Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Mar 1.
Published in final edited form as: Neuropharmacology. 2011 Feb 1;62(3):1564–1573. doi: 10.1016/j.neuropharm.2011.01.044

Schizophrenia and tobacco smoking comorbidity: nAChR agonists in the treatment of schizophrenia-associated cognitive deficits

Manoranjan S D’Souza 1, Athina Markou 1
PMCID: PMC3116036  NIHMSID: NIHMS276442  PMID: 21288470

Abstract

Tobacco smoking is a preventable cause of morbidity and mortality throughout the world. Very high rates of tobacco smoking are seen in patients with schizophrenia. Importantly, smokers with schizophrenia generally have higher nicotine dependence scores, experience more severe withdrawal symptoms upon smoking cessation, have lower cessation rates than healthy individuals, and suffer from significant smoking-related morbidity and premature mortality compared with the general population. Interestingly, significant disturbances in cholinergic function are reported in schizophrenia patients. The high smoking-schizophrenia comorbidity observed in schizophrenia patients may be an attempt to compensate for this cholinergic dysfunction. Cholinergic neurotransmission plays an important role in cognition and is hypothesized to play an important role in schizophrenia-associated cognitive deficits. In this review, preclinical evidence highlighting the beneficial effects of nicotine and subtype-selective nicotinic receptor agonists in schizophrenia-associated cognitive deficits, such as working memory and attention, is discussed. Furthermore, some of the challenges involved in the development of procognitive medications, particularly subtype-selective nicotinic receptor agonists, are also discussed. Amelioration of schizophrenia-associated cognitive deficits may help in the treatment of schizophrenia-smoking comorbidity by promoting smoking cessation and thus help in the better management of schizophrenia patients.

1. Introduction

Tobacco dependence in the form of cigarette smoking is often associated with psychiatric comorbidity. Individuals with a psychiatric disorder consume approximately 46% of all cigarettes smoked in the United States (Grant et al., 2004). Exceptionally high smoking rates (60–90%) are reported in patients suffering from schizophrenia (Chapman et al., 2009; de Leon et al., 1995; de Leon and Diaz, 2005; Dome et al., 2010; Hughes et al., 1986; Leonard et al., 2001). Schizophrenia patients also have a higher risk of being current smokers compared with people from the general population (Dome et al., 2010).

Smokers with schizophrenia can be classified as “heavy smokers” and take a greater number of puffs, have shorter interpuff intervals, and consume larger total cigarette puff volumes compared with smokers in the general population (Tidey et al., 2005). Furthermore, smokers with schizophrenia had higher blood and saliva nicotine levels compared with control smokers matched for cigarette smoking (Williams et al., 2005). Additionally, smokers with schizophrenia reported greater reinforcing effects from smoking, making them more vulnerable to developing nicotine dependence (Spring et al., 2003). Importantly, withdrawal symptoms upon abstinence from smoking were generally more severe in schizophrenia patients than smokers without schizophrenia (Weinberger et al., 2007). Thus, smoking cessation in these patients poses a significant challenge compared with smokers without schizophrenia. Finally, heavy smoking habits and the inability to abstain from smoking often lead to significant cardiovascular and pulmonary morbidity, resulting in premature deaths in schizophrenia patients (Hennekens, 2007; Shanmugam et al., 2007; de Leon and Diaz, 2005).

To promote smoking cessation in schizophrenia patients, understanding some of the possible reasons for high smoking-schizophrenia comorbidity is critical. In this article, we focus on cholinergic neurotransmission dysfunction reported in schizophrenia patients and its possible role in mediating some of the schizophrenia-associated cognitive deficits. Additionally, some of the procognitive effects of nicotine and subtype-selective nicotinic acetylcholine receptor (nAChR) agonists important for the treatment of schizophrenia-associated cognitive deficits are discussed.

2. Theories explaining the high prevalence of smoking among schizophrenia patients

Several hypotheses have been offered to explain the high prevalence of smoking in patients with schizophrenia. A popular hypothesis is the “self-medication hypothesis,” in which smoking may help schizophrenia patients manage their symptoms, such as positive symptoms, negative symptoms, and cognitive symptoms, by compensating for the underlying neurobiological deficits associated with the disorder (Adler et al., 1998b; Dome et al., 2010; Khantzian, 1985; Leonard et al., 2001; Leonard et al., 1998; Markou et al., 1998). An additional or complementary hypothesis is that the high comorbidity may be attributable to the hypothesized beneficial effects of smoking on some of the aversive effects of psychiatric medications (Barr et al., 2008a). For example, smoking is believed to relieve some of the extrapyramidal symptoms caused by classical antipsychotic treatments. Finally, some studies suggest that smoking and psychiatric disorders may have common risk factors (Chambers et al., 2001). The discussion of the merits and evidence supporting each of these hypotheses is beyond the scope of this review. In this review, we tacitly support the “self-medication hypothesis” by implicating deficits in cholinergic neurotransmission (discussed below) reported in schizophrenia patients as a possible cause for the high incidence of smoking-schizophrenia comorbidity.

3. Dysfunction in cholinergic neurotransmission in schizophrenia: Evidence from clinical and preclinical studies

Postmortem brain analysis of patients with a history of schizophrenia reported elevated levels of choline acetyltransferase, an enzyme involved in the synthesis of acetylcholine, in several brain areas, such as the hippocampus, caudate, putamen, thalamus, and septal areas, compared with controls (McGeer and McGeer, 1977). In contrast, decreased levels of choline acetyltransferase were reported in the nucleus accumbens and pons in schizophrenia patients (Bird et al., 1977; Karson et al., 1993). Furthermore, a postmortem histochemical analysis reported an increase in the number of cholinergic neurons in the pedunculopontine nucleus of the brainstem in patients with a history of schizophrenia compared with controls (Garcia-Rill et al., 1995). Finally, studies using magnetic resonance spectroscopy revealed increased choline levels in several brain regions, such as the thalamus, anterior cingulate, and caudate nucleus in antipsychotic-naive patients diagnosed with schizophrenia compared with matched healthy volunteers (Bustillo et al., 2002; Theberge et al., 2004). Although the status of schizophrenia patients with respect to smoking was not reported in the aforementioned postmortem and clinical studies, data from a majority of studies support increased acetylcholine levels in the brains of schizophrenia patients. These increased acetylcholine levels are hypothesized to compensate for nAChR dysfunction.

