. Bliss TV, Collingridge GL. A synaptic model of memory: long-term potentiation in the hippocampus. Nature. 1993;361:31–39. doi: 10.1038/361031a0. [DOI] [PubMed] [Google Scholar]
  31. Boada-Rovira M, Brodaty H, Cras P, Baloyannis S, Emre M, Zhang R, et al. Efficacy and safety of donepezil in patients with Alzheimer's disease: results of a global, multinational, clinical experience study. Drugs Aging. 2004;21(1):43–53. doi: 10.2165/00002512-200421010-00004. [DOI] [PubMed] [Google Scholar]
  32. Bockaert J, Claeysen S, Compan V, Dumuis A. 5-HT4 receptors. Curr Drug Targets CNS Neurol Diord. 2004;3:39–51. doi: 10.2174/1568007043482615. [DOI] [PubMed] [Google Scholar]
  33. Boess FG, Hendrix M, van der Staay FJ, Erb C, Schreiber R, van Staveren W, et al. Inhibition of Phosphodiesterase 2 increases neuronal cGMP, synaptic plasticity and memory performance. Neuropharmacology. 2004;47:1081–1092. doi: 10.1016/j.neuropharm.2004.07.040. [DOI] [PubMed] [Google Scholar]
  34. Bonaventure P, Hall H, Gommeren W, Cras P, Langlois X, Jurzak M, et al. Mapping of serotonin 5-HT4 receptor mRNA and ligand binding sites in the post-mortem human brain. Synapse. 2000;36:35–46. doi: 10.1002/(SICI)1098-2396(200004)36:1<35::AID-SYN4>3.0.CO;2-Y. [DOI] [PubMed] [Google Scholar]
  35. Bonaventure P, Letavic M, Dugovic C, Wilson S, Aluisio L, Pudiak C, et al. Histamine H3 receptor antagonist: from target identification to drug leads. Biochem Pharmacol. 2007;73:1084–1096. doi: 10.1016/j.bcp.2006.10.031. [DOI] [PubMed] [Google Scholar]
  36. Bos M, Sleight AJ, Godel T, Martin JR, Reimer C. Stadler H, 5-HT6 receptor antagonists: lead optimization and biological evaluation of N-aryl and N-heteroaryl 4-amino-benzene sulfonmides. Eur J Med Chem. 2001;36:165–178. doi: 10.1016/s0223-5234(00)01209-5. [DOI] [PubMed] [Google Scholar]
  37. Bourtchouladze R, Lidge R, Catapano R, Stanley J, Gossweiler S, Romashko D, et al. A mouse model of Rubinstein-Taybi syndrome: defective long-term memory is ameliorated by inhibitors of Phosphodiesterase 4. Proc Natl Acad Sci USA. 2003;100:10518–10522. doi: 10.1073/pnas.1834280100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Bradley SR, Lameh J, Ohrmund L, Thomas S, Bajpai A, Nguyen D, et al. AC-260584, an orally bioavailable M1 muscarinic receptor allosteric agonist, improves cognitive performance in an animal model. Neuropharm. 2010;58:365–373. doi: 10.1016/j.neuropharm.2009.10.003. [DOI] [PubMed] [Google Scholar]
  39. Brandeis R, Sapir M, Hafif N, Abraham S, Oz N, Stein E, et al. AF150(S): a new functionally selective M1 agonist improves cognitive performance in rats. Pharmacol Biochem Behav. 1999;51(4):667–674. doi: 10.1016/0091-3057(94)00435-l. [DOI] [PubMed] [Google Scholar]
  40. Bratt AM, Kelly ME, Domeney AM, Naylor RJ, Costall B. Ondansetron fails to attenuate a scopolamine-induced deficit in a stone maze task. Neuroreport. 1994;5:1921–1924. doi: 10.1097/00001756-199410000-00020. [DOI] [PubMed] [Google Scholar]
  41. Briggs CA, Anderson DJ, Brioni JD, Buccafusco JJ, Buckley MJ, Campell JE, et al. Functional characterization of the novel neuronal nicotinic receptor ligand GTS-21 in vitro and in vivo. Pharmacol Biochem Behav. 1997;57:231–241. doi: 10.1016/s0091-3057(96)00354-1. [DOI] [PubMed] [Google Scholar]
  42. Brioni JD, Kim DJ, O’Neill AB. Nicotine cue: lack of effect of the alpha 7 nicotinic receptor antagonist methyllycaconitine. Eur J Pharmacol. 1996;301:1–5. doi: 10.1016/0014-2999(96)00010-6. [DOI] [PubMed] [Google Scholar]
  43. Browman KE, Komater VA, Curzon P, Rueter LE, Hancock AA, Decker MW, et al. Enhancement of prepulse inhibition of startle in mice by the H3 receptor antagonists thioperamide and ciproxifan. Behav Brain Res. 2004;153:69–76. doi: 10.1016/j.bbr.2003.11.001. [DOI] [PubMed] [Google Scholar]
  44. Buccafusco JJ. Neuronal nicotinic receptor subtypes: defining therapeutic targets. Mol Interv. 2004;4(5):285–295. doi: 10.1124/mi.4.5.8. [DOI] [PubMed] [Google Scholar]
  45. Buccafusco JJ, Terry AV, Jr, Decker MW, Gopalakrishnan M. Profile of nicotinic acetylcholine receptor agonists ABT-594 and A-582941, with differential subtype selectivity, on delayed matching accuracy by young monkeys. Biochem Pharmacol. 2007;74:1202–1211. doi: 10.1016/j.bcp.2007.07.010. [DOI] [PubMed] [Google Scholar]
  46. Buccafusco JJ, Terry AV., Jr A reversible model of the cognitive impairment associated with schizophrenia in monkeys: potential therapeutic effects of two nicotinic acetylcholine receptor agonists. Biochem Pharmacol. 2009;78(7):852–862. doi: 10.1016/j.bcp.2009.06.102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Buccafusco JJ, Terry AV, Jr, Goren T, Blaugrun E. Potential cognitive actions of (n-propargyl-(3r)-aminoindan-5-yl)-ethyl, methyl carbamate (tv3326), a novel neuroprotective agent, as assessed in old rhesus monkeys in their performance of versions of a delayed matching task. Neuroscience. 2003;119:669–678. doi: 10.1016/s0306-4522(02)00937-5. [DOI] [PubMed] [Google Scholar]
  48. Buccafusco JJ, Beach JW, Terry AV., Jr Desensitization of nicotinic acetylcholine receptors as a strategy for drug development. J Pharmacol Exp Ther. 2009 Feb;328(2):364–370. doi: 10.1124/jpet.108.145292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Buccafusco JJ. Emerging cognitive enhancing drugs. Expert Opin Emerg Drugs. 2009 Dec;14(4):577–589. doi: 10.1517/14728210903257796. [DOI] [PubMed] [Google Scholar]
  50. Burnham KE, Baxter MG, Bainton JR, Southam E, Dawson LA, Bannerman DM, et al. Activation of 5-HT6 receptors facilitates attentional set shifting. Psychopharmacology. 2010;208:13–21. doi: 10.1007/s00213-009-1701-6. [DOI] [PubMed] [Google Scholar]
  51. Caccamo A, Oddo S, Billings LM, Green KN, Martinez-Coria H, Fisher A, et al. M1 receptors play a central role in modulating AD-like pathology in transgenic mice. Neuron. 2006;49:671–682. doi: 10.1016/j.neuron.2006.01.020. [DOI] [PubMed] [Google Scholar]
  52. Cachard-Chastel M, Lezoualc’h F, Dewachter I, Delomenie C, Croes S, Devijver H, et al. 5-HT4 receptor agonists increase sAPPα levels in the cortex and hippocampus of male C57BL/6j mice. Brit J Pharmacol. 2007;150:883–892. doi: 10.1038/sj.bjp.0707178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Cachard-Chastel M, Devers S, Sicsic S, Langlois M, Lezoualc’h F, Gardier AM, et al. Prucalopride and donepezil act synergistically to reverse scopolamine-induced memory deficit in C57BL/6j mice. Behav Brain Res. 2008;187:455–461. doi: 10.1016/j.bbr.2007.10.008. [DOI] [PubMed] [Google Scholar]
  54. Callahan PM, Ilch CP, Rowe WB, Tehim A. Charcterization of the selective 5-HT6 receptor antagonist SB-271046 in behavioral models of cognition. Soc Neurosi Abstr. 2004 776.19. [Google Scholar]
  55. Callahan PM, Wang S, Xie W, Dragan S, Sun S, Michael T, Herbert B, Rowe WB, Tehim A, Lowe DA, Barrett JE. Pharmacological characterization of MEM 3454, a novel nicotinic alpha7 receptor partial agonist: Therapeutic potential for the cognitive deficits associated with Alzheimer’s disease and schizophrenia. Neuropsychopharmacology. 2006;31 Supl 1:S198. [Google Scholar]
  56. Callahan PM, Rowe WB, Hsu CC, Tehim A. Characterization of the 5-HT6 receptor agonist, WAY 181187 and antagonist, SB 742457: effects on recognition, associative and spatial reference memory function in young animals. Soc Neurosci Abstr. 2009 883.3. [Google Scholar]
  57. Carey GJ, Costall B, Domeney AM, Gerrard PA, Jones DN, Naylor RJ, et al. Ondansetron and arecoline prevent scopolamine-induced cognitive deficits in the marmoset. Pharmacol Biochem Behav. 1992;42:75–83. doi: 10.1016/0091-3057(92)90449-p. [DOI] [PubMed] [Google Scholar]
  58. Carey GJ, Billard W, Binch H, 3rd, Cohen-Williams M, Crosby G, Grzelak M, Guzik H, Kozlowski JA, Lowe DB, Pond AJ, Tedesco RP, Watkins RW, Coffin VL. SCH 57790, a selective muscarinic M2 receptor antagonist, releases acetylcholine and produces cognitive enhancement in laboratory animals. Eur. J. Pharmacol. 2001;16:189–200. doi: 10.1016/s0014-2999(01)01440-6. [DOI] [PubMed] [Google Scholar]
  59. Carli M, Lushi R, Sammanin R. Dose related impairments of spatial learning by intrahippocampal scopolamine and antagonism by ondansetron, a 5-HT3 receptor antagonist. Behav Brain Res. 1997;82:185–194. doi: 10.1016/s0166-4328(97)80988-6. [DOI] [PubMed] [Google Scholar]
  60. Carlsson CM. Lessons learned from failed and discontinued clinical trials for the treatment of Alzheimer' disease: future directions. J Alzheimers Dis. 2008;15:327–338. doi: 10.3233/jad-2008-15214. [DOI] [PubMed] [Google Scholar]
  61. Cassel JC, Jeltsch H. Serotonergic modulation of cholinergic function in the central nervous system: cognitive implications. Neuroscience. 1995;69:1–41. doi: 10.1016/0306-4522(95)00241-a. [DOI] [PubMed] [Google Scholar]
  62. Cavalli A, Bolognesi ML, Capsoni S, Andrisano V, Bartolini M, Margotti E, et al. A small molecule targeting the multifactorial nature of Alzheimer's disease. Angew Chem Int Ed Engl. 2007;46(20):3689–3692. doi: 10.1002/anie.200700256. [DOI] [PubMed] [Google Scholar]
  63. Centers for Disease Control and Prevention and The Merck Company Foundation. The State of Aging and Health in America, 2007. Whitehouse Station, NJ: The Merck Company Foundation; 2007. [Google Scholar]
  64. Chen L, Wang H, Zhang Z, Li Z, He D, Sokabe M, et al. DMXB (GTS-21) ameliorates the cognitive deficits in beta amyloid(25–35(−)) injected mice through preventing the dysfunction of alpha7 nicotinic receptor. J Neurosci Res. 2010;88(8):1784–1794. doi: 10.1002/jnr.22345. [DOI] [PubMed] [Google Scholar]
  65. Chen Z. Effect of histamine H3-receptor antagonist clobenpropit on spatial memory of radial maze performance in rats. Acta Pharmacol Sin. 2000;21:905–910. [PubMed] [Google Scholar]
  66. Chen Z, Xu AJ, Li R, Wei EQ. Reversal of scopolamine-induced spatial memory deficits in rats by TAK-147. Acta Pharmacol Sin. 2002 Apr;23(4):355–360. [PubMed] [Google Scholar]
  67. Clader JW, Wang Y. Muscarinic receptor agonists and antagonists in the treatment of Alzheimer’s disease. Curr Pharmaceutical Des. 2005;11:3353–3361. doi: 10.2174/138161205774370762. [DOI] [PubMed] [Google Scholar]
  68. Clapham J, Kilpatrick GJ. Thioperamide, the selective histamine H3 receptor antagonist, attenuates stimulant-induced locomotor activity in the mouse. Eur J Pharmacol. 1994;259:107–114. doi: 10.1016/0014-2999(94)90498-7. [DOI] [PubMed] [Google Scholar]
  69. Cole DC, Lennox WJ, Lombardi S, Ellingboe JW, Bernotas RC, Tawa GJ, et al. Discovery of 5-arlsulfonamido-3(pyrrolidin-2-ylmethyl)-1H-indole derivatives as potent, selective 5-HT6 receptor agonists and antagonists. J Med Chem. 2005;48:353–356. doi: 10.1021/jm049243i. [DOI] [PubMed] [Google Scholar]
  70. Comery TA, Martone RL, Aschmies S, Atchison KP, Diamantidis G, Gong X, et al. Acute γ-secretase inhibition improves contextual fear conditioning in the Tg2576 mouse model of Alzheimer’s disease. J Neurosci. 2005;25:8898–8902. doi: 10.1523/JNEUROSCI.2693-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Conn PJ, Jones CK, Lindsley CW. Subtype-selective allosteric modulators of muscarinic receptors for the treatment of CNS disorders. Trends Pharmacol Sci. 2009;30:148–155. doi: 10.1016/j.tips.2008.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Consolo S, Amaboldi S, Giorgi S, Russi G, Ladinsky H. 5-HT4 receptor stimulation facilitates acetylcholine release in rat frontal cortex. Neuroreport. 2004;5:1230–1232. doi: 10.1097/00001756-199406020-00018. [DOI] [PubMed] [Google Scholar]
  73. Coyle JT, Price DL, DeLong MR. Alzheimer's disease: a disorder of cortical cholinergic innervation. Science. 1983;219(4589):1184–1190. doi: 10.1126/science.6338589. [DOI] [PubMed] [Google Scholar]
  74. Coyle JT, Geerts H, Sorra K, Amatniek J. Beyond in vitro data: a review of in vivo evidence regarding the allosteric potentiating effect of galantamine on nicotinic acetylcholine receptors in Alzheimer’s neuropathology. J Alzheimers Dis. 2007;11:491–507. doi: 10.3233/jad-2007-11411. [DOI] [PubMed] [Google Scholar]
  75. Cui YH, Si W, Yin L, An SM, Jin J, Deng SN, et al. A novel derivative of xanomeline improved memory function in aged mice. Neurosci Bull. 2008a;24(4):251–257. doi: 10.1007/s12264-008-0204-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Cui Y, Wang D, Si W, Lv W, Niu Y, Lei X, et al. Enhancement of memory function in aged mice by a novel derivative of xanomeline. Cell Res. 2008b;18:1151–1153. doi: 10.1038/cr.2008.292. [DOI] [PubMed] [Google Scholar]
  77. Davies P, Maloney AJ. Selective loss of central cholinergic neurons in Alzheimer's disease. Lancet. 1976;2(8000):1403. doi: 10.1016/s0140-6736(76)91936-x. [DOI] [PubMed] [Google Scholar]
  78. Dawson GR, Bentley G, Draper F, Rycroft W, Iversen SD, Pagella PG. The behavioral effects of heptyl physostigmine, a new cholinesterase inhibitor, in tests of long-term and working memory in rodents. Pharmacol Biochem Behav. 1991;39(4):865–871. doi: 10.1016/0091-3057(91)90045-4. [DOI] [PubMed] [Google Scholar]
  79. Dawson LA, Nguyen HQ, Li P. In vivo effects of the 5-HT(6)antagonist SB-271046 on striatal and frontal cortex extracellular concentrations of noradrenaline, dopamine, 5-HT, glutamate and aspartate. Br J Pharacol. 2000;130:23–26. doi: 10.1038/sj.bjp.0703288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Dawson LA, Nguyen HQ, Li P. The 5-HT(6) receptor antagonist SB-271046 selectively enhances excitatory neurotransmission in the rat frontal cortex and hippocampus. Neuropsychopharmacology. 2001;25:662–668. doi: 10.1016/S0893-133X(01)00265-2. [DOI] [PubMed] [Google Scholar]
  81. Day M, Pan JB, Buckley MJ, Cronin E, Hollingsworth PR, Hirst WD, et al. Differential effects of ciproxifan and nicotine on impulsivity and attention measures in the 5-choice serial reaction time test. Biochem Pharmacol. 2007;73:1123–1134. doi: 10.1016/j.bcp.2006.12.004. [DOI] [PubMed] [Google Scholar]
  82. Decker MW, Bannon AW, Curzon P, Gunther KL, Brioni JD, Holladay MW, et al. ABT-089 [2-methyl-3-(2-(S)-pyrrolidinylmethoxy)pyridine dihydrochloride]: II. A novel cholinergic channel modulator with effects on cognitive performance in rats and monkeys. J Pharmacol Exp Ther. 1997;283:247–258. [PubMed] [Google Scholar]
  83. De Ferrari GV, Canales MA, Shin I, Weiner LM, Silman I, Inestrosa NC. A structural motif of acetylcholinesterase that promotes amyloid beta-peptide fibril formation. Biochemistry. 2001;40:10447–10457. doi: 10.1021/bi0101392. [DOI] [PubMed] [Google Scholar]
  84. de Filippi G, Mogg AJ, Phillips KG, Zwart R, Sher E, Chen Y. The subtype-selective nicotinic acetylcholine receptor positive allosteric modulator 2087101 facilitates neurotransmission in the brain. Eur J Pharmacol. 2010;643(2–3):218–224. doi: 10.1016/j.ejphar.2010.06.064. [DOI] [PubMed] [Google Scholar]
  85. de Los Ríos C, Egea J, Marco-Contelles J, León R, Samadi A, Iriepa I, et al. Synthesis, inhibitory activity of cholinesterases, and neuroprotective profile of novel 1,8-naphthyridine derivatives. J Med Chem. 2010;53(14):5129–5143. doi: 10.1021/jm901902w. [DOI] [PubMed] [Google Scholar]
  86. Devan BD, Sierra-Mercado D, Jr, Jimenez M, Bowker JL, Duffy KB, Spangler EL, et al. Phosphodiesterase inhibition by sidenafil citrate attenuates the learning impairment induced by blockade of cholinergic muscarinic receptors in rats. Pharmacol Bichem Behav. 2004;79:691–699. doi: 10.1016/j.pbb.2004.09.019. [DOI] [PubMed] [Google Scholar]
  87. Devan BD, Bowker JL, Duffy KB, Bharati IS, Jimenez M, Sierra-Mercado D, Jr, et al. Phosphodiesterase inhibition by sidenafil citrate attenuates a maze learning impairment in rats induced by nitric oxide synthase inhibition. Psychopharmacology. 2006;183:439–445. doi: 10.1007/s00213-005-0232-z. [DOI] [PubMed] [Google Scholar]
  88. Diez-Ariza M, Garcia-Alloza M, Lasheras B, Del Rio J, Ramirez MJ. GABA(A) receptor antagonists ehance cortical acetylcholine release induced by 5-HT(3) receptor blockade in freely moving rats. Brain Res. 2002;956:81–85. doi: 10.1016/s0006-8993(02)03483-2. [DOI] [PubMed] [Google Scholar]
  89. Doody RS, Gavrilova SI, Sano M, Thomas RG, Aisen PS, Bachurin SO, et al. Effect of dimebon on cognition, activities of daily living, behaviour, and global function in patients with mild-to-moderate Alzheimer's disease: a randomised, double-blind, placebo-controlled study. Lancet. 2006;372(9634):207–215. doi: 10.1016/S0140-6736(08)61074-0. [DOI] [PubMed] [Google Scholar]
  90. Domek-Lopacinska K, Strosznajder JB. The effect of selective inhibiton of cyclic GMP hydrolyzing phosphodiesterase 2 and 5 on learning and memory processes and nitric oxide synthase activity in brain during aging. Brain Res. 2008;1216:68–77. doi: 10.1016/j.brainres.2008.02.108. [DOI] [PubMed] [Google Scholar]
  91. Domeney AM, Costall B, Gerrard PA, Jones DN, Naylor RJ, Tyers MB. The effect of ondansetron on cognitive performance in the marmoset. Pharmacol Biochem Behav. 1991;38:169–175. doi: 10.1016/0091-3057(91)90606-3. [DOI] [PubMed] [Google Scholar]
  92. Dysken M, Kuskowski M, Love S. Ondansetron in the treatment of cognitive decline in Alzheimer’s dementia. Am J Geriatr Psychiatry. 2002;10:212–215. [PubMed] [Google Scholar]
  93. Egawa T, Mishima K, Matsumoto Y, Iwasaki K, Fujiwara M. Rolipram and its optical isomers, Phosphodiesterase 4 inhibitors, attenuated the scopolamine-induced impairments of learning and memory in rats. Jpn J Pharmacol. 1997;75:275–281. doi: 10.1254/jjp.75.275. [DOI] [PubMed] [Google Scholar]
  94. Elrod K, Buccafusco JJ, Jackson WJ. Nicotine enhances delayed matching-to-sample performance by primates. Life Sci. 1988;43:277–287. doi: 10.1016/0024-3205(88)90318-9. [DOI] [PubMed] [Google Scholar]
  95. Eglen RM, Wong EHF, Dumuis A, Bockaert J. Central 5-HT4 receptors. Trends Pharmacol Sci. 1995;16:391–398. doi: 10.1016/s0165-6147(00)89081-1. [DOI] [PubMed] [Google Scholar]
  96. Esbenshade TA, Browman KE, Bitner RS, Strakhova M, Cowart MD, Brioni JD. The histamine H3 receptor: an attractive target for the treatment of cognitive disorders. Brit J Pharmacol. 2008;154:1166–1181. doi: 10.1038/bjp.2008.147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Fernández-Bachiller MI, Pérez C, González-Muñoz GC, Conde S, López MG, Villarroya M, et al. Novel tacrine-8-hydroxyquinoline hybrids as multifunctional agents for the treatment of Alzheimer's disease, with neuroprotective, cholinergic, antioxidant, and copper-complexing properties. J Med Chem. 2010;53(13):4927–4937. doi: 10.1021/jm100329q. [DOI] [PubMed] [Google Scholar]
  98. Fisher A, Karton Y, Heldman E, Gurwitz D, Haring R, Meshulam H, et al. Progress in medicinal chemistry of novel selective muscarinic agonists. Drug Des Discov. 1993;9(3–4):221–235. [PubMed] [Google Scholar]
  99. Fisher A, Brandeis R, Chapman S, Pittel Z, Michaelson DM. M1 muscarinic agonist treatment reverses cognitive and cholinergic impairments of apolipoprotein E-deficient mice. J Neurochem. 1998;70:1991–1997. doi: 10.1046/j.1471-4159.1998.70051991.x. [DOI] [PubMed] [Google Scholar]
  100. Fisher A, Brandeis R, Haring Nira Bar-Ner R, Kliger-Spatz M, Natan N, Sonego H, et al. AF150(S) and AF267B: M1 muscarinic agonists as innovative therapies for Alzheimer’s disease. J Mol Neurosci. 2002;19(1–2):145–153. doi: 10.1007/s12031-002-0025-3. [DOI] [PubMed] [Google Scholar]
  101. Flynn DD, Ferrari-DiLeo G, Levey AI, Mash DC. Differential alterations in muscarinic receptor subtypes in Alzheimer’s disease: implications for cholinergic-based therapies. Life Sci. 1995;56:869–876. doi: 10.1016/0024-3205(95)00022-x. [DOI] [PubMed] [Google Scholar]
  102. Foley AG, Murphy KJ, Hirst WD, Gallagher HC, Hagan JJ, Upton N, et al. The 5-HT6 receptor antagonist SB-271046 reverses scopolamine-disrupted consolidation of a passive avoidance task and ameliorates spatial task deficits in aged rats. Neuropsychopharmacology. 2004;29:58–67. doi: 10.1038/sj.npp.1300332. [DOI] [PubMed] [Google Scholar]
  103. Fone KCF. An update on the role of the 5-hydroxytryptamine6 receptor in cognitive function. Neuropharmacology. 2008;55:1015–1022. doi: 10.1016/j.neuropharm.2008.06.061. [DOI] [PubMed] [Google Scholar]
  104. Fontana DJ, Daniels SE, Henderson C, Eglen RM, Wong EH. Ondansetron improves cognitive performance in the Morris water maze spatial navigation task. Psychopharmacology. 1995;120:408–417. doi: 10.1007/BF02245812. [DOI] [PubMed] [Google Scholar]
  105. Fontana DJ, Daniels SE, Wong EH, Clark RD, Eglen RM. The effects of novel, selective 5-hydroxytryptamine (5-HT)4 receptor ligands in rat spatial navigation. Neuropharmacology. 1997;36:689–696. doi: 10.1016/s0028-3908(97)00055-5. [DOI] [PubMed] [Google Scholar]
  106. Fox GB, Pan JB, Radek RJ, Lewis AM, Bitner RS, Esbenshade TA, et al. Two novel and selective nonimidazole H3 receptor antagonists A-304121 and A-317920: II. In vivo behavioral and neurophysiological characterization. J Pharmacol Exp Ther. 2003;305:897–908. doi: 10.1124/jpet.102.047241. [DOI] [PubMed] [Google Scholar]
  107. Fox GB, Esbenshade TA, Pan JB, Radek RJ, Krueger KM, Yao BB, et al. Pharmacological properties of ABT-239 [4-(2-{2-[(2R)-2-methylpyrrolidinyl]ethyl}-benzofuran-5-yl)benzonitril e]: II. Neurophysiological characterization and broad preclinical efficacy in cognition and schizophrenia of a potent and selective histamine H3 receptor antagonist. J Pharmacol Exp Ther. 2005;313:176–190. doi: 10.1124/jpet.104.078402. [DOI] [PubMed] [Google Scholar]
  108. Freedman R, Hall M, Adler LE, Leonard S. Evidence in post-mortem brain tissue for decreased numbers of hippocampal nicotinic receptors in schizophrenia. Biologic Psychiat. 1995;38:22–33. doi: 10.1016/0006-3223(94)00252-X. [DOI] [PubMed] [Google Scholar]
  109. Frey U, Huang YY, Kandel ER. Effects of cAMP simulate a late stage pf LTP in hippocampal CA1 neurons. Science. 1993;260:1661–1664. doi: 10.1126/science.8389057. [DOI] [PubMed] [Google Scholar]
  110. Frolich L, Eckerwall G, Jonas N. Sirocco-Investigators. A multicenter, double-blind, placebo-controlled phase IIB proof-of-concept dose-ranging study of AZD3480 and donepezil over 12 weeks in patients with mild to moderate Alzheimer’s disease. Alzheimers Dement. 2009;5(4) suppl 1:P85. [Google Scholar]
  111. Galeotti N, Ghelardini C, Bartolini A. Role of 5HT4 receptors in mouse passive avoidance test. J Pharmacol Exp Ther. 1998;286:1115–1121. [PubMed] [Google Scholar]
  112. Galici R, Boggs JD, Aluisio L, Frazer IC, Bonaventure P, Lord B, et al. JNJ-10181457, a selective non-imidazole histamine H3 receptor antagonist, normalizes acetylcholine neurotransmission and has efficacy in translational rat models of cognition. Neuropharmacology. 2009;56:1131–1137. doi: 10.1016/j.neuropharm.2009.03.011. [DOI] [PubMed] [Google Scholar]
  113. Gallagher M, Rapp PR, Fanelli RJ. Opiate antagonist facilitation of time-dependent memory processes: dependence upon intact norepinephrine function. Brain Res. 1985;347:284–290. doi: 10.1016/0006-8993(85)90188-x. [DOI] [PubMed] [Google Scholar]
  114. Gannon KS, King M, Fone KCF, Shacham S, Fichman M, Melendez R, Cheruku S, Lobera M, Orbach P. PRX-07034, a potent and selective 5-HT6 antagonist, enhances novel object discrimination (NOD) memory and augments the effect of Aricept. Soc Neurosci Abstr. 2006:64.9. [Google Scholar]
  115. Gao Y, Tang XC, Guan LC, Kuang PZ. Huperzine A reverses scopolamine- and uscimol-induced memory deficits in chick. Acta Pharmocol Sin. 2000;21:1169–1173. [PubMed] [Google Scholar]
  116. Gard PR, Rusted JM. Angiotensin and Alzheimer’s disease: therapeutic prospects. Expert Rev Neurotherapeut. 2004;4:87–96. doi: 10.1586/14737175.4.1.87. [DOI] [PubMed] [Google Scholar]
  117. Gatto GJ, Bohme GA, Caldwell WS, Letchworth SR, Traina VM, Obinu MC, et al. TC-1734: an orally active neuronal nicotinic acetylcholine receptor modulator with antidepressant, neuroprotective, and long-lasting cognitive effects. CNS Drug Rev. 2004;10(2):147–166. doi: 10.1111/j.1527-3458.2004.tb00010.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Ghelardini C, Galeotti N, Gualtieri F, Romanelli MN, Bucherelli C, Baldi E, et al. DM235 (sunifiram): a novel nootropic with potential as a cognitive enhancer. Naunyn Schmiedebergs Arch Pharmacol. 2002;365:419–426. doi: 10.1007/s00210-002-0577-3. [DOI] [PubMed] [Google Scholar]
  119. Giovannini MG, Bartolini L, Bacciottini L, Greco L, Blandina P. Effects of histamine H3 receptor agonists and antagonists on cognitive performance and scopolamine-induced amnesia. Behav Brain Res. 1999;104:147–155. doi: 10.1016/s0166-4328(99)00063-7. [DOI] [PubMed] [Google Scholar]
  120. Gong B, Vitolo OV, Trinchese F, Liu S, Shelanski M, Arancio O. Persistent improvement in synaptic and cognitive functions in an Alzheimer mouse model after rolipram treatment. J Clin Invest. 2004;114:1624–1634. doi: 10.1172/JCI22831. [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Grauer SM, Pulito VL, Navarra RL, Kelly MP, Kelley C, Graf R, et al. Phosphodiesterase 10A inhibitor activity in preclinical models of positive, cognitive, and negative symptoms of schizophrenia. J Pharmacol Exper Ther. 2009;331:574–590. doi: 10.1124/jpet.109.155994. [DOI] [PubMed] [Google Scholar]
  122. Grigorev VV, Dranyi OA, Bachurin SO. Comparative study of action mechanisms of dimebon and memantine on AMPA- and NMDA-subtypes glutamate receptors in rat cerebral neurons. Bull Exp Biol Med. 2003;136:474–477. doi: 10.1023/b:bebm.0000017097.75818.14. [DOI] [PubMed] [Google Scholar]
  123. Grigorev VV, Garibova TL, Voronina TA, Litvinova SA, Bachurin SO. Effects of chronic administration of dimebon on behavior and memory of SAMP 10 mice. Eksp Klin Farmakol. 2009;72(1):52–56. [PubMed] [Google Scholar]