Indeed, disturbances in nAChR expression and function have also been reported after postmortem evaluations of brains of schizophrenia patients compared with brains of subjects without schizophrenia. Decreased expression of α7 nAChRs was found in several brain regions, such as the hippocampus, reticular nucleus of the thalamus, and prefrontal cortex in schizophrenia patients (Court et al., 1999; Freedman et al., 1995; Guan et al., 1999; Olincy and Stevens, 2007). Furthermore, autoradiographic studies indicated decreased α4β2 nAChR binding in the hippocampus, cortex, and striatum in schizophrenia patients compared with controls (Breese et al., 2000; Durany et al., 2000). Finally, autoantibodies against nAChRs have been found in patients with schizophrenia (Chandley et al., 2009; Margutti et al., 2006).

In addition to changes in the expression or function of nAChRs, evidence from genetic studies links schizophrenia with nAChR genes. Schizophrenia-associated polymorphisms in the neuregulin-1 gene were associated with decreased α7 nAChR binding and decreased α7 nAChR mRNA levels in the prefrontal cortex (Mathew et al., 2007). Furthermore, schizophrenia-associated sensory motor gating deficits, measured by P50 evoked potentials for auditory stimuli, have been linked to α7 subunit gene (CHRNA7) polymorphisms (Leonard et al., 1998). Altogether, the above studies suggest decreased cholinergic neurotransmission in schizophrenia patients.

Disturbances in cholinergic neurotransmission are also observed in putative animal models of schizophrenia that involve administration of N-methyl-D-aspartate (NMDA) receptor antagonists, such as ketamine and phencyclidine (PCP), in the induction of schizophrenia-like behavioral abnormalities. Interestingly, acute and repeated administration of the NMDA receptor antagonists ketamine and PCP resulted in increased release of acetylcholine in several forebrain regions (Kim et al., 1999; Nelson et al., 2002; Sato et al., 1996). In mice, PCP administration was associated with decreased α7 nAChR expression (Hashimoto et al., 2008).

Interestingly, antipsychotics have anticholinergic properties and affect nAChR expression (Grinevich et al., 2009; Levin and Rezvani, 2007; Singhal et al., 2007; Terry et al., 2003). In unperturbed healthy rats, long-term administration of typical antipsychotic medications, such as haloperidol and chlorpromazine, and atypical antipsychotics, such as risperidone and olanzapine, resulted in decreased density of α7 nAChRs in the hippocampus, frontal cortex, and basal forebrain (Terry et al., 2005). In contrast, long-term administration of typical (haloperidol and chlorpromazine) and atypical (risperidone and olanzapine) antipsychotics in rats did not affect α4β2 nAChR density in the hippocampus (Terry et al., 2006). Finally, long-term administration of risperidone and haloperidol decreased choline acetyltransferase activity in the hippocampus and striatum. In contrast, long-term administration of clozapine and olanzapine did not affect choline acetyltransferase activity in the striatum and hippocampus. Altogether, the above preclinical data suggest that antipsychotics possibly worsen already decreased cholinergic neurotransmission in schizophrenia patients.

4. Cholinergic system and cognition

Schizophrenia is a complex, chronic multisymptom disorder associated with positive, negative, and cognitive symptoms that result in severe disability and often lead to social isolation and economic instability. Although disturbances in cholinergic transmission are seen in schizophrenia, it Is not clear what role this dysfunction plays in the pathophysiology of schizophrenia.

Optimal functioning of the cholinergic system is essential for normal cognitive function. The frontal cortex and hippocampus are two brain regions critically involved in cognitive function, and these two brain areas are tightly regulated by extensive cholinergic projections from several forebrain regions (Hasselmo and Sarter, 2010; Lysakowski et al., 1989; Mesulam, 2004; Rye et al., 1984; Sarter et al., 2005). Disruption of cholinergic system function, both in humans and animals via administration of cholinergic receptor blockers, resulted in cognitive impairment in tasks involving working memory and attention (Levin, 1988; Liljequist and Mattila, 1979; Nuotto, 1983). Furthermore, forebrain cholinergic neurons are activated in animals during the performance of tasks that assess attention and working memory (Masuda et al., 1997; Pepeu and Giovannini, 2004). Finally, pharmacological enhancement of cholinergic neurotransmission facilitated working memory in both humans and animals (Baratti et al., 1979; Cox and Tye, 1973; Davis and Mohs, 1982; Drachman, 1977). These studies highlight the critical role of cholinergic neurotransmission in cognitive function, including memory and attention.

5. Schizophrenia-associated cognitive impairment and smoking

Cognitive impairments are present in the majority of schizophrenia patients. However, the degree of severity of schizophrenia-associated cognitive deficits varies between individuals (Gold and Harvey, 1993). Importantly, these cognitive deficits often lead to an inability to perform day-to-day functions. Schizophrenia-associated cognitive deficits affect several domains, such as verbal learning and memory, attention, speed of processing, problem solving, visual learning and memory, social cognition, and working memory (Nuechterlein et al., 2004). Schizophrenia-associated cognitive deficits are a major problem because current schizophrenia treatments mainly target the positive symptoms (classical and atypical antipsychotics) and negative symptoms (atypical antipsychotics) with either no or limited procognitive effects (Velligan and Miller, 1999). Unfortunately, classical antipsychotics are known to worsen schizophrenia-associated cognitive deficits (Kasper and Resinger, 2003). Treatment of schizophrenia-associated cognitive deficits is critical because, as mentioned above, these deficits correlate greatly with day-to-day functioning, and attenuation or reversal of these cognitive deficits may improve functional outcomes and social rehabilitation in schizophrenia patients.

Interestingly, nicotine administration in both smoking and nonsmoking patients with schizophrenia resulted in the alleviation of cognitive deficits. For example, nicotine administration via nasal spray resulted in improvements in spatial organization and memory in patients suffering from schizophrenia (Barr et al., 2008b; Sacco et al., 2005; Smith et al., 2006). Furthermore, administration of nicotine in the form of nicotine gum or transdermal patch improved attention in schizophrenia patients (Barr et al., 2008b; Harris et al., 2004). Finally, abstinence from smoking in smokers with schizophrenia worsened deficits in spatial working memory and attention, which were alleviated by smoking (Sacco et al., 2005). Furthermore, alleviation of the aforementioned cognitive deficits upon resumption of smoking was blocked by administration of the nAChR antagonist mecamylamine (Sacco et al., 2005). The above studies suggest that tobacco smoking has beneficial procognitive effects in schizophrenia patients.