  124. Grön G, Brandenburg I, Wunderlich AP, Riepe MW. Inhibition of hippocampal function in mild cognitive impairment: targeting the cholinergic hypothesis. Neurobiol Aging. 2006;(1):78–87. doi: 10.1016/j.neurobiolaging.2004.12.005. [DOI] [PubMed] [Google Scholar]
  125. Guan JS, Haggarty SJ, Giacometti E, Dannenberg JH, Joseph N, Gao J, et al. HDAC2 negatively regulates memory formation and synaptic plasticity. Nature. 2009 May 7;459(7243):55–60. doi: 10.1038/nature07925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Gwee MC, Cheah LS. Actions of cimetidine and ranitidine at some cholinergic sites: implications in toxicology and anesthesia. Life Sci. 1986;39:383–388. doi: 10.1016/0024-3205(86)90516-3. [DOI] [PubMed] [Google Scholar]
  127. Hahnen E, Hauke J, Tränkle C, et al. Histone deacetylase inhibitors: possible implications for neurodegenerative disorders. Expert Opin Investig Drugs. 2008;17:169–184. doi: 10.1517/13543784.17.2.169. [DOI] [PubMed] [Google Scholar]
  128. Hannon J, Hoyer D. Molecular biology of 5-HT receptors. Behav Brain Res. 2008;195:198–213. doi: 10.1016/j.bbr.2008.03.020. [DOI] [PubMed] [Google Scholar]
  129. Harder JA, Maclean CJ, Alder JT, Francis PT, Ridley RM. The 5-HT1A antagonist, WAY-100635, ameliorates the cognitive impairment induced by fornix transaction in the marmoset. Psychopharmacology. 1996;127:245–254. [PubMed] [Google Scholar]
  130. Harder JA, Ridley RM. The 5-HT1A antagonist, WAY 100635, alleviates cognitive impairments induced by dizocilpine (MK-801) in monkeys. Neuropharmacology. 2000;39:547–552. doi: 10.1016/s0028-3908(99)00179-3. [DOI] [PubMed] [Google Scholar]
  131. Hasselmo ME. The role of acetylcholine in learning and memory. Curr Opin Neurobiol. 2006;16(6):710–715. doi: 10.1016/j.conb.2006.09.002. Epub 2006 Sep 29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Hatcher PD, Brown VJ, Tait DS, Bate S, Overend P, Hagan JJ, Jones DNC. 5-HT6 receptor antagonists improve performance in an attentional set shifting task in rats. Psychopharmacology. 2005;181:253–259. doi: 10.1007/s00213-005-2261-z. [DOI] [PubMed] [Google Scholar]
  133. Heal DJ, Smith SL, Fias A, Codony X, Buschmann H. Selective 5-HT6 receptor ligands: progress in the development of a novel pharmacological approach to the treatment of obesity and related metabolic disorders. Pharmacol Ther. 2008;117:207–231. doi: 10.1016/j.pharmthera.2007.08.006. [DOI] [PubMed] [Google Scholar]
  134. Hebb AL, Robertson HA. Denovan-Wright EM. Phosphodiesterase 10A inhibition is associated with locomotor and cognitive deficits and increased anxiety in mice. Eur Neuropsychopharmacol. 2008;18:339–363. doi: 10.1016/j.euroneuro.2007.08.002. [DOI] [PubMed] [Google Scholar]
  135. Hebert LE, Scherr PA, Bienias JL, Bennett DA, Evans DA. “Alzheimer’s disease in the U.S. population: Prevalence estimates using the 2000 census.”. Archives of Neurology. 2003;60:1119–1122. doi: 10.1001/archneur.60.8.1119. [DOI] [PubMed] [Google Scholar]
  136. Hille C, Bate S, Davis J, Gonzalez MI. 5-HT4 receptor agonism in the five-choice serial reaction time task. Behav Brain Res. 2008;195:180–186. doi: 10.1016/j.bbr.2008.08.007. [DOI] [PubMed] [Google Scholar]
  137. Hilt D, Gawryl M, Koenig G. EVP-6124_StudyGroup. EVP-6124: safety, tolerability, and cognitive effects of a novel A7 nicotinic receptor agonist in Alzheimer’s disease patients on stable donepezil or rivastigmine therapy; International Conference on Alzheimer’s disease; Vienna, Austria. July 11–16, 2009. [Google Scholar]
  138. Hirst WD, Stean TO, Rogers DC, Sunter D, Pugh P, Moss SF, et al. SB-399885 is a potent, selective 5-HT6 receptor antagonist with cognitive enhancing properties in aged rat water maze and novel object recognition models. Eur J Pharmacol. 2006;553:109–119. doi: 10.1016/j.ejphar.2006.09.049. [DOI] [PubMed] [Google Scholar]
  139. Hirst WD, Andree TH, Aschmies S, Childers WE, Comery TA, Dawson LA, et al. Correlating efficacy in rodent cognition models with in vivo 5-hydroxytryptamine1A receptor occupancy by a novel antagonist, (R)-N-(2-methyl-(4-indolyl-1-piperazinyl)ethyl)-N-(2-pyridinyl)-cycloh exane carboxamide (WAY-101405) J Pharmacol Exp Ther. 2008;325:134–145. doi: 10.1124/jpet.107.133082. [DOI] [PubMed] [Google Scholar]
  140. Hodges H, Sowinski P, Sinden JD, Netto CA, Fletcher A. The selective 5HT3 antagonist, WAY-100289, enhances spatial memory in rats with ibotenate lesions of the forebrain cholinergic projection system. Psychopharmacology. 1995;117:318–332. doi: 10.1007/BF02246107. [DOI] [PubMed] [Google Scholar]
  141. Howe WM, Ji J, Parikh V, Williams S, Mocaer E, Trocme-Thibierge C, et al. Enhancement of attentional performance by selective stimulation of α4β2* nAChRs: Underlying cholinergic mechanisms. Neuropsychopharm. 2010;35:1391–1401. doi: 10.1038/npp.2010.9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. Huang L, Luo Z, He F, Lu J, Li X. Synthesis and biological evaluation of a new series of berberine derivatives as dual inhibitors of acetylcholinesterase and butyrylcholinesterase. Bioorg Med Chem. 2010;18(12):4475–4484. doi: 10.1016/j.bmc.2010.04.063. [DOI] [PubMed] [Google Scholar]
  143. Huang M, Li Z, Prus AJ, Dai J, Callahan PM, Meltzer HY. The nicotinic alpha7 receptor agonist MEM 3454 increases dopamine and acetylcholine release in rat medial prefrontal cortex and hippocampus alone and in combination with risperidone. Neuropsychopharmacology. 2006;31 Supl. 1:S185. [Google Scholar]
  144. Huang YW, Hu WW, Chen Z, Zhang LS, Shen HQ, Timmerman H, et al. Effect of the histamine H3-antagonist clobenpropit on spatial memory deficits induced by MK-801 as evaluated by radial arm maze in Sprague-Dawley rats. Behav Brain Res. 2004;151:287–293. doi: 10.1016/j.bbr.2003.09.002. [DOI] [PubMed] [Google Scholar]
  145. Huang YY, Kandel ER. Low-frequency stimulation induces a pathway-specific late phase of LTP in the amygdale that is mediated by PKA and dependent on protein synthesis. Learn Mem. 2007;14:497–503. doi: 10.1101/lm.593407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  146. Huang Z, Dias R, Jones T, Liu S, Styhler A, Claveau D, et al. L-454,560, a potent and selective PDE4 inhibitor with in vivo efficacy in animal models of asthma and cognition. Biochem Pharmacol. 2007;73:1971–1981. doi: 10.1016/j.bcp.2007.03.010. [DOI] [PubMed] [Google Scholar]
  147. Hung TM, Na M, Dat NT, Ngoc TM, Youn U, Kim HJ, et al. Cholinesterase inhibitory and anti-amnesic activity of alkaloids from Corydalis turtschaninovii. J Ethnopharmacol. 2008 Sep 2;119(1):74–80. doi: 10.1016/j.jep.2008.05.041. Epub 2008 Jun 13. [DOI] [PubMed] [Google Scholar]
  148. Hulme EC, Lu ZL, Bee MS. Scanning mutagenesis studies of the M1 muscarinic acetylcholine receptor. Receptors Channels. 1990;9:215–228. [PubMed] [Google Scholar]
  149. Hurst RS, Hajos M, Raggenbass M, Wall TM, Higdon NR, Lawson JA, et al. A novel positive allosteric modulator of the alpha7 neuronal nicotinic acetylcholine receptor: in vitro and in vivo characterization. J Neurosci. 2005;25(17):4396–4405. doi: 10.1523/JNEUROSCI.5269-04.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Ikari H, Spangler EL, Greig NH, Pei XF, Brossi A, Speer D, et al. Maze learning in aged rats is enhanced by phenserine, a novel anticholinesterase. Neuroreport. 1995;6(3):481–484. doi: 10.1097/00001756-199502000-00019. [DOI] [PubMed] [Google Scholar]
  151. Imanishi T, Sawa A, Ichimaru Y, Miyashiro M, Kato S, Yamamoto T, et al. Ameliorating effects of rolipram on experimentally induced impairments of learning and memory in rodents. Eur J Pharmacol. 1997;321:272–278. doi: 10.1016/s0014-2999(96)00969-7. [DOI] [PubMed] [Google Scholar]
  152. Imhof L, Wallhagen MI, Mahrer-Imhof R, Monsch AU. Becoming forgetful: how elderly people deal with forgetfulness in everyday life. Am J Alzheimers Dis Other Demen. 2006 Oct–Nov;21(5):347–353. doi: 10.1177/1533317506292499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  153. Impey S, Mark M, Villacres EC, Poser S, Chavkin C, Storm DR. Induction of CRE-mediated gene expression by stimuli that generate long-lasting LTP in area CA1 of the hippocampus. Neuron. 1996;16:973–982. doi: 10.1016/s0896-6273(00)80120-8. [DOI] [PubMed] [Google Scholar]
  154. Janas AM, Cunningham SC, Duffy KB, Devan BD, Greig NH, Holloway HW, et al. The cholinesterase inhibitor, phenserine, improves Morris water maze performance of scopolamine-treated rats. Life Sci. 2005;76(10):1073–1081. doi: 10.1016/j.lfs.2004.06.028. [DOI] [PubMed] [Google Scholar]
  155. Jones CK, Brady AE, Davis AA, Xiang Z, Bubser M, Tantawy MN, et al. Novel selective allosteric activator of the M1 muscarinic acetylcholine receptor regulates amyloid processing and produces antipsychotic-like activity in rats. J Neurosci. 2008;28:10422–10433. doi: 10.1523/JNEUROSCI.1850-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  156. Jones S, Sudweeks S, Yakel JL. Nicotinic receptors in the brain: correlating physiology with function. Trends Neurosci. 1999;22:555–561. doi: 10.1016/s0166-2236(99)01471-x. [DOI] [PubMed] [Google Scholar]
  157. Karlin A. Emerging structure of the nicotinic acetylcholine receptors. Nat Rev Neurosci. 2002;3:102–114. doi: 10.1038/nrn731. [DOI] [PubMed] [Google Scholar]
  158. Kem WR. The brain alpha7 nicotinic receptor may be an important therapeutic target for the treatment of Alzheimer’s disease: studies with DMXBA (GTS-21) Behav Brain Res. 2000;113:169–181. doi: 10.1016/s0166-4328(00)00211-4. [DOI] [PubMed] [Google Scholar]
  159. Kendall I, Slotten HA, Codony X, Burgueno J, Pauwels PJ, Vela JM, et al. E-6801, a 5-HT6 receptor agonist, improves recognition memory by combined modulation of cholinergic and glutamatergic neurotransmission in the rat. Psychopharmacology. 2010 doi: 10.1007/s00213-010-1854-3. (Epub ahead of print) [DOI] [PubMed] [Google Scholar]
  160. Kennedy D. Just treat, or enhance? Science. 2004;304:317. doi: 10.1126/science.304.5667.17. [DOI] [PubMed] [Google Scholar]
  161. Kikuchi T, Okamura T, Fukushi K, Irie T. Piperidine-4-methanthiol ester derivatives for a selective acetylcholinesterase assay. Biol Pharm Bull. 2010;33(4):702–706. doi: 10.1248/bpb.33.702. [DOI] [PubMed] [Google Scholar]
  162. King MV, Sleight AJ, Woolley ML, Topham IA, Marsden CA, Fone KCF. 5-HT6 receptor antagonists reverse delay-dependent deficits in novel object discrimination by enhancing consolidation-an effect sensitive to NMDA receptor antagonism. Neuropharmacology. 2004;47:195–204. doi: 10.1016/j.neuropharm.2004.03.012. [DOI] [PubMed] [Google Scholar]
  163. King MV, Sleight AJ, Fone KCF, Marsden CA. The selective 5-HT6 receptor antagonist Ro)4–6790 reverses age-related and chronic intermittent PCP-induced cognitive dysfunction in rats. J Psychopharmacol. 2005;19:A12. [Google Scholar]
  164. King MV, Marsden CA, Fone KCF. A role for the 5-HT1A, 5-HT4 and 5-HT6 receptors in learning and memory. Trends Pharmacol Sci. 2008;29:482–492. doi: 10.1016/j.tips.2008.07.001. [DOI] [PubMed] [Google Scholar]
  165. King MV, Spicer CH, Sleight AJ, Marsden CA, Fone KCF. Impact of regional 5-HT depletion on the cognitive enhancing effects of a typical 5-ht6 receptor antagonist, Ro 04-6790, in the novel object discrimination task. Psychopharmacology. 2009;202:111–123. doi: 10.1007/s00213-008-1334-1. [DOI] [PubMed] [Google Scholar]
  166. Kinsella K, He W. An Aging World. U.S. Census Bureau, International Population Reports, P95/09–1. Washington, D.C.: U.S. Government Printing Office; 2009
  167. Knapp MJ, Knopman DS, Solomon PR, Pendlebury WW, Davis CS, Gracon SI. A 30-week randomized controlled trial of high-dose tacrine in patients with Alzheimer's disease. The Tacrine Study Group. JAMA. 1994;271(13):985–991. [PubMed] [Google Scholar]
  168. Kohen R, Metcalf MA, Khan N, Druck T, Huebner K, Lachowicz JE, et al. Cloning, characterization, and chromosomal localization of a human 5-HT6 serotonin receptor. J Neurochem. 1996;66:47–56. doi: 10.1046/j.1471-4159.1996.66010047.x. [DOI] [PubMed] [Google Scholar]
  169. Komater VA, Buckley MJ, Browman KE, Pan JB, Hancock AA, Decker MW, et al. Effects of histamine H3 receptor antagonists in two models of spatial learning. Behav Brain Res. 2005;159:295–300. doi: 10.1016/j.bbr.2004.11.008. [DOI] [PubMed] [Google Scholar]
  170. Kulla A, Manahan-Vaughan D. Modulation by serotonin 5-HT(4) receptors of long-term potentiation and depotentiation in the dentate gyrus of freely moving rats. Cereb Cortex. 2002;12:150–162. doi: 10.1093/cercor/12.2.150. [DOI] [PubMed] [Google Scholar]
  171. Kumar YC, Malviya M, Chandra JN, Sadashiva CT, Kumar CS, Prasad SB, et al. Effect of novel N-aryl sulfonamide substituted 3-morpholino arecoline derivatives as muscarinic receptor 1 agonists in Alzheimer’s dementia models. Bioorg Med Chem. 2008;16(9):5157–5163. doi: 10.1016/j.bmc.2008.03.019. [DOI] [PubMed] [Google Scholar]
  172. Kwon SH, Kim HC, Lee SY, Jang CG. Loganin improves learning and memory impairments induced by scopolamine in mice. Eur J Pharmacol. 2009;619(1–3):44–49. doi: 10.1016/j.ejphar.2009.06.062. [DOI] [PubMed] [Google Scholar]
  173. Lachowicz JE, Duffy RA, Ruperto V, Kozlowski J, Zhou G, Clader J, Billard W, Binch H, 3rd, Crosby G, Cohen-Williams M, Strader CD, Coffin V. Facilitation of acetylcholine release and improvement in cognition by a selective M2 muscarinic antagonist, SCH 72788. Life Sci. 2001;68:2585–2592. doi: 10.1016/s0024-3205(01)01056-6. [DOI] [PubMed] [Google Scholar]
  174. Lacroix LP, Dawson LA, Hagan JJ, Heidbreder CA. 5-HT6 receptor antagonist SB-271046 enhances extracellular levels of monoamines in the rat medial prefrontal cortex. Synapse. 2004;51:158–164. doi: 10.1002/syn.10288. [DOI] [PubMed] [Google Scholar]
  175. Lamirault L, Simon H. Enhancement of place and object recognition memory in young adult and old rats by RS 67333, a partial agonist of 5-HT4 receptors. Neuropharmacology. 2001;41:844–853. doi: 10.1016/s0028-3908(01)00123-x. [DOI] [PubMed] [Google Scholar]
  176. Lanni C, Lenzken SC, Pascale A, Del Vecchio I, Racchi M, Pistoia F, et al. Cognition enhancers between treating and doping the mind. Pharmacol Res. 2008 Mar;57(3):196–213. doi: 10.1016/j.phrs.2008.02.004. [DOI] [PubMed] [Google Scholar]
  177. Lee KY, Sung SH, Kim SH, Jang YP, Oh TH, Kim YC. Cognitive-enhancing activity of loganin isolated from Cornus officinalis in scopolamine-induced amnesic mice. Arch Pharm Res. 2009;32(5):677–683. doi: 10.1007/s12272-009-1505-6. [DOI] [PubMed] [Google Scholar]
  178. Lee I, Choe YS, Ryu EK, Choi BW, Choi JY, Choi Y, et al. Synthesis and evaluation of radioiodine-labelled CP-118,954 for the in-vivo imaging of acetylcholinesterase. Nucl Med Commun. 2007;28(7):561–566. doi: 10.1097/MNM.0b013e328194f1f7. [DOI] [PubMed] [Google Scholar]
  179. Leiser SC, Bowlby MR, Comery TA, Dunlop J. A cog in cognition: how the α7 nicotinic acetylcholine receptor is geared towards improving cognitive deficits. Pharmacol & Ther. 2009;122:302–311. doi: 10.1016/j.pharmthera.2009.03.009. [DOI] [PubMed] [Google Scholar]
  180. Lermontova NN, Redkozubov AE, Shevtsova EF, Serkova TP, Kireeva EG, Bachurin SO. Dimebon and tacrine inhibit neurotoxic action of beta-amyloid in culture and block L-type Ca(2+) channels. Bull Exp Biol Med. 2001;132(5):1079–1083. doi: 10.1023/a:1017972709652. [DOI] [PubMed] [Google Scholar]
  181. Leurs R, Bakker RA, Timmerman H, de Esch IJP. The histamine H3 receptor: from gene cloning to H3 receptor drugs. Nature Rev Drug Discov. 2005;4:107–120. doi: 10.1038/nrd1631. [DOI] [PubMed] [Google Scholar]
  182. Levallet G, Hotte M, Boulouard M, Dauphin F. Increased particulate Phosphodiesterase 4 in the prefrontal cortex supports 5-HT4 receptor-induced improvement of object recognition memory in the rat. Psychopharmacology. 2009;202:125–139. doi: 10.1007/s00213-008-1283-8. [DOI] [PubMed] [Google Scholar]
  183. Levey AI, Kitt CA, Simonds WF, Price DL, Brann MR. Identification and localization of muscarinic acetylcholine receptor proteins in brain with subtype-specific antibodies. J Neurosci. 1991;11:3218–3226. doi: 10.1523/JNEUROSCI.11-10-03218.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  184. Levin ED. Nicotinic receptor subtypes and cognitive function. J Neurobiol. 2002;53:633–640. doi: 10.1002/neu.10151. [DOI] [PubMed] [Google Scholar]
  185. Levin ED, Rezvani AH. Nicotinic interactions with antipsychotic drugs, models of schizophrenia and impacts on cognitive function. Biochem Pharmacol. 2007;74:1182–1191. doi: 10.1016/j.bcp.2007.07.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  186. Lezoualc’h F. 5-HT4 receptor and Alzheimer’s disease: the amyloid connection. Exper Neurol. 2007;205:325–329. doi: 10.1016/j.expneurol.2007.02.001. [DOI] [PubMed] [Google Scholar]
  187. Li J, Huang H, Miezan Ezoulin JM, Gao XL, Massicot F, et al. Pharmacological profile of PMS777, a new AChE inhibitor with PAF antagonistic activity. Int J Neuropsychopharmacol. 2006;(1):21–29. doi: 10.1017/S1461145705006425. Epub 2006 Jan 23. [DOI] [PubMed] [Google Scholar]
  188. Li NC, Lee A, Whitmer RA, Kivipelto M, Lawler E, Kazis LE, et al. Use of angiotensin receptor blockers and risk of dementia in a predominantly male population: prospective cohort analysis. BMJ. 2010 Jan 12;340:b5465. doi: 10.1136/bmj.b5465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  189. Li Z, Huang M, Prus AJ, Dai J, Meltzer HY. 5-HT6 receptor antagonist SB-399885 potentiates haloperidol and risperidone-induced dopamine efflux in the medial prefrontal cortex or hippocampus. Brain Res. 2007;1134:70–78. doi: 10.1016/j.brainres.2006.11.060. [DOI] [PubMed] [Google Scholar]
  190. Ligneau X, Lin J-S, Vanni-Mercier G, Jouvet M, Muir JL, Ganellin CR, et al. Neurochemical and behavioral effects of ciproxifan, a potent histamine H3 receptor antagonist. J Pharmacol Exp Ther. 1998;287:658–666. [PubMed] [Google Scholar]
  191. Ligneau X, Perrin D, Landais L, Camelin J-C, Calmels TPG, Berrebi-Bertrand I, et al. BF2.649 [1-{3-[3-(4-chlorophenyl)propoxy]propyl}piperidine, hydrochloride], a nonimidazole inverse agonist/antagonist at the human histamine H3 receptor: preclinical pharmacology. J Pharmacol Exp Ther. 2007;320:365–375. doi: 10.1124/jpet.106.111039. [DOI] [PubMed] [Google Scholar]
  192. Loiseau F, Dekeyne A, Millan MJ. Pro-cognitive effects of 5-HT6 receptor antagonists in the social recognition procedure in rats: implication of the frontal cortex. Psychopharmacology. 2008;196:93–104. doi: 10.1007/s00213-007-0934-5. [DOI] [PubMed] [Google Scholar]
  193. Lopez-Hernandez G, Placzek AN, Thinschmidt JS, Lestage P, Trocme-Thibierge C, Morain P, et al. Partial agonist and neuromodulatory activity of S 24795 for alpha7 nAChR responses of hippocampal interneurons. Neuropharmacology. 2007;53(1):134–144. doi: 10.1016/j.neuropharm.2007.04.007. [DOI] [PubMed] [Google Scholar]
  194. Lowenstein PR, Castro MG. Uncertainty in the translation of preclinical experiments to clinical trials. Why do most phase III clinical trials fail? Curr Gene Ther. 2008;9:368–374. doi: 10.2174/156652309789753392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  195. Lu YF, Hawkins RD. Ryanodine receptors contribute to cGMP-induced late-phase LTP and CREB phosphorylation in the hippocampus. J Neurophysiol. 2002;88:1270–1278. doi: 10.1152/jn.2002.88.3.1270. [DOI] [PubMed] [Google Scholar]
  196. Luck T, Luppa M, Briel S, Riedel-Heller SG. Incidence of mild cognitive impairment: a systematic review. Dement Geriatr Cogn Disord. 2010;29(2):164–175. doi: 10.1159/000272424. [DOI] [PubMed] [Google Scholar]
  197. Luques L, Shoham S, Weinstock M. Chronic brain cytochrome oxidase inhibition selectively alters hippocampal cholinergic innervation and impairs memory: prevention by ladostigil. Exp Neurol. 2007;206(2):209–219. doi: 10.1016/j.expneurol.2007.04.007. Epub 2007 Apr 27. [DOI] [PubMed] [Google Scholar]
  198. Luttgen M, Elvander E, Madjid N, Ogren SO. Analysis of the role of 5-HT1A receptors in spatial and aversive learning in the rat. Neuropharmacology. 2005;48:830–852. doi: 10.1016/j.neuropharm.2005.01.007. [DOI] [PubMed] [Google Scholar]
  199. Lynch G, Palmer LC, Gall CM. The likelihood of cognitive enhancement. Pharmacol Biochem Behavior. 2011;Jan(6) doi: 10.1016/j.pbb.2010.12.024. [Epub ahead of print] [DOI] [PMC free article] [PubMed] [Google Scholar]
  200. Maher-Edwards G, Zvartau-Hind M, Hunter AJ, Gold M, Hopton G, Jacobs G, et al. Double-blind, controlled phase II study of a 5-HT6 receptor antagonist, SB-742457, in Alzheimer’s disease. Curr Alzheimer Res. 2010;7:374–385. doi: 10.2174/156720510791383831. [DOI] [PubMed] [Google Scholar]
  201. Malviya M, Kumar YC, Asha D, Chandra JN, Subhash MN, Rangappa KS. Muscarinic receptor 1 agonist activity of novel N-arylthioureas substituted 3-morpholino arecoline derivatives in Alzheimer’s presenile dementia models. Bioorg Med Chem. 2008;16(15):7095–7101. doi: 10.1016/j.bmc.2008.06.053. [DOI] [PubMed] [Google Scholar]
  202. Malviya M, Sunil Kumar YC, Mythri RB, Venkateshappa C, Subhash MN, Rangappa KS. Muscarinic receptor 1 agonist activity of novel N-aryl carboxamide substituted 3-morpholino arecoline derivatives in Alzheimer’s presenile dementia models. Bioorg Med Chem. 2009;17:5526–5534. doi: 10.1016/j.bmc.2009.06.032. [DOI] [PubMed] [Google Scholar]
  203. Marchetti E, Chaillan FA, Dumuis A, Bockaert J, Soumireu-Mourat B, Roman FS. Modulation of memory processes and cellular excitability in the dentate gyrus of freely moving rats by a 5-HT4 receptor partial agonist, and an antagonist. Neuropharmacology. 2004;47:1021–1035. doi: 10.1016/j.neuropharm.2004.06.033. [DOI] [PubMed] [Google Scholar]
  204. Marcos B, Gil-Bea FJ, Hirst WD, Garcia-Alloza M, Ramirez MJ. Lack of localization of 5-Ht6 receptors on cholinergic neurons: implication of multiple neurotransmitter systems in 5-HT6 receptor-mediated acetylcholine release. Eur J Neurosci. 2006;24:1299–1306. doi: 10.1111/j.1460-9568.2006.05003.x. [DOI] [PubMed] [Google Scholar]
  205. Marcos B, Chuang TT, Gil-Bea FJ, Ramirez MJ. Effects of 5-HT6 receptor antagonism and cholinesterase inhibition in models of cognitive impairment in the rat. Brit J Pharmacol. 2008;155:434–440. doi: 10.1038/bjp.2008.281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  206. Martinez-Mir MI, Pollard H, Moreay J, Arrang JM, Ruat M, Traiffort E, et al. Three histamine receptors (H1, H2 and H3) visualized in the brain of human and non-human primates. Brain Res. 1990;526:322–327. doi: 10.1016/0006-8993(90)91240-h. [DOI] [PubMed] [Google Scholar]
  207. Maruyama W, Akao Y, Youdim MB, Davis BA, Naoi M. Transfection- enforced Bcl-2 overexpression and an anti-Parkinson drug, rasagiline, prevent nuclear accumulation of glyceraldehyde-3-phosphate dehydrogenase induced by an endogenous dopaminergic neurotoxin, N-methyl(R)salsolinol. Journal of Neurochemistry. 2001;78:727–735. doi: 10.1046/j.1471-4159.2001.00448.x. [DOI] [PubMed] [Google Scholar]
  208. Mash DC, Flynn DD, Potter LT. Loss of M2 muscarine receptors in the cerebral cortex in Alzheimer’s disease and experimental cholinergic denervation. Science. 1985;228:1115–1117. doi: 10.1126/science.3992249. [DOI] [PubMed] [Google Scholar]
  209. Matsumoto M, Togashi H, Mori K, Ueno KI, Ohashi S, Kojima T, et al. Evidence for involvement of central 5-HT4 receptors in cholinergic function associated with cognitive processes: behavioral electrophysiological, and neurochemical studies. J Pharmacol Exp Ther. 2001;296:676–682. [PubMed] [Google Scholar]
  210. Mattsson C, Sonesson C, Sandahl A, Greiner HE, Gassen M, Plaschke J, et al. 2-Alkyl-3-(1,2,3,6-tetrahydropyridin-4-yl)-1H-indoles as novel 5-HT6 receptor agonists. Bioorg Med Chem Lett. 2005;15:4230–4234. doi: 10.1016/j.bmcl.2005.06.067. [DOI] [PubMed] [Google Scholar]
  211. McArthur RA, Gray J, Schreiber R. Cognitive effects of muscarinic M1 functional agonists in non-human primates and clinical trials. Curr Opin Investig Drugs. 2010;11:740–760. [PubMed] [Google Scholar]
  212. McGehee DS, Role LW. Physiological diversity of nicotinic acetylcholine receptors expressed by vertebrate neurons. Annu Rev Physiol. 1995;57:521–546. doi: 10.1146/annurev.ph.57.030195.002513. [DOI] [PubMed] [Google Scholar]
  213. Medhurst AD, Atkins AR, Beresford IJ, Brackenborough K, Briggs MA, Calver AR, et al. GSK189254, a novel H3 receptor antagonist that binds to histamine H3 receptors in Alzheimer’s disease brain and improves cognitive performance in preclinical models. J Pharmacol Exp Ther. 2007a;321:1032–1045. doi: 10.1124/jpet.107.120311. [DOI] [PubMed] [Google Scholar]
  214. Medhurst AD, Briggs MA, Bruton G, Calver AR, Chessell I, Crook B, et al. Structurally novel histamine H3 receptor antagonists GSK207040 and GSK334429 improve scopolamine-induced memory impairment and capsaicin-induced secondary allodynia in rats. Biochem Pharmacol. 2007b;73:1182–1194. doi: 10.1016/j.bcp.2007.01.007. [DOI] [PubMed] [Google Scholar]
  215. Meguro K, Yanai K, Sakai N, Sakurai E, Maeyama K, Sasaki H, et al. Effects of thioperamide, a histamine H3 antagonist, on the step-through passive avoidance response and histidine decarboxylase activity in senescence-accelerated mice. Pharmacol Biochem Behav. 1995;50:321–325. doi: 10.1016/0091-3057(95)00248-u. [DOI] [PubMed] [Google Scholar]
  216. Meneses A. 5-HT system and cognition. Neurosci Biobehav Rev. 1999;23:1111–1125. doi: 10.1016/s0149-7634(99)00067-6. [DOI] [PubMed] [Google Scholar]
  217. Meneses A. Role of 5-Ht6 receptors in memory formation. Drug News Perspect. 2001;4:396–400. doi: 10.1358/dnp.2001.14.7.660941. [DOI] [PubMed] [Google Scholar]
  218. Meneses A, Perez-Garcia G. 5-HT(1A) receptors and memory. Neurosci Biobehav Rev. 2007;31:705–727. doi: 10.1016/j.neubiorev.2007.02.001. [DOI] [PubMed] [Google Scholar]