In summary, deficits in cholinergic function seen in schizophrenia patients may play a critical role in mediating impaired working memory, spatial organization, and attention in schizophrenia patients. Furthermore, tobacco smoking or nicotine administration alleviated schizophrenia-associated deficits in attention and memory. Thus, treatment of schizophrenia-associated cognitive deficits may promote smoking cessation in schizophrenia patients and significantly improve the health of schizophrenia patients.

4. Effects of nicotine and subtype-selective nAChR agonists on cognitive function

As discussed above, several cognitive domains are impaired in schizophrenia. Because of the limited literature on the beneficial effects of nicotine on cognitive domains other than attention and memory, this review is limited to preclinical studies that evaluated the effects of nicotine and subtype-selective nAChR agonists on attention and working memory in both unperturbed animals and putative animal models of schizophrenia. The available clinical studies that support a role of nicotine and subtype-selective nAChR agonists in attention and memory tasks are presented. Preclinical tasks used to assess the beneficial effects of nicotine and subtype-selective nAChR agonists on attention and memory are briefly described in Table 1.

Table 1.

Preclinical tasks assessing memory and attention.

Cognitive function Task Description Reference
Memory Radial arm maze The radial arm maze consisted of an octagonal central chamber with eight attached arms. Each of the arms was baited with food, and the animal was expected to retrieve the reward (food) from each arm without revisiting a previously visited arm. Spatial working memory was measured by the number of baited arms entered prior to entering a previously visited arm. Chronic nicotine administration using subcutaneously implanted glass and silastic pellets (12 mg/kg salt) improved working memory 1 week after pellet implantation compared with saline controls. Subjects were female Sprague Dawley rats. Levin et al., 1990
Morris water maze task In this task, animals were placed in the Morris water maze at different starting locations during multiple trials. The animals were expected to escape from drowning by locating a platform submerged under the water. The average distance traveled by the animal to locate the submerged platform from various start locations over multiple trials was used as a measure of working memory. The α7 agonist ABBF (1 mg/kg, per oral [p.o.]) improved working memory in aged rats in the Morris water maze task. Boess et al., 2007
Novel object recognition task This task was performed over two trials separated by 24 h. In the first trial, the subject was allowed to explore two objects. The second trial, conducted 24 h after the first trial, consisted of presentation of an object from the previous trial and a novel object. The discrimination index, defined as the difference in time spent exploring the novel object minus the time spent exploring the familiar object divided by the total time spent exploring the two objects, was used to assess novel object recognition memory. The α7 agonist ABBF (0.3–1 mg/kg, p.o.) improved object recognition memory in mice. Boess et al., 2007
Social memory task This task was performed over two trials separated by 24 h. On day 1, the adult rat being assessed was presented with a juvenile rat for a period of 2 min. Social investigation in the form of sniffing, grooming of body parts, anogenital sniffing, and close following was scored over the period of 2 min to build a composite social investigation score. On the following day (i.e., after 24 h), the adult rat was again presented with the same juvenile rat. A decreased social investigation score was indicative of social memory. The α7 agonist ABBF (0.3–1 mg/kg, p.o.) improved social recognition memory in rats. Boess et al., 2007
Delayed matching-to- sample task This computerized task was designed to assess memory in nonhuman primates. The task involved presentation of a stimulus of a specific color on the computer screen. The subject was allowed to respond after a delay of up to120 s by pressing one of two illuminated keys. One of the two illuminated keys was of the same color as the stimulus presented on the screen. Selection of the key matching the color of the stimulus presented was considered a correct response. The percentage of correct responses was used as a measure of attention. The percentage of correct responses after the longest delay was significantly greater after nicotine (7.5 μg/kg, intramuscular injection) administration in aged monkeys compared with saline controls. Buccafusco and Jackson, 1991
Attention 5-Choice serial reaction time task The experimental subject was expected to monitor a light stimulus that was presented in one of the five equidistant located apertures. The animal signaled the detection of the light stimulus by nosepoking in the aperture where the light stimulus appeared. Correct responses were rewarded, and incorrect responses were punished with a timeout period. Accuracy, a measure of attention, was defined as the total number of correct responses divided by the total number of correct and incorrect responses. Chronic nicotine administration (9 mg/kg/day, salt) via minipumps improved accuracy 4–6 days after minipump implantation compared with saline controls in Wistar rats. Semenova et al., 2007
Sustained attention task In this task, experimental animals were required to detect a light “signal” that varied in intensity. In a given trial, the signal may or may not have been presented prior to presentation of two levers in the chamber. The animal was expected to respond by responding on one of the two levers, and the correct lever was determined based on where the light signal was presented prior to the introduction of the levers in the chamber. Attention was assessed by determining the correct number of hits (i.e., responding on the correct lever when the signal was presented, defined as the total number of correct detections/total number of signal trials) or false alarms (i.e., animal incorrectly pressed the signal lever when no signal had been presented; number of false alarms/number of blank trials). Low doses of nicotine (0.0125, 0.025, and 0.05 mg/kg, salt; administered subcutaneously) dose-dependently increased the percent correct rejection in adult female Sprague Dawley rats. The same nicotine doses did not affect correct detections of the signal (i.e., percent hits). Rezvani et al., 2002

4.1. Effects of nicotine on memory and attention

Both acute and chronic nicotine administration enhanced working memory in the radial-arm maze in young adult and aged rats (Levin et al., 1993; Levin et al., 1990; Levin and Rose, 1990, 1991; Levin and Torry, 1996). The ventral hippocampus plays a critical role in the memory-enhancing effects of chronic nicotine. Ibotenic acid-induced lesion of the ventral hippocampus attenuated the memory-enhancing effect of chronic nicotine (Levin et al., 1999). Furthermore, nicotine administration enhanced working memory in delayed matching-to-sample tasks in aged nonhuman primates (Buccafusco and Jackson, 1991). Importantly, nicotine administration reversed the memory-impairing effects of some antipsychotic medications, such as clozapine and olanzapine, in the radial arm maze task in rats (Addy and Levin, 2002; Levin et al., 2005). Nicotine has also been shown to attenuate NMDA antagonist-induced impairments in working memory in the radial-maze task (Levin et al., 1998).