  219. Meneses A, Perez-Garcia G, Liy-Salmeron G, Flores-Galvez D, Castillo C, Castillo E. The effects of the 5-HT6 receptor agonist EMD and the 5-HT7 receptor agonist AS19 on memory formation. Behav Brain Res. 2008;195:112–119. doi: 10.1016/j.bbr.2007.11.023. [DOI] [PubMed] [Google Scholar]
  220. Millan MJ, Gobert A, Roux S, Porsolt R, Meneses A, Carli M, et al. The serotonin1A receptor partial agonist S15535 [4-(benzodioxan-5yl)1-(indan-2-yl)piperazine] enhances cholinergic transmission and cognitive function in rodents:a combined neurochemcial and behavioral analysis. J Pharmacol Exp Ther. 2004;311:190–203. doi: 10.1124/jpet.104.069625. [DOI] [PubMed] [Google Scholar]
  221. Miller G. Pharmacology. The puzzling rise and fall of a dark-horse Alzheimer's drug. Science. 2010;327(5971):1309. doi: 10.1126/science.327.5971.1309. [DOI] [PubMed] [Google Scholar]
  222. Mintzer JE, Kershaw P. The efficacy of galantamine in the treatment of Alzheimer's disease: comparison of patients previously treated with acetylcholinesterase inhibitors to patients with no prior exposure. Int J Geriatr Psychiatry. 2003;18:292–297. doi: 10.1002/gps.826. [DOI] [PubMed] [Google Scholar]
  223. Minzenberg MJ, Carter CS. Modafinil: a review of neurochemical actions and effects on cognition. Neuropsychopharmacology. 2008;33:1477–1502. doi: 10.1038/sj.npp.1301534. [DOI] [PubMed] [Google Scholar]
  224. Misane I, Ogren SO. Selective 5-HT1A antagonists WAY 100635 and NAD-299 attenuate the impairment of passive avoidance caused by scopolamine in the rat. Neuropsychopharmacology. 2003;28:253–264. doi: 10.1038/sj.npp.1300024. [DOI] [PubMed] [Google Scholar]
  225. Mitchell ES, Hoplight BJ, Lear SP, Neumaier JF. BGC20-761, a novel tryptamine analog, enhances memory consolidation and reverses scopolamine-induced memory deficit in social and visuospatial memory tasks through a 5-HT6 receptor-mediated mechanism. Neuropharmacology. 2006;50:412–420. doi: 10.1016/j.neuropharm.2005.10.002. [DOI] [PubMed] [Google Scholar]
  226. Miyamoto M, Takahashi H, Kato K, Hirai K, Ishihara Y, Goto G. Effects of 3-[1-(phenylmethyl)-4-piperidinyl]-1-(2,3,4,5-tetrahydro-1 -H-1-benzazepin-8-yl)-1-propanone fumarate (TAK-147), a novel acetylcholinesterase inhibitor, on impaired learning and memory in animal models. J Pharmacol Exp Ther. 1996;277(3):1292–1304. [PubMed] [Google Scholar]
  227. Miyazaki S, Onodera K, Imaizumi M, Timmerman H. Effects of clobenpropit (VUF-9153), a histamine H3-receptor antagonist, on learning and memory, and on cholinergic and monoaminergic systems in mice. Life Sci. 1997;61:355–361. doi: 10.1016/s0024-3205(97)00406-2. [DOI] [PubMed] [Google Scholar]
  228. Mohamed T, Rao PP. Design, synthesis and evaluation of 2,4-disubstituted pyrimidines as cholinesterase inhibitors. Bioorg Med Chem Lett. 2010;20(12):3606–3609. doi: 10.1016/j.bmcl.2010.04.108. [DOI] [PubMed] [Google Scholar]
  229. Mohler EG, Shacham S, Noiman S, Lezoualc’h F, Robert S, Gastineau M, et al. VRX-03011, a novel 5-HT4 agonist, enhances memory and hippocampal acetylcholine efflux. Neuropharmacology. 2007;53:563–573. doi: 10.1016/j.neuropharm.2007.06.016. [DOI] [PubMed] [Google Scholar]
  230. Monsma FJ, Shen Y, Ward RP, Hamblin MW, Sibley DR. Cloning and expression of a novel serotonin receptor with high-affinity for tricyclic psychotropic-drugs. Mol Pharmacol. 1993;43:320–327. [PubMed] [Google Scholar]
  231. Monti B, Berteotti C, Contestabile A. Subchronic rolipram delivery activates hippocampal CREB and Arc, enhances retention and slows down extinction of conditioned fear. Neuropsychpharmacology. 2006;31:278–286. doi: 10.1038/sj.npp.1300813. [DOI] [PubMed] [Google Scholar]
  232. Moser PC, Bergis OE, Jegham S, Lochead A, Duconseille E, Terranova JP, et al. SL65.0155, a novel 5-hydroxytryptamine4 receptor partial agonist with potent cognition-enhancing properties. J Pharmacol Exp Ther. 2002;302:731–741. doi: 10.1124/jpet.102.034249. [DOI] [PubMed] [Google Scholar]
  233. Murray KN, Abeles N. Nicotine’s effect on neural and cognitive functioning in an aging population. Aging & Mental Health. 2002;6:129–138. doi: 10.1080/13607860220126808. [DOI] [PubMed] [Google Scholar]
  234. Naghdi N, Harooni HE. The effect of intrahippocampal injections of ritanserin (5-HT2A/2C antagonist) and granisetron (5-HT3 antagonist) on learning as assessed in the spatial version of the water maze. Behav Brain Res. 2005;157:205–210. doi: 10.1016/j.bbr.2004.06.024. [DOI] [PubMed] [Google Scholar]
  235. Navakkode S, Sajikumar S, Frey JU. The type IV-specific phosphodiesterase inhibitor rolipram and its effect on hippocampal long-term potentiation and synaptic tagging. J Neurosci. 2004;24:7740–7744. doi: 10.1523/JNEUROSCI.1796-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  236. Ng HJ, Whittemore ER, Tran MB, Hogenkamp DJ, Broide RS, Johnstone TB, et al. Nootropic alpha7 nicotinic receptor allosteric modulator derived from GABAA receptor modulators. Proc Natl Acad Sci. 2007;104(19):8059–8064. doi: 10.1073/pnas.0701321104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  237. Ohara T, Fukaya H, Itanaka K, Seno N. Ameliorating effects of SDZ ENA 713 on age-associated decreases in learning performance and brain choline acetyltransferase activity in rats. Brain Res Bull. 1997;43:39–42. doi: 10.1016/s0361-9230(96)00349-8. [DOI] [PubMed] [Google Scholar]
  238. Ohman A, Josephsson S, Nygård L. Awareness through interaction in everyday occupations: experiences of people with Alzheimer's disease. Scand J Occup Ther. 2008 Mar;15(1):43–51. doi: 10.1080/11038120701441080. [DOI] [PubMed] [Google Scholar]
  239. Ogren SO, Eriksson TM, Elvander-Tottie E, D’Addario C, Ekstrom JC, Svenningsson P, et al. The role of 5-HT1A receptors in learning and memory. Behav Brain Res. 2008;195:54–77. doi: 10.1016/j.bbr.2008.02.023. [DOI] [PubMed] [Google Scholar]
  240. Ou LY, Tang XC, Cai JX. Effect of huperzine A on working memory in reserpine- or yohimbine-treated monkeys. Eur J Pharmacol. 2001;433:151–156. doi: 10.1016/s0014-2999(01)01500-x. [DOI] [PubMed] [Google Scholar]
  241. Parkinson study group. Randomized placebo-controlled study of the nicotinic agonist SIB-1508Y in Parkinson disease. Neurology. 2006;66(3):408–410. doi: 10.1212/01.wnl.0000196466.99381.5c. [DOI] [PubMed] [Google Scholar]
  242. Pascoli V, Boer-Saccomani C, Hermant J-F. H3 receptor antagonists reverse delay-dependent deficits in novel object discrimination by enhancing retrieval. Psychopharmacology. 2009;202:141–152. doi: 10.1007/s00213-008-1171-2. [DOI] [PubMed] [Google Scholar]
  243. Patat A, Parks V, Raje S, Plotka A, Chassard D, Coz FL. Safety, tolerability, pharmacokinetics and pharmacodynamics of ascending single and multiple doses of lecozotan in healthy young and elderly subjects. Brit J Clin Pharmacol. 2008;67:299–308. doi: 10.1111/j.1365-2125.2008.03348.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  244. Patel N, Spangler EL, Greig NH, Yu QS, Ingram DK, Meyer RC. Phenserine, a novel acetylcholinesterase inhibitor, attenuates impaired learning of rats in a 14-unit T-maze induced by blockade of the N-methyl-D-aspartate receptor. Neuroreport. 1998;9(1):171–176. doi: 10.1097/00001756-199801050-00035. [DOI] [PubMed] [Google Scholar]
  245. Patil CS, Singh VP, Singh S, Kulkarni SK. Modulatory effect of the PDE-5 inhibitor sildenafil in diabetic neuropathy. Pharmacology. 2004;72:192–195. doi: 10.1159/000080104. [DOI] [PubMed] [Google Scholar]
  246. Prendergast MA, Jackson WJ, Terry AV, Jr, Decker MW, Arneric SP, Buccafusco JJ. Central nicotinic receptor agonists ABT-418, ABT-089, and (−)-nicotine reduce distractibility in adult monkeys. Psychopharmacol (Berl) 1998;136(1):50–58. doi: 10.1007/s002130050538. [DOI] [PubMed] [Google Scholar]
  247. Peng Y, Lee DY, Jiang L, Ma Z, Schachter SC, Lemere CA. Huperzine A regulates amyloid precursor protein processing via protein kinase C and mitogenactivated protein kinase pathways in neuroblastoma SK-N-SH cells over-expressing wild type human amyloid precursor protein 695. Neuroscience. 2007;150:386–395. doi: 10.1016/j.neuroscience.2007.09.022. [DOI] [PubMed] [Google Scholar]
  248. Perez-Garcia G, Meneses A. Oral administration of the5-HT6 receptor antagonists SB-357134 and SB-399885 improves memory formation in an autoshaping learning task. Pharmacol Biochem Behav. 2005;81:673–682. doi: 10.1016/j.pbb.2005.05.005. [DOI] [PubMed] [Google Scholar]
  249. Petersen RC. Mild cognitive impairment as a diagnostic entity. J Intern Med. 2004 Sep;256(3):183–194. doi: 10.1111/j.1365-2796.2004.01388.x. [DOI] [PubMed] [Google Scholar]
  250. Pillot C, Ortiz J, Heron A, Ridray S, Schwartz JC, Arrang JM. Ciproxifan, a histamine H3-receptor antagonist/inverse agonist, potentiates neurochemical and behavioral effects of haloperidol in the rat. J Neurosci. 2002;22:7272–7280. doi: 10.1523/JNEUROSCI.22-16-07272.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  251. Pisani L, Catto M, Giangreco I, Leonetti F, Nicolotti O, Stefanachi A, et al. Design, Synthesis, and Biological Evaluation of Coumarin Derivatives Tethered to an Edrophonium-like Fragment as Highly Potent and Selective Dual Binding Site Acetylcholinesterase Inhibitors. ChemMedChem. 2010 Aug 2; doi: 10.1002/cmdc.201000210. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
  252. Pitsikas N, Bramibilla A, Borsini F. DAU 6215, a novel 5-HT3 receptor antagonist, improves performance in the aged rat in the Morris water maze task. Neurobiol Aging. 1993;14:561–564. doi: 10.1016/0197-4580(93)90039-e. [DOI] [PubMed] [Google Scholar]
  253. Pitsikas N, Borsini F. Itasetron (DAU 6215) prevents age-related memory deficits in the rat in a multiple choice avoidance task. Eur J Pharmacol. 1996;311:115–119. doi: 10.1016/0014-2999(96)00586-9. [DOI] [PubMed] [Google Scholar]
  254. Pitsikas N, Borsini F. Different effects of tropisetron and ondansetron in learning and memory paradigms. Pharmacol Biochem Behav. 1997;56:571–576. doi: 10.1016/s0091-3057(96)00516-3. [DOI] [PubMed] [Google Scholar]
  255. Pitsikas N, Rigamonti AE, Cella SG, Muller EE. The 5-HT1A receptor antagonist WAY 100635 improves rats performance in different models of amnesia evaluated by the object recognition task. Brain Res. 2003;983:215–222. doi: 10.1016/s0006-8993(03)03091-9. [DOI] [PubMed] [Google Scholar]
  256. Pollard H, Moreau J, Arrang JM, Schwartz JC. A detailed autoradiographic mapping of histamine H3 receptors in rat brain areas. Neuroscience. 1993;52:169–189. doi: 10.1016/0306-4522(93)90191-h. [DOI] [PubMed] [Google Scholar]
  257. Prendergast MA, Jackson WJ, Terry AV, Jr, Decker MW, Arneric SP, Buccafusco JJ. Central nicotinic receptor agonists ABT-418, ABT-089, and (−)-nicotine reduce distractibility in adult monkeys. Psychopharmacol (Berl) 1998;136(1):50–58. doi: 10.1007/s002130050538. [DOI] [PubMed] [Google Scholar]
  258. Prickaerts J, Steinbusch HW, Smits JF, de Vente J. Possible role of nitric oxide-cyclic GMP pathway in object recognition memory: effects of 7-nitroindazole and zaprinast. Eur J Pharmacol. 1997;337:125–136. doi: 10.1016/s0014-2999(97)01301-0. [DOI] [PubMed] [Google Scholar]
  259. Prickaerts J, van Staveren WC, Sik A, Markerink-van Ittersum M, Niewohner U, van der Staay FJ, et al. Effects of two selective Phosphodiesterase type 5 inhibitors, sidenafil and vardenafil, on object recognition memory and hippocampal cyclic GMP levels in the rat. Neuroscience. 2002;113:351–361. doi: 10.1016/s0306-4522(02)00199-9. [DOI] [PubMed] [Google Scholar]
  260. Prickaerts J, Sik A, van der Staay FJ, de Vente J, Blokland A. Dissociable effects of acetylcholinesterase inhibitors and phosphodiesterase type 5 inhibitors on object recognition memory: acquisition versus consolidation. Psychopharmacology. 2005;177:381–390. doi: 10.1007/s00213-004-1967-7. [DOI] [PubMed] [Google Scholar]
  261. Qiu C, Kivipelto M, von Strauss E. Epidemiology of Alzheimer's disease: occurrence, determinants, and strategies toward intervention. Dialogues Clin Neurosci. 2009;11(2):111–128. doi: 10.31887/DCNS.2009.11.2/cqiu. [DOI] [PMC free article] [PubMed] [Google Scholar]
  262. Quirion R. Cholinergic markers in Alzheimer disease and the autoregulation of acetylcholine release. J Psychiatr Neurosci. 1993;18:226–234. [PMC free article] [PubMed] [Google Scholar]
  263. Quirion R, Wilson A, Rowe W, Aubert I, Richard J, Doods H, et al. Facilitation of acetylcholine release and cognitive performance by an M2-muscarinic receptor antagonist in aged memory-impaired rats. J Neurosci. 1995;15:1455–1462. doi: 10.1523/JNEUROSCI.15-02-01455.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  264. Racchi M, Mazzucchelli M, Lenzken SC, Porrello E, Lanni C, Govoni S. Role of acetylcholinesterase inhibitors in the regulation of amyloid beta precursor protein (AbetaPP) metabolism. Chem. Biol. Interact. 2005:157–158. 335–338. doi: 10.1016/j.cbi.2005.10.099. [DOI] [PubMed] [Google Scholar]