N-methyl-D-aspartate receptor dysfunction is hypothesized to be a key player in mediating the pathophysiology of schizophrenia, particularly schizophrenia-associated cognitive deficits (Goff and Coyle, 2001; Konradi and Heckers, 2003). Indeed, administration of the NMDA receptor antagonists PCP and ketamine produced a schizophrenia-like state in healthy humans and worsened existing symptoms in schizophrenia patients (Allen and Young, 1978; Bubenikova-Valesova et al., 2008; Malhotra et al., 1997; Steinpreis, 1996). Importantly, NMDA receptor antagonists, such as PCP and ketamine, profoundly disrupted cognitive function in both healthy subjects and schizophrenia patients (Krystal et al., 2000; Krystal et al., 1994; Malhotra et al., 1996), including disruptions in attention and working memory (Adler et al., 1998a; Krystal et al., 1994; Malhotra et al., 1996; Morgan et al., 2004). Consistent with this finding, NMDA receptor antagonists also disrupted performance in attention and working memory tasks in experimental animals (Amitai et al., 2007; Greco et al., 2005; Handelmann et al., 1987; Kesner and Dakis, 1993; Rezvani et al., 2008). Thus, NMDA receptor antagonist administration is often used as an inducing condition to model schizophrenia-like cognitive deficits in humans and animals (e.g., Amitai and Markou, 2010).

Nicotine also enhanced attention in several experimental animal procedures that assess attention. The 5-choice serial reaction time task (5-CSRTT) is extensively used to assess attention in animals (for review, see Chudasama and Robbins, 2004; Robbins, 2002; Young et al., 2009). The enhancement of attention by acute nicotine in unperturbed rats and mice, measured in the 5-CSRTT, is not very robust and can be best described as inconsistent (Blondel et al., 2000; Grottick and Higgins, 2000; Mirza and Bright, 2001; Muir et al., 1995; Semenova et al., 2007; Young et al., 2004). Importantly, however, the attention-enhancing effects of acute nicotine in the 5-CSRTT in unperturbed rats have been reported under challenge conditions, such as reducing the length of the visual stimulus, decreasing the intertrial interval, and other similar manipulations that increase attentional load (Hahn et al., 2002; Stolerman et al., 2000). In contrast to findings in unperturbed rats, acute nicotine administration enhanced performance in the 5-CSRTT in rats with lesions of cholinergic neurons in the basal nucleus of Meynert (Muir et al., 1995). Attention in rodents has also been evaluated using the sustained attention task, which is also sometimes referred to as the lateralized visual signal detection task (Bushnell, 1998; McGaughy and Sarter, 1995). The effects of acute nicotine on attention in this task in rats have also been mixed, with some studies reporting beneficial effects (Grilly, 2000; Rezvani et al., 2002) and others reporting no effects of nicotine (Bushnell et al., 1997; Turchi et al., 1995).

Several hypotheses have been proposed to explain the inconsistent effects of nicotine on attention in unperturbed animals. One hypothesis is that the cholinergic system is maximally activated in unperturbed animals. Therefore, nicotine administration cannot further improve performance (Turchi et al., 1995). Another hypothesis states that acute nicotine can produce dysphoric effects that may interfere with performance on attentional tasks (Heishman et al., 1993). Furthermore, repeated administration of nicotine is hypothesized to result in the development of tolerance to these dysphoric effects and consequently improve performance on attentional tasks. Indeed, chronic nicotine administration via subcutaneous osmotic minipumps in rats increased attention in the 5-CSRTT (Semenova et al., 2007). An earlier study also reported beneficial effects of chronic nicotine in rats in a go/no go task that had an attentional component (Nelsen and Goldstein, 1972). In summary, although chronic nicotine improved attentional performance, the effects of acute nicotine on attention are inconsistent, with studies reporting either pro-attentional or no effects on attention. Clearly, further work is required to fully understand the effects of nicotine on attentional processes.

Very few studies have assessed the effects of nicotine in putative animal models of schizophrenia. Chronic nicotine treatment did not improve PCP-induced cognitive deficits in the 5-CSRTT in rats (Amitai and Markou, 2009). In contrast, nicotine attenuated memory and attentional impairments induced by another NMDA receptor antagonist, dizocilpine, in the lateralized visual signal detection task in rats (Rezvani et al., 2008). The differences in the effects of nicotine on attention in these two studies could be attributable to subtle differences in the pharmacological profiles of the two NMDA receptor antagonists, phencyclidine and dizocilpine, that induce cognitive deficits in animals that resemble those seen in schizophrenia. Alternatively, the differential effects of nicotine could also be attributable to the cognitive tasks (5-CSRTT vs. lateralized visual detection task) used in the two studies. Both tasks assess attention, but the lateralized visual detection task may be more able to detect the proattention effects of nicotine than the 5-CSRTT. In summary, the above data suggest that acute and chronic nicotine enhanced attention in both unperturbed animals and putative animal models of schizophrenia.

4.2. Effects of subtype-selective nicotinic receptor agonists on memory and attention

α4β2 and α7 nAChRs play a critical role in mediating the memory-enhancing effect of nicotine. Nicotine-induced memory enhancement in the radial-arm maze task was blocked by the nAChR antagonists dihydro-β-erythroidine and methyllycaconitine, which exhibit relative selectivity for these two receptors, respectively (Levin et al., 2002). Furthermore, the α7 nAChR agonists N-[(3R)-1-azabicyclo[2.2.2]oct-3-yl]-7-[2-(methoxy)phenyl]-1-benzofuran-2-carboxamide (ABBF) and GTS-21 enhanced working memory in the radial arm maze and the Morris water maze in aged rats (Arendash et al., 1995; Boess et al., 2007).

In addition to working memory, several α7 and α4β2 nAChR ligands have shown procognitive effects in recognition-based memory tasks, such as the social recognition memory and novel object recognition tasks. For example, administration of the α7 nAChR agonists ABBF and AR-R17779 enhanced novel object and social recognition memory in unperturbed rats (Boess et al., 2007; Van Kampen et al., 2004). The α7 nAChR agonists ABT-089, GTS-21, and A-582941 also improved working memory in the delayed matching-to-sample task in nonhuman primates (Briggs et al., 1997; Buccafusco et al., 2007). In a transgenic th(tk-)/th(tk-) mouse model that shares many of the anatomical and biochemical dysfunctions seen in schizophrenia, administration of the α7 nAChR agonist TC-5619 improved novel object recognition memory (Hauser et al., 2009). Finally, the α4β2 receptor agonist varenicline enhanced memory in a novel object recognition task in unperturbed rats (Rollema et al., 2009).

The role of α7 nAChRs in attention is debatable because opposite findings have been reported in pharmacological and genetic studies. For example, the selective α7 nAChR agonist AR-R17779 did not have beneficial effects on attention in the 5-CSRTT (Grottick and Higgins, 2000; Hahn et al., 2003), suggesting that α7 nAChRs have possibly no role or a limited role in attention. In contrast, impairments in attention have been reported in α7 nAChR knockout mice in the 5-CSRTT (Hoyle et al., 2006; Young et al., 2007; Young et al., 2004). However, compensatory neurobiological mechanisms occurring in α7 nAChR knockout mice could be responsible for some of the attention-impairing effects in these mice. Additionally, differences in species (rats vs. mice) could also contribute to the different results between these two studies. Altogether, these findings suggest that α7 nAChRs may be required for attention, but activation of these receptors in unperturbed animals by agonists may not enhance attention beyond a certain optimal level. Importantly, α7 nAChRs are permeable to calcium and play an important role as heteroreceptors that modulate the release of several neurotransmitters (Dani, 2001). A small proof-of-concept trial showed beneficial effects of the α7 nAChR partial agonist 3-[(2,4-dimethoxy)benzylidene]anabaseine (DMXB-A) on attention in nonsmoking patients with schizophrenia (Olincy et al., 2006). However, a larger clinical study in nonsmoking schizophrenia patients did not show beneficial effects of the same α7 nAChR partial agonist (Freedman et al., 2008). The effects of α7 nAChR agonists on attention need further evaluation in putative animal models of schizophrenia and smoking and nonsmoking schizophrenia patients.

α4β2 nAChRs are also reported to play a role in attention, and ligands activating these receptors have shown beneficial effects on attention. Administration of the α4β2 nAChR agonist ABT-418 enhanced attention in the 5-CSRTT in unperturbed rats (Hahn et al., 2003). In clinical studies, repeated administration of the α4β2-selective agonist TC-1734 improved attention in healthy adults compared with placebo (Dunbar et al., 2007). Two small proof-of-concept clinical studies demonstrated improvement in attention after administration of the α4β2-selective agonist ABT-089 and ABT-418 in adults with attention deficit disorder (Wilens et al., 1999; Wilens et al., 2006). Furthermore, selective α4β2 receptor agonists may be hypothesized to have superior pro-attentional efficacy compared with nicotine (Howe et al., 2010). Further studies that evaluate α4β2 nAChRs in animal models of schizophrenia and schizophrenia patients are warranted.

In summary, nAChR agonists, especially those acting at α4β2 and α7 nAChRs, have great potential to alleviate attentional and working memory deficits seen in schizophrenia. Currently, very few studies have evaluated compounds acting on these receptors in animal models of schizophrenia or schizophrenia patients. More importantly, these compounds need to be evaluated in smokers with schizophrenia. Several clinical studies are currently ongoing that assess the procognitive effects of α4β2 and α7 nAChR agonists in schizophrenia patients (http://www.clinicaltrials.gov).

5. nAChR agonists as procognitive medications for schizophrenia-associated cognitive dysfunction: Potential challenges and future directions

The use of nicotine for the treatment of schizophrenia-associated cognitive deficits is limited by its aversive effects seen at high doses and tachyphylaxis upon prolonged treatment (Buchanan et al., 2007). Recently, several subtype-selective positive allosteric modulators targeting the different nAChRs have been identified (Taly et al., 2009). These subtype-selective nAChR positive modulators/agonists may provide more tolerable and efficacious alternatives to nicotine (Taly et al., 2009). However, several challenges exist in the discovery and development of nAChR agonists as procognitive medications for schizophrenia-associated cognitive deficits. Cognition is a broad term that encompasses several domains. To perform tasks involved with daily living, either all domains or a combination of certain domains is required. A major challenge for the development of nAChR agonists as procognitive agents is identifying agents that are effective in improving several cognitive domains. Evidence currently suggests that subtype-selective nAChR agonists may enhance certain domains, while not being effective in others. Specifically, preclinical evidence suggests that α4β2 and α7 nAChR agonists have beneficial effects on memory and attention in preclinical animal models and clinical studies. Thus, compounds that positively target a large number of cognitive domains need to be identified. Interestingly, nAChRs undergo desensitization upon activation. Thus, activation of nAChRs may have very short-lasting effects. In this respect, identifying compounds that activate nAChRs without desensitization may prove to be extremely useful in the clinical development of these nAChR agonists as procognitive compounds. Finally, preclinical studies have shown that repeated administration of nicotine upregulates nAChRs (Flores et al., 1992; Schwartz and Kellar, 1983; Yates et al., 1995). Therefore, determining the effects of chronic administration of nAChR agonists on receptor expression and function and how these changes might alter the beneficial effects of these compounds on cognition is important.

Furthermore, novel nAChR-based cognitive enhancers may have interactions with currently used and under-development antipsychotic medications, resulting in unanticipated adverse effects in schizophrenia patients. Identifying possible interactions between nAChR compounds and antipsychotic medications using preclinical animal models will help limit the adverse effects or diminished efficacy resulting from the combined administration of nAChR-based procognitive medications and antipsychotic drugs. One way to overcome the problem of antipsychotic-nAChR interactions is to look for dual-use medications, such as nAChR-based cognitive enhancers with potential antipsychotic activity or antipsychotic compounds that will have potential procognitive effects. Recently, several allosteric modulators of muscarinic cholinergic receptors have been identified that have antipsychotic-like activity in preclinical animal models and procognitive properties (Bridges et al., 2010; Jones et al., 2008; Maehara et al., 2008). Muscarinic cholinergic receptors also play an important role in cognition and may be used as potential treatments for schizophrenia-associated cognitive deficits (Sellin et al., 2008), but the present review is limited to the discussion of nAChRs. Finally, interactions between nAChR agonists and medications other than antipsychotic treatments that schizophrenia patients may be taking need to be determined. For example, a preclinical study reported a possible interaction between nicotine and the α2 adrenergic receptor antagonist idazoxan. Specifically, repeated administration of nicotine and idazoxan in rats with dizocilpine-induced cognitive deficits resulted in the development of seizures in some rats (Timofeeva and Levin, 2008). Although idazoxan is not used in humans, α2 adrenergic receptor antagonists are used in the treatment of hypertension in humans. Whether the interaction between nicotine and idazoxan was specific to idazoxan or would be seen with other α2 adrenergic receptor antagonists remains to be determined.