  265. Ramirez MJ, Cenarruzabeitia E, Lasheras B, Del Rio J. Invovlement of GABA systems in acetylcholine release induced by 5-HT3 receptor blockade in slices from rat entorhinal cortex. Brain Res. 1996;712:274–280. doi: 10.1016/0006-8993(95)01471-3. [DOI] [PubMed] [Google Scholar]
  266. Ramos BP, Birnbaum SG, Lindenmeyer I, Newton SS, Duman RS, Arnsten AF. Dysregulation of protein kinase a signaling in the aged prefrontal cortex: new strategy for treating age-related cognitive decline. Neuron. 2003;40:835–845. doi: 10.1016/s0896-6273(03)00694-9. [DOI] [PubMed] [Google Scholar]
  267. Randt CT, Judge ME, Bonnet KA, Quartermain D. Brain cyclic AMP and memory in mice. Pharmacol Biochem Behav. 1982;17:677–680. doi: 10.1016/0091-3057(82)90344-6. [DOI] [PubMed] [Google Scholar]
  268. Raskind MA, Peskind ER, Wessel T, Yuan W. Galantamine in AD: A 6-month randomized, placebo-controlled trial with a 6-month extension. The Galantamine USA-1 Study Group. Neurology. 2000;54(12):2261–2268. doi: 10.1212/wnl.54.12.2261. [DOI] [PubMed] [Google Scholar]
  269. Reisine TD, Yamamura HI, Bird ED, Spokes E, Enna SJ. Pre- and postsynaptic neurochemical alterations in Alzheimer's disease. Brain Res. 1978 Dec 29;159(2):477–481. doi: 10.1016/0006-8993(78)90562-0. [DOI] [PubMed] [Google Scholar]
  270. Reneerkens OAH, Rutten K, Steinbusch HWM, Blokland A, Prickaerts J. Selective Phosphodiesterase inhibitors: a promising target for cognition enhancement. Psychopharmacology. 2009;202:419–443. doi: 10.1007/s00213-008-1273-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  271. Rezvani AH, Kholdebarin E, Brucato FH, Callahan PM, Lowe DA, Levin ED. Effect of R3487/MEM3454, a novel nicotinic alpha7 receptor partial agonist and 5-HT3 antagonist on sustained attention in rats. Prog Neuropsychopharmacol Biol Psychiatry. 2009;33(2):269–275. doi: 10.1016/j.pnpbp.2008.11.018. [DOI] [PubMed] [Google Scholar]
  272. Riemer C, Borroni E, Levet-Trafit B, Martin JR, Poli S, Porter RHP, et al. Influence of the 5-HT6 receptor on acetylcholine release in the cortex: pharmacological characterization of 4-(2-bromo-6-pyrrolidin-1-ylpyridine-4-sulfonyl)phenylamine, a potent, selective 5-HT6 receptor antagonist. J Med Chem. 2003;46:1273–1276. doi: 10.1021/jm021085c. [DOI] [PubMed] [Google Scholar]
  273. Rizzo S, Bartolini M, Ceccarini L, Piazzi L, Gobbi S, Cavalli A, et al. Targeting Alzheimer's disease: Novel indanone hybrids bearing a pharmacophoric fragment of AP2238. Bioorg Med Chem. 2010 Mar 1;18(5):1749–1760. doi: 10.1016/j.bmc.2010.01.071. Epub 2010 Feb 4. [DOI] [PubMed] [Google Scholar]
  274. Robert SJ, Zugaza JL, Fischmeister R, Gardier AM, Lezoualc’h F. The human serotonin 5-HT4 receptor regulates secretion of non-amyloidogenic precursor protein. J Biol Chem. 2001;276:44881–44888. doi: 10.1074/jbc.M109008200. [DOI] [PubMed] [Google Scholar]
  275. Roberts JC, Reavill C, East SZ, Harrison PJ, Patel S, Routledge C, et al. The distribution of 5-HT6 receptors in rat brain: an autoradiographic binding study using the radiolabelled 5-HT6 receptor antagonist I-125 SB-258585. Brain Res. 2002;934:49–57. doi: 10.1016/s0006-8993(02)02360-0. [DOI] [PubMed] [Google Scholar]
  276. Rodefer JS, Murphy ER, Baxter MG. PDE10A inhibition reverses subchronic PCP-induced deficits in attentional set-shifitng in rats. Eur J Neurosci. 2005;21:1070–1076. doi: 10.1111/j.1460-9568.2005.03937.x. [DOI] [PubMed] [Google Scholar]
  277. Rodefer JS, Baseile JL, Callahan PM, Van Natta K, Yeo H, Lowe DA, Santarelli L, Vivian JA. Attentional set-shifting in rats and monkeys: Effects of cholinergic challenges. Soc Neurosci Abstr. 2007 746.4. [Google Scholar]
  278. Rodefer JS, Nguyen TN, Karlsson JJ, Arnt J. Reversal of subchronic PCP-induced deficits in attentional set shifting in rats by sertindole and a 5-HT6 receptor antagonist: comparison among antipsychotics. Neuropsychopharmacology. 2008;33:2657–2666. doi: 10.1038/sj.npp.1301654. [DOI] [PubMed] [Google Scholar]
  279. Rogers DC, Hagan JJ. 5-HT6 receptor antagonists enhance retention of a water-maze task in the rat. Psychopharmacology. 2001;158:114–119. doi: 10.1007/s002130100840. [DOI] [PubMed] [Google Scholar]
  280. Rollema H, Hajos M, Seymour PA, Kozak R, Majchrzsak MJ, Guanowsky V, et al. Preclinical pharmacology of the α4β2 nAChR partial agonist varenicline related to effects on reward, mood and cognition. Biochem Pharmacol. 2009;78:813–824. doi: 10.1016/j.bcp.2009.05.033. [DOI] [PubMed] [Google Scholar]
  281. Romero G, Sanchez E, Pujol M, Perez P, Codony X, Holenz J, et al. Efficacy of selective 5-HT6 receptor ligands determined by monitoring 5-HT6 receptor-mediated cAMP signaling pathways. Brit J Pharmacol. 2006;148:1133–1143. doi: 10.1038/sj.bjp.0706827. [DOI] [PMC free article] [PubMed] [Google Scholar]
  282. Roncarati R, Scali C, Comery TA, Grauer SM, Aschmi S, Bothmann H, et al. Pro-cognitive and neuroprotective activity of a novel α7 nicotinic acetylcholine receptor agonist for treatment of neurodegenerative and cognitive disorders. J Pharmacol Exp Ther. 2009;329(2):459–468. doi: 10.1124/jpet.108.150094. [DOI] [PubMed] [Google Scholar]
  283. Rose GM, Hopper AH, De Vivo M, Tehim A. Phosphodiesterase inhibitors for cognitive enhancement. Curr Pharm Design. 2005;11:3329–3334. doi: 10.2174/138161205774370799. [DOI] [PubMed] [Google Scholar]
  284. Roth BL, Hanizavareh SM, Blum AE. Serotonin receptors represent highly favorable molecular targets for cognitive enhancement in schizophrenia and other disorders. Pyschopharmacology. 2004;174:17–24. doi: 10.1007/s00213-003-1683-8. [DOI] [PubMed] [Google Scholar]
  285. Routledge C, Bromidge SM, Moss SF, Price GW, Hirst W, Newman H, et al. Characterization of SB-271046: a potent, selective and orally active 5-HT6 receptor antagonist. Brit J Pharmacol. 2000;130:1606–1612. doi: 10.1038/sj.bjp.0703457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  286. Rowe WB, O'Donnell JP, Pearson D, Rose GM, Meaney MJ, Quirion R. Long-term effects of BIBN-99, a selective muscarinic M2 receptor antagonist, on improving spatial memory performance in aged cognitively impaired rats. Behav Brain Res. 2003;145:171–178. doi: 10.1016/s0166-4328(03)00116-5. [DOI] [PubMed] [Google Scholar]
  287. Rowe WB, DiSomma RM, Hsu CC, Solomon K, Callahan PM, Wang D, et al. Histamine H3 receptor antagonists elicit pro-cognitive effects on episodic and spatial reference memories in young and aged animals. Alzheimer’s and Dementia. 2006;2:S647. [Google Scholar]
  288. Rowe WB, Blalock EM, Chen KC, Kadish I, Wang D, Barrett JE, et al. Hippocampal expression analyses reveal selective association of immediate-early, neuroenergetic, and myelinogenic pathways with cognitive impairment in aged rats. J Neurosci. 2007;27:3098–3110. doi: 10.1523/JNEUROSCI.4163-06.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  289. Rowe WB, Callahan PM, Xie W, Hsu CC, Dunn R, Murray M, et al. Serotonin 5-HT6 receptor antagonists modulate episodic, spatial and working memory highlightinh a potential therapeutic role in alleviating deficits associated with mild cognitive impairment and Alzheimer’s disease. Soc Neurosci Abstr. 2008;685:9. [Google Scholar]
  290. Rowe WB, Callahan PM, Hsu CC, Tehim A. Characterization of the 5-HT6 receptor agonist, WAY 181187 and antagonist, SB 742457: effects on spatial reference and spatial working memory function in aged F344 rats. Soc Neurosci Abstr. 2009;883:2. [Google Scholar]
  291. Ruat M, Traiffort E, Arrang JM, Tardivellacombe J, Diaz J, Leurs R, et al. A novel rat serotonin (5-HT6) receptor-molecular cloning, localization and stimulation of cAMP accumulation. Biochem Biophys Res Comm. 1993;193:268–276. doi: 10.1006/bbrc.1993.1619. [DOI] [PubMed] [Google Scholar]
  292. Rupniak NM, Tye SJ, Brazell C, Heald A, Iversen SD, Pagella PG. Reversal of cognitive impairment by heptyl physostigmine, a long-lasting cholinesterase inhibitor, in primates. J Neurol Sci. 1992;107(2):246–249. doi: 10.1016/0022-510x(92)90296-w. [DOI] [PubMed] [Google Scholar]
  293. Ruske AC, White KG. Facilitation of memory performance by a novel muscarinic agonist in young and old rats. Pharmacol Biochem Behav. 1999;63(4):663–667. doi: 10.1016/s0091-3057(99)00037-4. [DOI] [PubMed] [Google Scholar]
  294. Rutten K, Basile JL, Prickaerts J, Blokland A, Vivian JA. Selective PDE inhibitors rolipram and sildenafil improve object retrieval performance in adult cynomolgus macaques. Psychopharmacology. 2008;196:643–648. doi: 10.1007/s00213-007-0999-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  295. Rutten K, Prickaerts J, Hendrix M, van der Staay FJ, Sik A, Blokland A. Time-dependent involvement of cAMP and cGMP in consolidation of object memory: studies using selective phosphodiesterase type 2, 4 and 5 inhibitors. Eur J Pharmacol. 2007;558:107–112. doi: 10.1016/j.ejphar.2006.11.041. [DOI] [PubMed] [Google Scholar]
  296. Rutten K, Prickaerts J, Blokland A. Rolipram reverses scopolamine-induced and time-dependent memory deficits in object recognition by different mechanisms of action. Neurobiol Learn Mem. 2006;85:132–138. doi: 10.1016/j.nlm.2005.09.002. [DOI] [PubMed] [Google Scholar]
  297. Rutten K, Vente JD, Sik A, Ittersum MM, Prickaerts J, Blokland A. The selective PDE5 inhibitor, sildenafil, improves object memory in Swiss mice and increases cGMP levels in hippocampus. Behav Brain Res. 2005;164:11–16. doi: 10.1016/j.bbr.2005.04.021. [DOI] [PubMed] [Google Scholar]
  298. Sabbagh MN. Drug development for Alzheimer’s disease: where are we now and where are we headed? Am J Geriatr Pharm. 2009;7:167–185. doi: 10.1016/j.amjopharm.2009.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  299. Sadashiva CT, Narendra Sharath Chandra JN, Kavitha CV, Thimmegowda A, Subhash MN, Rangappa KS. Synthesis and pharmacological evaluation of novel N-alkyl/aryl substituted thiazolidinone arecoline analogues as muscarinic M1 receptor agonists in Alzheimer’s dementia models. Eur J Med Chem. 2009;44:4848–4854. doi: 10.1016/j.ejmech.2009.07.026. [DOI] [PubMed] [Google Scholar]
  300. Sagi Y, Weinstock M, Youdim MB. Attenuation of MPTP-induced dopaminergic neurotoxicity by TV3326, a cholinesterase-monoamine oxidase inhibitor. J. Neurochem. 2003;86:290–297. doi: 10.1046/j.1471-4159.2003.01801.x. [DOI] [PubMed] [Google Scholar]
  301. Samadi A, Marco-Contelles J, Soriano E, Alvarez-Pérez M, Chioua M, Romero A, et al. Multipotent drugs with cholinergic and neuroprotective properties for the treatment of Alzheimer and neuronal vascular diseases. I. Synthesis, biological assessment, and molecular modeling of simple and readily available 2-aminopyridine-, and 2-chloropyridine-3,5-dicarbonitriles. Bioorg Med Chem. 2010;18(16):5861–5872. doi: 10.1016/j.bmc.2010.06.095. [DOI] [PubMed] [Google Scholar]
  302. Schaffhauser H, Mathiasen JR, Dicamillo A, Huffman MJ, Lu LD, McKenna BA, et al. Dimebolin is a 5-HT6 antagonist with acute cognition enhancing activities. Biochem Pharmacol. 2009;78:1035–1042. doi: 10.1016/j.bcp.2009.06.021. [DOI] [PubMed] [Google Scholar]
  303. Schechter LE, Lin Q, Smith DL, Zhang G, Shan Q, Platt B, et al. Neuropharmacological profile of novel and selective 5-HT6 receptor agonists: WAY-181187 and WAY-208466. Neuropsychopharmacology. 2008;33:1323–1335. doi: 10.1038/sj.npp.1301503. [DOI] [PubMed] [Google Scholar]
  304. Schechter LE, Smith DL, Rosenzweig-Lipson S, Sukoff SJ, Dawson LA, Marquis K, et al. Lecozotan (SRA-333): a selective serotonin 1A receptor antagonist that enhances the stimulated release of glutamate and acetylcholine in the hippocampus and possesses cognitive-enhancing properties. J Pharmacol Exp Ther. 2005;314:1274–1289. doi: 10.1124/jpet.105.086363. [DOI] [PubMed] [Google Scholar]
  305. Schlicker E, Malinowska B, Kathmann M, Gothert M. Modulation of neurotransmitter release via histamine H3 heteroreceptors. Fund Clin Pharmacol. 1994;8:128–137. doi: 10.1111/j.1472-8206.1994.tb00789.x. [DOI] [PubMed] [Google Scholar]
  306. Schmidt CJ, Chapin DS, Cianfrogna J, Corman ML, Hajos M, Harms JF, et al. Preclinical characterization of selective phosphodiesterase 10A inhibitiors: a new therapeutic approach to the treatment of schizophrenia. J Pharmacol Exper Ther. 2008;325:681–690. doi: 10.1124/jpet.107.132910. [DOI] [PubMed] [Google Scholar]
  307. Schneider JS, Tinker JP, Van Velson M, Menzaghi F, Lloyd GK. Nicotinic acetylcholine receptor agonist SIB-1508Y improves cognitive functioning in chronic low-dose MPTP-treated monkeys. J Pharmacol Exp Ther. 1999;290(2):731–739. [PubMed] [Google Scholar]