Additionally, some general challenges exist in the discovery of procognitive medications. One of these challenges is the lack of translational tests across species to assess the procognitive benefits of compounds (Markou et al., 2009; Young et al., 2009). The development of procognitive medications is also hampered by the lack of animal models of cognitive deficits that have predictive validity (Markou et al., 2009). The establishment of predictive validity in terms of pharmacological isomorphism (Geyer and Markou, 1995) is hampered by the lack of an effective treatment for schizophrenia-associated cognitive deficits as a positive control. To improve the identification and development of new procognitive medications, the United States National Institute of Mental Health has so sponsored three initiatives, namely the Measurement and Treatment Research to Improve Cognition in Schizophrenia (MATRICS), Cognitive Neuroscience Treatment Research to Improve Cognition in Schizophrenia (CNTRICS), and Treatment Units for Research on Neurocognition and Schizophrenia (TURNS) (Buchanan et al., 2007; Carter et al., 2008; Marder and Fenton, 2004). These initiatives have attempted to bring together academic, industrial, and government bodies to help in the development of new medications for the treatment of schizophrenia-associated cognitive deficits. Efforts are increasingly underway to develop tests for assessing cognitive function across species. These efforts may eventually help identify novel medications that target schizophrenia-associated cognitive deficits. Such medications may also ultimately help reduce schizophrenia-smoking comorbidity and improve the overall health of schizophrenia patients.

6. Negative symptoms in schizophrenia and the beneficial effects of nicotine

This review has largely focused on the beneficial role of tobacco smoking in schizophrenia-associated cognitive deficits. However, schizophrenia patients also suffer from significant negative symptoms, such as decreased motivation, anhedonia, altered expression of emotions, and a decreased amount or content of speech. The reinforcing effects of nicotine are well documented in both humans and animals (Corrigall and Coen, 1989; Goldberg and Henningfield, 1988). Therefore, high levels of smoking in schizophrenia patients could be an attempt to ameliorate the negative symptoms associated with schizophrenia. Supporting this hypothesis, a recent clinical study involving the α7 receptor agonist DMXB-A showed considerable improvement in anhedonia and alogia (i.e., poverty of speech) in patients with schizophrenia (Freedman et al., 2008). However, a complete review of the beneficial effects of nicotine and nAChRs on the negative symptoms of schizophrenia is beyond the scope of this review.

7. Conclusion

A high degree of comorbidity exists between smoking and schizophrenia. Over the years, research has highlighted significant disturbances in the expression of nAChRs, especially α7 and α4β2 nAChRs, in both schizophrenia patients and putative animal models of schizophrenia. Furthermore, the data suggest that administration of nicotine or nAChR subtype-selective agonists results in beneficial cognitive effects on attention and working memory in preclinical animal models. Because of the limitations of chronic tobacco use, α7 and α4β2 nAChR-based strategies are currently being evaluated as possible therapies for schizophrenia-associated cognitive deficits. Finally, although this review has focused primarily on nAChR-based strategies for the amelioration of schizophrenia-associated cognitive deficits, it must be understood that nAChRs are widely distributed as heteroreceptors on presynaptic nerve terminals and can influence the release of several different neurotransmitters.

In conclusion, addressing the problem of tobacco smoking in smokers with schizophrenia is critical for decreasing smoking-associated morbidity in schizophrenia patients. The treatment of symptoms not alleviated by antipsychotics, such as cognitive and negative symptoms, may be useful in promoting abstinence in schizophrenia patients. More work is clearly required to identify the neurobiological substrates mediating schizophrenia-smoking comorbidity. A better understanding of this comorbidity will help provide alternative and better treatments for smoking cessation, schizophrenia, and the comorbidity of these two disorders.

Acknowledgments

This work was supported by NIH research grants 2R01MH62527, 1R01DA11946, 2R01DA232090, and 2U19DA026838 to AM. MSD was supported by fellowship 19FT-0045 from the Tobacco-Related Disease Research Program (TRDRP) of the State of California. The authors would like to thank Mr. Michael Arends for outstanding editorial assistance.

Abbreviations

nAChR

nicotinic acetylcholine receptor

NMDA

N-methyl-D-aspartate

PCP

phencyclidine

5-CSRTT

5-choice serial reaction time task

ABBF

N-[(3R)-1-azabicyclo[2.2.2]oct-3-yl]-7-[2-(methoxy)phenyl]-1-benzofuran-2-carboxamide