  308. Scheider JS, Tinker JP, Menzaghi F, Lloyd GK. The subtype selective nicotinic acetylcholine receptor agonist SIB-1553A improves both attention and working memory components of a spatial working memory task in chronic low dose 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine-treated monkeys. J Pharmacol Exp Ther. 2003;306(1):401–406. doi: 10.1124/jpet.103.051912. [DOI] [PubMed] [Google Scholar]
  309. Schreiber R, Vivian J, Hedley L, Szczepanski K, Secchi RL, Zuzow M, et al. Effects of the novel 5-HT6 receptor antagonist RO4368554 in rat models for cognition and sensorimotor gating. Eur Neuropsychopharm. 2007;17:277–288. doi: 10.1016/j.euroneuro.2006.06.009. [DOI] [PubMed] [Google Scholar]
  310. Setter SM, Iltz JL, Fincham JE, Campbell RK, Baker DE. Phosphodiesterase 5 inhibitors for erectile dysfunction. Ann Pharmacother. 2005;39:1286–1295. doi: 10.1345/aph.1E487. [DOI] [PubMed] [Google Scholar]
  311. Seu E, Lang A, Rivera RJ, Jentsch JD. Inhibition of the norepinephrine transporter improves behavioral flexibility in rats and monkeys. Psychopharmacology (Berl) 2009;202:505–519. doi: 10.1007/s00213-008-1250-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  312. Sheng R, Xu Y, Hu C, Zhang J, Lin X, Li J, et al. Design, synthesis and AChE inhibitory activity of indanone and aurone derivatives. Eur. J. Med. Chem. 2009;44:7–17. doi: 10.1016/j.ejmech.2008.03.003. [DOI] [PubMed] [Google Scholar]
  313. Shafiei M, Mahmoudian M, Rostami P, Nemati F. Effect of sidenafil (Viagra) on memory retention of a passive avoidance response in rats. Acta Physiol Hung. 2006;93:53–59. doi: 10.1556/APhysiol.93.2006.1.6. [DOI] [PubMed] [Google Scholar]
  314. Shirazi-Southall S, Rodriguez DE, Nomikos GG. Effects of typical and atypical antipsychotoics and receptor selective compounds on acetylcholine efflux in the hippocampus of the rat. Neuropsychopharmacology. 2002;26:583–594. doi: 10.1016/S0893-133X(01)00400-6. [DOI] [PubMed] [Google Scholar]
  315. Shoham S, Bejar C, Kovalev E, Schorer-Apelbaum D, Weinstock M. Ladostigil prevents gliosis, oxidative-nitrative stress and memory deficits induced by intracerebroventricular injection of streptozotocin in rats. Neuropharmacology. 2007;52(3):836–843. doi: 10.1016/j.neuropharm.2006.10.005. Epub 2006 Nov 22. [DOI] [PubMed] [Google Scholar]
  316. Si W, Zhang X, Niu Y, Yu H, Lei X, Chen H, et al. A novel derivative of xanomeline improves fear cognition in aged mice. Neurosci Lett. 2010;473:115–119. doi: 10.1016/j.neulet.2010.02.031. [DOI] [PubMed] [Google Scholar]
  317. Simon R. Lost in translation: problems and pitfalls in translating laboratory observations to clinical utility. Eur J Cancer. 2008;44:2707–2713. doi: 10.1016/j.ejca.2008.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  318. Son H, Lu YF, Zhuo M, Arancio O, Kandel ER, Hawkins RD. The specific role of cGMP in hippocampal LTP. Learn Mem. 1998;5:231–245. [PMC free article] [PubMed] [Google Scholar]
  319. Sood A, Beach WJ, Webster SJ, Terry AV, Buccafusco JJ. The effects of JWB1-84-1 on memory related task performance by amyloid Abeta transgenic mice and by young and aged monkeys. Neuropharm. 2007;53(5):588–600. doi: 10.1016/j.neuropharm.2007.06.028. [DOI] [PubMed] [Google Scholar]
  320. Southam E, Cilia J, Gartlon JE, Woolley ML, Lacroix LP, Jennings CA, et al. Preclinical investigations into the antipsychotic potential of the novel histamine H3 receptor antagonist GSK207040. Psychopharmacology. 2009;201:483–494. doi: 10.1007/s00213-008-1310-9. [DOI] [PubMed] [Google Scholar]
  321. Spalding TAMAJN, Ott TR, Friberg M, Bajpai A, Bradley SR, et al. Structural requirements of transmembrane domain 3 for activation by the M1 muscarinic receptor agonists AC-42, AC-260584, clozapine, and N-desmethylclozapine: evidence for three distinct modes of receptor activation. Mol Pharmacol. 2006;70(6):1974–1983. doi: 10.1124/mol.106.024901. [DOI] [PubMed] [Google Scholar]
  322. Spencer JP, Brown JT, Richardson JC, Medhurst AD, Sehmi SS, Calver AR, et al. Modulation of hippocampal excitability by 5-HT4 receptor agonists persists in a transgenic model of Alzheimer’s disease. Neuroscience. 2004;129:49–54. doi: 10.1016/j.neuroscience.2004.06.070. [DOI] [PubMed] [Google Scholar]
  323. Standridge JB. Pharmacotherapeutic approaches to the treatment of Alzheimer's disease. Clin Ther. 2004 May;26(5):615–630. doi: 10.1016/s0149-2918(04)90064-1. [DOI] [PubMed] [Google Scholar]
  324. Stean TO, Hirst WD, Thomas DR, Price GW, Rogers D, Riley G, et al. Pharmacological profile of SB-357134: a potent, selective brain penetrant, and orally active 5-HT(6) receptor antagonist. Pharmacol Biochem Behav. 2002;71:645–654. doi: 10.1016/s0091-3057(01)00742-0. [DOI] [PubMed] [Google Scholar]
  325. Sterling J, Herzig Y, Goren T, Finkelstein N, Lerner D, Goldenberg W, et al. Novel dual inhibitors of AChE and MAO derived from hydroxy aminoindan and phenethylamine as potential treatment for Alzheimer’s disease. J. Med. Chem. 2002;45:5260–5279. doi: 10.1021/jm020120c. [DOI] [PubMed] [Google Scholar]
  326. Studzinski CM, Araujo JA, Milgram NW. The canine model of human cognitive aging and dementia: pharmacological validity of the model for assessment of human cognitive-enhancing drugs. Prog Neuropsychopharmacol Biol Psychiatry. 2005;29(3):489–498. doi: 10.1016/j.pnpbp.2004.12.014. [DOI] [PubMed] [Google Scholar]
  327. Sullivan JP, Donnelly-Roberts D, Briggs CA, Anderson DJ, Gopalakrishnan M, Xue IC, et al. ABT-089 [2-methyl-3-(2-(S)-pyrrolidinylmethoxy)pyridine]: I. A potent and selective cholinergic channel modulator with neuroprotective properties. J Pharmacol Exp Ther. 1997;283(1):235–246. [PubMed] [Google Scholar]
  328. Svenningsson P, Tzavara ET, Qi HS, Carruthers R, Witkin JM, Nomikos GG, et al. Biochemical and behavioral evidence for antidepressant-like effects of 5-HT6 receptor stimulation. J Neurosci. 2007;27:4201–4209. doi: 10.1523/JNEUROSCI.3110-06.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  329. Sydserff S, Sutton EJ, Song D, Quirk MC, Maciag C, Li C, et al. Selective alpha7 nicotinic receptor activation by AZD0328 enhances cortical dopamine release and improves learning and attentional processes. Biochem Pharmacol. 2009;78(7):880–888. doi: 10.1016/j.bcp.2009.07.005. [DOI] [PubMed] [Google Scholar]
  330. Taly A, Corringer PJ, Guedin D, Lestage P, Changeux JP. Nicotinic receptors: allosteric transitions and therapeutic targets in the nervous system. Nat Rev Drug Dis. 2009;8(9):733–750. doi: 10.1038/nrd2927. [DOI] [PubMed] [Google Scholar]
  331. Tecle H, Schwarz RD, Barrett SD, Callahan MJ, Caprathe BW, Davis RE, et al. CI-1017, a functionally M1-selective muscarinic agonist: design, synthesis, and preclinical pharmacology. Pharm Acta Helv. 2000;74(2–3):141–148. doi: 10.1016/s0031-6865(99)00027-8. [DOI] [PubMed] [Google Scholar]
  332. Terry AV, Jr, Buccafusco JJ, Prendergast MA, Jackson WJ, Fontana DL, Wong EH, et al. The 5-HT3 receptor antagonist, RS-56812, enhances delayed matching performance in monkeys. Neuroreport. 1996;8:49–54. doi: 10.1097/00001756-199612200-00011. [DOI] [PubMed] [Google Scholar]
  333. Terry AV, Jr, Buccafusco JJ, Jackson WJ, Prendergast MA, Fontana DJ, Wong EHF, et al. Enhanced delayed matching performance in younger and older macaques administered the 5HT4 receptor agonist, RS 17017. Psychopharmacology. 1998;135:407–415. doi: 10.1007/s002130050529. [DOI] [PubMed] [Google Scholar]
  334. Terry AV, Jr, Gattu M, Buccafusco JJ, Sowell JW, Kosh JW. Ranitidine Analog, JWS USC 75IX, Enhances Memory Related Task Performance in Rats. Drug Development Research. 1999;47:97–106. [Google Scholar]
  335. Terry AV, Jr, Buccafusco JJ. The Cholinergic Hypothesis of Age and Alzheimer’s Disease-Related Cognitive Deficits: Recent Challenges and Their Implications for Novel Drug Development. J Pharmacol Exper Ther. 2003;306(3):821–827. doi: 10.1124/jpet.102.041616. [DOI] [PubMed] [Google Scholar]
  336. Terry AV, Jr, Hernandez CM, Hohnadel EJ, Bouchard KP, Buccafusco JJ. Cotinine. A Neuroactive Metabolite of Nicotine: Potential for Treating Disorders of Impaired Cognition. CNS Drug Reviews. 2005a;11:229–252. doi: 10.1111/j.1527-3458.2005.tb00045.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  337. Terry AV, Jr, Buccafusco JJ, Bartoszyk GD. Selective serotonin 5-HT2A receptor antagonist EMD 281014 improves delayed matching performance in young and aged rhesus monkeys. Psychopharmacology. 2005b;179:725–732. doi: 10.1007/s00213-004-2114-1. [DOI] [PubMed] [Google Scholar]
  338. Terry AV, Jr, Buccafusco JJ, Wilson C. Cognitive dysfunction in neuropsychiatric disorders: selected serotonin receptor subtypes as therapeutic targets. Behav Brain Res. 2008;195:30–38. doi: 10.1016/j.bbr.2007.12.006. [DOI] [PubMed] [Google Scholar]
  339. Terry AV, Buccafusco JJ, Herman EJ, Callahan PM, Beck WD, Warner S, Vandenhuerk L, Bouchard K, Schwarz GM, Gao J, Chapman JM. Prototypical ranitidine analog, JWS-USC-75-IX, improves information processing and cognitive function in animal models. J Pharmacol Exper Ther. 2011 doi: 10.1124/jpet.110.175422. (in press) [DOI] [PMC free article] [PubMed] [Google Scholar]
  340. Tzavara ET, Bymaster FP, Felder CC, Wade M, Gomeza J, Wess J, McKinzie DL, Nomikos GG. Dysregulated hippocampal acetylcholine neurotransmission and impaired cognition in M2, M4 and M2/M4 muscarinic receptor knockout mice. Mol. Psychiatry. 2003;8:673–679. doi: 10.1038/sj.mp.4001270. [DOI] [PubMed] [Google Scholar]
  341. The Centers for Disease Control and Prevention. Public health and aging: trends in aging – United States and worldwide. JAMA. 2003;289:1371–1373. [PubMed] [Google Scholar]
  342. Thomas SJ, Grossberg GT. Memantine: a review of studies into its safety and efficacy in treating Alzheimer's disease and other dementias. Clin Interv Aging. 2009;4:367–377. doi: 10.2147/cia.s6666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  343. Timmermann DB, Gronlien JH, Kohlhaas KL, Neilsen EO, Dam E, Jorgensen TD, et al. An allosteric modulator of the alpha7 nicotinic acetylcholine receptor possessing cognition-enhancing properties in vivo. J Pharmacol Exp Ther. 2007;323:294–307. doi: 10.1124/jpet.107.120436. [DOI] [PubMed] [Google Scholar]
  344. Trabace L, Cassano T, Colaianna M, Castrignanò S, Giustino A, Amoroso S, et al. Neurochemical and neurobehavioral effects of ganstigmine (CHF2819), a novel acetylcholinesterase inhibitor, in rat prefrontal cortex: an in vivo study. Pharmacol Res. 2007;56(4):288–294. doi: 10.1016/j.phrs.2007.07.006. Epub 2007 Jul 27. [DOI] [PubMed] [Google Scholar]
  345. Tully T, Bourtchouladze R, Scott R, Tallman J. Targeting the CREB pathway for memory enhancers. Nat Rev Drug Discov. 2003;2:267–277. doi: 10.1038/nrd1061. [DOI] [PubMed] [Google Scholar]
  346. United Nations. World Population Ageing 2007. New York: United Nations; 2007
  347. Upton N, Chuang TT, Hunter AJ, Virley DJ. 5-HT6 receptor antagonists as novel cognitive enhancing agents for Alzheimer’s disease. Neurotherapeutics. 2008;5:458–469. doi: 10.1016/j.nurt.2008.05.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  348. Valli MJ, Tang Y, Kosh JW, Chapman JM, Jr, Sowell JW., Sr Synthesis and cholinergic properties of N-aryl-2-[[[5-[(dimethylamino)methyl]-2-furanyl]methyl]thio]ethylamino analogs of ranitidine. J Med Chem. 1992;35:3141–3147. doi: 10.1021/jm00095a008. [DOI] [PubMed] [Google Scholar]
  349. Van der Schyf CJ, Mandel S, Geldenhuys WJ, Amit T, Avramovich Y, Zheng H, et al. Novel multifunctional anti-Alzheimer drugs with various CNS neurotransmitter targets and neuroprotective moieties. Curr Alzheimer Res. 2007;4:522–536. doi: 10.2174/156720507783018226. [DOI] [PubMed] [Google Scholar]
  350. van der Staay FJ, Rutten K, Barfacker L, DeVry J, Erb C, Heckroth H, et al. The novel selective PDE9 inhibitor BAY 73–6691 improves learning and memory in rodents. Neuropharmacology. 2008;55:908–918. doi: 10.1016/j.neuropharm.2008.07.005. [DOI] [PubMed] [Google Scholar]
  351. van Donkelaar EL, Rutten K, Blokland A, Akkerman S, Steinbusch HWM, Prickaerts J. Phosphodiesterase 2 and 5 inhibiton attenuates the object memory deficit induced by acute tryptophan depletion. Eur J Pharmacol. 2008;600:98–104. doi: 10.1016/j.ejphar.2008.10.027. [DOI] [PubMed] [Google Scholar]
  352. van Kampen M, Selbach K, Schneider R, Schiegel E, Boess F, Schreiber R. AR-R 17779 improves social recognition in rats by activation of nicotinic alpha7 receptors. 2004;172(4):375–383. doi: 10.1007/s00213-003-1668-7. [DOI] [PubMed] [Google Scholar]
  353. van Marum RJ. Update on the use of memantine in Alzheimer's disease. Neuropsychiatr Dis Treat. 2009;5:237–247. doi: 10.2147/ndt.s4048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  354. Vilaro MT, Cortes R, Mengod G. Serotonin 5-HT4 receptors and their mRNAs in rat and guinea pig brain: distribution and effects of neurotoxic lesions. J Comp Neurol. 2005;484:418–439. doi: 10.1002/cne.20447. [DOI] [PubMed] [Google Scholar]