DMXB-A

3-[(2,4-dimethoxy)benzylidene]anabaseine

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. Addy N, Levin ED. Nicotine interactions with haloperidol, clozapine and risperidone and working memory function in rats. Neuropsychopharmacology. 2002;27:534–541. doi: 10.1016/S0893-133X(02)00327-5. [DOI] [PubMed] [Google Scholar]
  2. Adler CM, Goldberg TE, Malhotra AK, Pickar D, Breier A. Effects of ketamine on thought disorder, working memory, and semantic memory in healthy volunteers. Biol Psychiatry. 1998a;43:811–816. doi: 10.1016/s0006-3223(97)00556-8. [DOI] [PubMed] [Google Scholar]
  3. Adler LE, Olincy A, Waldo M, Harris JG, Griffith J, Stevens K, Flach K, Nagamoto H, Bickford P, Leonard S, Freedman R. Schizophrenia, sensory gating, and nicotinic receptors. Schizophr Bull. 1998b;24:189–202. doi: 10.1093/oxfordjournals.schbul.a033320. [DOI] [PubMed] [Google Scholar]
  4. Allen RM, Young SJ. Phencyclidine-induced psychosis. Am J Psychiatry. 1978;135:1081–1084. doi: 10.1176/ajp.135.9.1081. [DOI] [PubMed] [Google Scholar]
  5. Amitai N, Markou A. Chronic nicotine improves cognitive performance in a test of attention but does not attenuate cognitive disruption induced by repeated phencyclidine administration. Psychopharmacology (Berl) 2009;202:275–286. doi: 10.1007/s00213-008-1246-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Amitai N, Markou A. Disruption of performance in the five-choice serial reaction time task induced by administration of N-methyl-D-aspartate receptor antagonists: relevance to cognitive dysfunction in schizophrenia. Biol Psychiatry. 2010;68:5–16. doi: 10.1016/j.biopsych.2010.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Amitai N, Semenova S, Markou A. Cognitive-disruptive effects of the psychotomimetic phencyclidine and attenuation by atypical antipsychotic medications in rats. Psychopharmacology (Berl) 2007;193:521–537. doi: 10.1007/s00213-007-0808-x. [DOI] [PubMed] [Google Scholar]
  8. Arendash GW, Sengstock GJ, Sanberg PR, Kem WR. Improved learning and memory in aged rats with chronic administration of the nicotinic receptor agonist GTS-21. Brain Res. 1995;674:252–259. doi: 10.1016/0006-8993(94)01449-r. [DOI] [PubMed] [Google Scholar]
  9. Baratti CM, Huygens P, Mino J, Merlo A, Gardella J. Memory facilitation with posttrial injection of oxotremorine and physostigmine in mice. Psychopharmacology (Berl) 1979;64:85–88. doi: 10.1007/BF00427350. [DOI] [PubMed] [Google Scholar]
  10. Barr AM, Procyshyn RM, Hui P, Johnson JL, Honer WG. Self-reported motivation to smoke in schizophrenia is related to antipsychotic drug treatment. Schizophr Res. 2008a;100:252–260. doi: 10.1016/j.schres.2007.11.027. [DOI] [PubMed] [Google Scholar]
  11. Barr RS, Culhane MA, Jubelt LE, Mufti RS, Dyer MA, Weiss AP, Deckersbach T, Kelly JF, Freudenreich O, Goff DC, Evins AE. The effects of transdermal nicotine on cognition in nonsmokers with schizophrenia and nonpsychiatric controls. Neuropsychopharmacology. 2008b;33:480–490. doi: 10.1038/sj.npp.1301423. [DOI] [PubMed] [Google Scholar]
  12. Bird ED, Spokes EG, Barnes J, MacKay AV, Iversen LL, Shepherd M. Increased brain dopamine and reduced glutamic acid decarboxylase and choline acetyl transferase activity in schizophrenia and related psychoses. Lancet. 1977;2:1157–1158. doi: 10.1016/s0140-6736(77)91542-2. [DOI] [PubMed] [Google Scholar]
  13. Blondel A, Sanger DJ, Moser PC. Characterisation of the effects of nicotine in the five-choice serial reaction time task in rats: antagonist studies. Psychopharmacology (Berl) 2000;149:293–305. doi: 10.1007/s002130000378. [DOI] [PubMed] [Google Scholar]
  14. Boess FG, De Vry J, Erb C, Flessner T, Hendrix M, Luithle J, Methfessel C, Riedl B, Schnizler K, van der Staay FJ, van Kampen M, Wiese WB, Koenig G. The novel α7 nicotinic acetylcholine receptor agonist N-[(3R)-1-azabicyclo[2.2.2]oct-3-yl]-7-[2-(methoxy)phenyl]-1-benzofuran-2-carboxamide improves working and recognition memory in rodents. J Pharmacol Exp Ther. 2007;321:716–725. doi: 10.1124/jpet.106.118976. [DOI] [PubMed] [Google Scholar]
  15. Breese CR, Lee MJ, Adams CE, Sullivan B, Logel J, Gillen KM, Marks MJ, Collins AC, Leonard S. Abnormal regulation of high affinity nicotinic receptors in subjects with schizophrenia. Neuropsychopharmacology. 2000;23:351–364. doi: 10.1016/S0893-133X(00)00121-4. [DOI] [PubMed] [Google Scholar]
  16. Bridges TM, LeBois EP, Hopkins CR, Wood MR, Jones CK, Conn PJ, Lindsley CW. The antipsychotic potential of muscarinic allosteric modulation. Drug News Perspect. 2010;23:229–240. doi: 10.1358/dnp.2010.23.4.1416977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Briggs CA, Anderson DJ, Brioni JD, Buccafusco JJ, Buckley MJ, Campbell JE, Decker MW, Donnelly-Roberts D, Elliott RL, Gopalakrishnan M, Holladay MW, Hui YH, Jackson WJ, Kim DJ, Marsh KC, O’Neill A, Prendergast MA, Ryther KB, Sullivan JP, Arneric SP. Functional characterization of the novel neuronal nicotinic acetylcholine 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]
  18. Bubenikova-Valesova V, Horacek J, Vrajova M, Hoschl C. Models of schizophrenia in humans and animals based on inhibition of NMDA receptors. Neurosci Biobehav Rev. 2008;32:1014–1023. doi: 10.1016/j.neubiorev.2008.03.012. [DOI] [PubMed] [Google Scholar]
  19. Buccafusco JJ, Jackson WJ. Beneficial effects of nicotine administered prior to a delayed matching-to-sample task in young and aged monkeys. Neurobiol Aging. 1991;12:233–238. doi: 10.1016/0197-4580(91)90102-p. [DOI] [PubMed] [Google Scholar]
  20. 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]
  21. Buchanan RW, Freedman R, Javitt DC, Abi-Dargham A, Lieberman JA. Recent advances in the development of novel pharmacological agents for the treatment of cognitive impairments in schizophrenia. Schizophr Bull. 2007;33:1120–1130. doi: 10.1093/schbul/sbm083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Bushnell PJ. Behavioral approaches to the assessment of attention in animals. Psychopharmacology (Berl) 1998;138:231–259. doi: 10.1007/s002130050668. [DOI] [PubMed] [Google Scholar]