  355. Vitolo OV, Sant’Angelo A, Costanzo V, Battaglia F, Arancio O, Shelanski M. Amyloid beta-peptide inhibition of the PKA/CREB pathway and long-term potentiation: reversibility by drugs that enhance cAMP signaling. Pro Natl Acad Sci USA. 2002;99:13217–13221. doi: 10.1073/pnas.172504199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  356. Wallace TL, Callahan PM, Tehim A, Bertrand D, Tombaugh G, Wang S, Xie W, Rowe WB, Ong V, Graham E, Terry AV, Jr, Rodefer JS, Herbert B, Murray M, Porter R, Santarelli L, Lowe DA. RG3487, a novel nicotinic α7 receptor partial agonist, improves cognition and sensorimotor gating in rodents. J Pharmacol Exp Ther. 2011;336:242–253. doi: 10.1124/jpet.110.171892. [DOI] [PubMed] [Google Scholar]
  357. Wallace-Boone TL, Chiu G, Dao H, Lowe D, Porter R, Santarelli L. R3487/MEM3454, a novel nicotinic alpha 7 receptor partial agonist, improves attention and working memory performance in cynomolgus macaques. Soc Neurosci Abstr. 2009:887.5. [Google Scholar]
  358. Wallace-Boone TL, Yeo H, Callahan PM, Tombaugh G, Misner DL, Grovey B, Thompson J, Van Natta K, Sahdeo S, Tehim A, Maag H, Santarelli L. Pro-cognitive properties of MEM 64368, a novel nicotinic alpha 7 receptor partial agonist, following acute and chronic administration. Biochem Pharmacol. 2007;74 SMA-45. [Google Scholar]
  359. Wang T, Tang XC. Reversal of scopolamine-induced deficits in radial maze performance by (−)-huperzine A: comparison with E2020 and tacrine. Eur J Pharmacol. 1998;349:137–142. doi: 10.1016/s0014-2999(98)00199-x. [DOI] [PubMed] [Google Scholar]
  360. Wang ZF, Tang LL, Yan H, Wang YJ, Tang XC. Effects of huperzine A on memory deficits and neurotrophic factors production after transient cerebral ischemia and reperfusion in mice. Pharmacol Biochem Behav. 2006;83(4):603–611. doi: 10.1016/j.pbb.2006.03.027. Epub 2006 May 9. [DOI] [PubMed] [Google Scholar]
  361. Wang J, Zhang HY, Tang XC. Huperzine a improves chronic inflammation and cognitive decline in rats with cerebral hypoperfusion. J Neurosci Res. 2010;88(4):807–815. doi: 10.1002/jnr.22237. [DOI] [PubMed] [Google Scholar]
  362. Watkins PB, Zimmerman HJ, Knapp MJ, Gracon SI, Lewis KW. Hepatotoxic effects of tacrine administration in patients with Alzheimer's disease. JAMA. 1994;271(13):992–998. [PubMed] [Google Scholar]
  363. Weinreb O, Bar-Am O, Amit T, Chillag-Talmor O, Youdim MBH. Neuroprotection via pro-survival protein kinase C isoforms associated with Bcl-2 family members. FASEB Journal. 2004;18:1471–1473. doi: 10.1096/fj.04-1916fje. [DOI] [PubMed] [Google Scholar]
  364. Weinreb O, Amit T, Bar-Am O, Yogev-Falach M, Youdim MB. The neuroprotective mechanism of action of the multimodal drug ladostigil. Front Biosci. 2008;13:5131–5137. doi: 10.2741/3069. [DOI] [PubMed] [Google Scholar]
  365. Weinstock M, Behar C, Wang RH, Poltyrev T, Gross A, Finberg JP, et al. TV3326, a novel neuroprotective drug with cholinesterase and monoamine oxidase inhibitory activities for the treatment of Alzheimer’s disease. J. Neural Transm. 2000;60:157–169. doi: 10.1007/978-3-7091-6301-6_10. [DOI] [PubMed] [Google Scholar]
  366. Weinstock M, Kirschbaum-Slager N, Lazarovici P, Bejar C, Youdim MBH, Shoham S. Neuroprotective effects of novel cholinesterase inhibitors derived from rasagiline as potential anti- Alzheimer drugs. Ann NY Acad Sci. 2001;939:148–162. doi: 10.1111/j.1749-6632.2001.tb03622.x. [DOI] [PubMed] [Google Scholar]
  367. Weinstock M, Poltyrev T, Bejar C, Youdim MB. Effect of TV3326, a novel monoamineoxidase cholinesterase inhibitor, in rat models of anxiety and depression. Psychopharmacology (Berl.) 2002;160:318–324. doi: 10.1007/s00213-001-0978-x. [DOI] [PubMed] [Google Scholar]
  368. Weinstock M, Gorodetsky E, Poltyrev T, Gross A, Sagi Y, Youdim M. A novel cholinesterase and brain-selective monoamine oxidase inhibitor for the treatment of dementia comorbid with depression and Parkinson’s disease. Prog. Neuropsychopharmacol. Biol. Psychiatry. 2003;27:555–561. doi: 10.1016/S0278-5846(03)00053-8. [DOI] [PubMed] [Google Scholar]
  369. Weinstock M, Luques L, Poltyrev T, Bejar C, Shoham S. Ladostigil prevents age-related glial activation and spatial memory deficits in rats. Neurobiol Aging. 2009 Jul 20; doi: 10.1016/j.neurobiolaging.2009.06.004. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
  370. Wesnes KA, McKeith IG, Ferrara R, Emre M, del Ser T, Spano PF, et al. Effects of rivastigmine on cognitive function in dementia with lewy bodies: a randomised placebo-controlled international study using the cognitive drug research computerised assessment system. Dement Geriatr Cogn Disord. 2002;13:183–192. doi: 10.1159/000048651. [DOI] [PubMed] [Google Scholar]
  371. Wesolowska A, Nikiforuk A. Effects of the brain-penetrant and selective 5-HT6 receptor antagonist SB-399885 in animal models of anxiety and depression. Neuropharmacology. 2007;52:1274–1283. doi: 10.1016/j.neuropharm.2007.01.007. [DOI] [PubMed] [Google Scholar]
  372. Wess J, Eglen RM, Gautam D. Muscarinic acetylcholine receptors: mutant mice provide new insights for drug development. Nature Reviews Drug Discovery. 2007;6:721–733. doi: 10.1038/nrd2379. [DOI] [PubMed] [Google Scholar]
  373. West PJ, Marcy VR, Marino MJ, Schaffhauser H. Activation of the 5-HT6 receptor attenuates long-term potentiation and facilitates GABAergic neurotransmission in rat hippocampus. Neuroscience. 2009;164:692–701. doi: 10.1016/j.neuroscience.2009.07.061. [DOI] [PubMed] [Google Scholar]
  374. Wilens TE, Decker MW. Neuronal nicotinic receptor agonists for the treatment of attention-deficit/hyperactivity disorder: focus on cognition. Biochem Pharmacol. 2007;74:1212–1223. doi: 10.1016/j.bcp.2007.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  375. Wilkinson DG, Francis PT, Schwam E, Payne-Parrish J. Cholinesterase inhibitors used in the treatment of Alzheimer's disease: the relationship between pharmacological effects and clinical efficacy. Drugs Aging. 2004;21(7):453–478. doi: 10.2165/00002512-200421070-00004. [DOI] [PubMed] [Google Scholar]
  376. Williams GV, Rao SG, Goldman-Rakic PS. The physiological role of 5-HT2A receptors in working memory. J Neurosci. 2002;22:2843–2854. doi: 10.1523/JNEUROSCI.22-07-02843.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  377. Winblad B, Engedal K, Soininen H, Verhey F, Waldemar G, Wimo A, et al. A 1-year, randomized, placebo-controlled study of donepezil in patients with mild to moderate AD. Neurology. 2001;57(3):489–495. doi: 10.1212/wnl.57.3.489. [DOI] [PubMed] [Google Scholar]
  378. Winstanley CA, Chudasama Y, Dalley JW, Theobald DEH, Glennon JC, Robbins TW. Intra-prefrontal 8-OH-DPAT and M100907 improve visuospatial attention and decrease impulsivity on the five-choice serial reaction time task in rats. Psychopharmacology. 2003;167:304–314. doi: 10.1007/s00213-003-1398-x. [DOI] [PubMed] [Google Scholar]
  379. Witkin JM, Nelson DL. Selective histamine H3 receptor antagonists for treatment of cognitive deficiencies and other disorders of the central nervous system. Pharmacol Ther. 2004;103:1–20. doi: 10.1016/j.pharmthera.2004.05.001. [DOI] [PubMed] [Google Scholar]
  380. Wolff MC, Overshiner C, Leander JD. 5-Ht6 receptor antagonists enhance memory in radial maze and object recognition tasks. Soc Neurosci Abstr. 2002:586.1. [Google Scholar]
  381. Woolley ML, Bentley JC, Sleight AJ, Mardsen CA, Fone KCF. A role for 5-Ht(6) receptors in retention of spatial learning in the Morris water maze. Neuropharmacology. 2001;41:210–219. doi: 10.1016/s0028-3908(01)00056-9. [DOI] [PubMed] [Google Scholar]
  382. Woolley ML, Marsden CA, Fone KCF. 5-ht6 receptors. Curr Drug Targets CNS Neurol Disord. 2004;3:59–79. doi: 10.2174/1568007043482561. [DOI] [PubMed] [Google Scholar]
  383. World Health Organization. The World is Fast Aging – Have We Noticed? 2008 http://www.who.int/ageing/en/index.html.
  384. Wu J, Li Q, Bezprozvanny I. Evaluation of Dimebon in cellular model of Huntington's disease. Mol Neurodegener. 2008 Oct 21;3:15. doi: 10.1186/1750-1326-3-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  385. Xu AJ, Chen Z, Li R, Zhu CY, Wei EQ. Effects of TAK-147, a novel acetylcholinesterase inhibitor, on spatial memory deficit as evaluated by Morris water maze of rats. Zhejiang Da Xue Xue Bao Yi Xue Ban. 2002a Apr;31(2):98–102. doi: 10.3785/j.issn.1008-9292.2002.02.009. [DOI] [PubMed] [Google Scholar]
  386. Xu AJ, Chen Z, Yanai K, Huang YW, Wei EQ. Effect of 3-[1-(phenylmethyl)-4-piperidinyl]-1-(2,3,4,5-tetrahydro-1H-1-benzazep in-8-yl)-1-propanone fumarate, a novel acetylcholinesterase inhibitor, on spatial cognitive impairment induced by chronic cerebral hypoperfusion in rats. Neurosci Lett. 2002b;331(1):33–36. doi: 10.1016/s0304-3940(02)00830-3. [DOI] [PubMed] [Google Scholar]
  387. Ye JW, Cai JX, Wang LM, Tang XC. Improving effects of huperzine A on spatial working memory in aged monkeys and young adult monkeys with experimental cognitive impairment. J Pharmacol Exp Ther. 1999;288:814–819. [PubMed] [Google Scholar]
  388. Ye JW, Shang YZ, Wang ZM, Tang XC. Huperzine A ameliorates the impaired memory of aged rat in the Morris water maze performance. Acta Pharmacol Sin. 2000;21:65–69. [PubMed] [Google Scholar]
  389. Yesavage JA, Tinklenberg JR, Hollister LE, Berger PA. Vasodilators in senile dementias: a review of the literature. Arch Gen Psychiatry. 1979;36:220–223. doi: 10.1001/archpsyc.1979.01780020110012. [DOI] [PubMed] [Google Scholar]
  390. Yogev-Falach M, Amit T, Bar-Am O, Sagi Y, Weinstock M, Youdim MBH. The involvement of mitogen-activated protein (MAP) kinase in the regulation of amyloid precursor protein processing by novel cholinesterase inhibitors derived from rasagiline. FASEB J. 2002;16(12):1674–1676. doi: 10.1096/fj.02-0198fje. [DOI] [PubMed] [Google Scholar]
  391. Yogev-Falach M, Bar-Am O, Amit T, Weinreb O, Youdim MB. A multifunctional, neuroprotective drug, ladostigil (TV3326), regulates holo-APP translation and processing. FASEB J. 2006;20:2177–2179. doi: 10.1096/fj.05-4910fje. [DOI] [PubMed] [Google Scholar]
  392. Youdim MB, Buccafusco JJ. Multi-functional drugs for various CNS targets in the treatment of neurodegenerative disorders. Trends Pharmacol Sci. 2005 Jan;26(1):27–35. doi: 10.1016/j.tips.2004.11.007. [DOI] [PubMed] [Google Scholar]
  393. Zhang HT, O’Donnell JM. Effects of rolipram on scopolamine-induced impairment of working and reference memory in the radial-arm maze test in rats. Psychopharmacology. 2000a;150:311–316. doi: 10.1007/s002130000414. [DOI] [PubMed] [Google Scholar]
  394. Zhang HT, Crissman AM, Dorairaj NR, Chandler LJ, O’Donnell JM. Inhibition of cyclic AMP phosphodiesterase (PDE4) reverses memory deficits associated with NMDA receptor antagonism. Neuropsychopharmacology. 2000b;23:198–204. doi: 10.1016/S0893-133X(00)00108-1. [DOI] [PubMed] [Google Scholar]
  395. Zhang HT, Zhao Y, Huang Y, Dorairaj NR, Chandler LJ, O’Donnell JM. Inhibition of the phosphodiesterase (PDE4) enzyme reverses memory deficits produced by infusion of the MEK inhibitor U0126 into the CA1 subregion of the rat hippocampus. Neuropsychopharmacology. 2004;29:1432–1439. doi: 10.1038/sj.npp.1300440. [DOI] [PubMed] [Google Scholar]
  396. Zhang HT, Huang Y, Suvarna NU, Deng C, Crissman AM, Hopper AT, et al. Effects of the novel PDE4 inhibitors MEM1018 and MEM 1091 on memory in the radial-arm maze and inhibitory avoidance tests in rats. Psychopharmacology. 2005;179:613–619. doi: 10.1007/s00213-004-2085-2. [DOI] [PubMed] [Google Scholar]
  397. Zhang J, Zhu D, Sheng R, Wu H, Hu Y, Wang F, et al. BZYX, a novel acetylcholinesterase inhibitor, significantly improved chemicals-induced learning and memory impairments on rodents and protected PC12 cells from apoptosis induced by hydrogen peroxide. Eur J Pharmacol. 2009;613(1–3):1–9. doi: 10.1016/j.ejphar.2009.03.054. [DOI] [PubMed] [Google Scholar]
  398. Zhang M, Ballard ME, Pan L, Roberts S, Faghih R, Cowart M, et al. Lack of cataleptogenic potentiation with non-imidazole H3 receptor antagonists reveals potential drug-drug interactions between imidazole-based H3 receptor antagonists and antipsychotic drugs. Brain Res. 2005;1045:142–149. doi: 10.1016/j.brainres.2005.03.018. [DOI] [PubMed] [Google Scholar]
  399. Zhang M-Y, Hughes ZA, Kerns EH, Lin Q, Beyer CE. Development of a liquid chromatography/tandem mass spectrometry method for the quantitation of acetylcholine and related neurotransmitters in brain microdialysis samples. J Pharm Biomed Anal. 2007;44:586–593. doi: 10.1016/j.jpba.2007.02.024. [DOI] [PubMed] [Google Scholar]
  400. Zhang ZJ, Kang WH, Li Q, Wang XY, Yao SM, Ma AQ. Beneficial effects of ondansetron as an adjunct to haloperidol for chronic, treatment-resistant schizophrenia: a double-blind, randomized, placebo-controlled study. Schizophr Res. 2006;88:102–110. doi: 10.1016/j.schres.2006.07.010. [DOI] [PubMed] [Google Scholar]

RESOURCES