  23. Bushnell PJ, Oshiro WM, Padnos BK. Detection of visual signals by rats: effects of chlordiazepoxide and cholinergic and adrenergic drugs on sustained attention. Psychopharmacology (Berl) 1997;134:230–241. doi: 10.1007/s002130050446. [DOI] [PubMed] [Google Scholar]
  24. Bustillo JR, Rowland LM, Lauriello J, Petropoulos H, Hammond R, Hart B, Brooks WM. High choline concentrations in the caudate nucleus in antipsychotic-naive patients with schizophrenia. Am J Psychiatry. 2002;159:130–133. doi: 10.1176/appi.ajp.159.1.130. [DOI] [PubMed] [Google Scholar]
  25. Carter CS, Barch DM, Buchanan RW, Bullmore E, Krystal JH, Cohen J, Geyer M, Green M, Nuechterlein KH, Robbins T, Silverstein S, Smith EE, Strauss M, Wykes T, Heinssen R. Identifying cognitive mechanisms targeted for treatment development in schizophrenia: an overview of the first meeting of the Cognitive Neuroscience Treatment Research to Improve Cognition in Schizophrenia Initiative. Biol Psychiatry. 2008;64:4–10. doi: 10.1016/j.biopsych.2008.03.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Chambers RA, Krystal JH, Self DW. A neurobiological basis for substance abuse comorbidity in schizophrenia. Biol Psychiatry. 2001;50:71–83. doi: 10.1016/s0006-3223(01)01134-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Chandley MJ, Miller MN, Kwasigroch CN, Wilson TD, Miller BE. Increased antibodies for the α7 subunit of the nicotinic receptor in schizophrenia. Schizophr Res. 2009;109:98–101. doi: 10.1016/j.schres.2009.01.023. [DOI] [PubMed] [Google Scholar]
  28. Chapman S, Ragg M, McGeechan K. Citation bias in reported smoking prevalence in people with schizophrenia. Aust N Z J Psychiatry. 2009;43:277–282. doi: 10.1080/00048670802653372. [DOI] [PubMed] [Google Scholar]
  29. Chudasama Y, Robbins TW. Psychopharmacological approaches to modulating attention in the five-choice serial reaction time task: implications for schizophrenia. Psychopharmacology (Berl) 2004;174:86–98. doi: 10.1007/s00213-004-1805-y. [DOI] [PubMed] [Google Scholar]
  30. Corrigall WA, Coen KM. Nicotine maintains robust self-administration in rats on a limited-access schedule. Psychopharmacology (Berl) 1989;99:473–478. doi: 10.1007/BF00589894. [DOI] [PubMed] [Google Scholar]
  31. Court J, Spurden D, Lloyd S, McKeith I, Ballard C, Cairns N, Kerwin R, Perry R, Perry E. Neuronal nicotinic receptors in dementia with Lewy bodies and schizophrenia: α-bungarotoxin and nicotine binding in the thalamus. J Neurochem. 1999;73:1590–1597. doi: 10.1046/j.1471-4159.1999.0731590.x. [DOI] [PubMed] [Google Scholar]
  32. Cox T, Tye N. Effects of physotigmine on the acquisition of a position discrimination in rats. Neuropharmacology. 1973;12:477–484. doi: 10.1016/0028-3908(73)90064-6. [DOI] [PubMed] [Google Scholar]
  33. Dani JA. Overview of nicotinic receptors and their roles in the central nervous system. Biol Psychiatry. 2001;49:166–174. doi: 10.1016/s0006-3223(00)01011-8. [DOI] [PubMed] [Google Scholar]
  34. Davis KL, Mohs RC. Enhancement of memory processes in Alzheimer’s disease with multiple-dose intravenous physostigmine. Am J Psychiatry. 1982;139:1421–1424. doi: 10.1176/ajp.139.11.1421. [DOI] [PubMed] [Google Scholar]
  35. de Leon J, Dadvand M, Canuso C, White AO, Stanilla JK, Simpson GM. Schizophrenia and smoking: an epidemiological survey in a state hospital. Am J Psychiatry. 1995;152:453–455. doi: 10.1176/ajp.152.3.453. [DOI] [PubMed] [Google Scholar]
  36. de Leon J, Diaz FJ. A meta-analysis of worldwide studies demonstrates an association between schizophrenia and tobacco smoking behaviors. Schizophr Res. 2005;76:135–157. doi: 10.1016/j.schres.2005.02.010. [DOI] [PubMed] [Google Scholar]
  37. Dome P, Lazary J, Kalapos MP, Rihmer Z. Smoking, nicotine and neuropsychiatric disorders. Neurosci Biobehav Rev. 2010;34:295–342. doi: 10.1016/j.neubiorev.2009.07.013. [DOI] [PubMed] [Google Scholar]
  38. Drachman DA. Memory and cognitive function in man: does the cholinergic system have a specific role? Neurology. 1977;27:783–790. doi: 10.1212/wnl.27.8.783. [DOI] [PubMed] [Google Scholar]
  39. Dunbar G, Boeijinga PH, Demazieres A, Cisterni C, Kuchibhatla R, Wesnes K, Luthringer R. Effects of TC-1734 (AZD3480), a selective neuronal nicotinic receptor agonist, on cognitive performance and the EEG of young healthy male volunteers. Psychopharmacology (Berl) 2007;191:919–929. doi: 10.1007/s00213-006-0675-x. [DOI] [PubMed] [Google Scholar]
  40. Durany N, Zochling R, Boissl KW, Paulus W, Ransmayr G, Tatschner T, Danielczyk W, Jellinger K, Deckert J, Riederer P. Human postmortem striatal α4β2 nicotinic acetylcholine receptor density in schizophrenia and Parkinson’s syndrome. Neurosci Lett. 2000;287:109–112. doi: 10.1016/s0304-3940(00)01144-7. [DOI] [PubMed] [Google Scholar]
  41. Flores CM, Rogers SW, Pabreza LA, Wolfe BB, Kellar KJ. A subtype of nicotinic cholinergic receptor in rat brain is composed of α4 and β2 subunits and is up-regulated by chronic nicotine treatment. Mol Pharmacol. 1992;41:31–37. [PubMed] [Google Scholar]
  42. Freedman R, Hall M, Adler LE, Leonard S. Evidence in postmortem brain tissue for decreased numbers of hippocampal nicotinic receptors in schizophrenia. Biol Psychiatry. 1995;38:22–33. doi: 10.1016/0006-3223(94)00252-X. [DOI] [PubMed] [Google Scholar]
  43. Freedman R, Olincy A, Buchanan RW, Harris JG, Gold JM, Johnson L, Allensworth D, Guzman-Bonilla A, Clement B, Ball MP, Kutnick J, Pender V, Martin LF, Stevens KE, Wagner BD, Zerbe GO, Soti F, Kem WR. Initial phase 2 trial of a nicotinic agonist in schizophrenia. Am J Psychiatry. 2008;165:1040–1047. doi: 10.1176/appi.ajp.2008.07071135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Garcia-Rill E, Biedermann JA, Chambers T, Skinner RD, Mrak RE, Husain M, Karson CN. Mesopontine neurons in schizophrenia. Neuroscience. 1995;66:321–335. doi: 10.1016/0306-4522(94)00564-l. [DOI] [PubMed] [Google Scholar]
  45. Geyer MA, Markou A. Animal models of psychiatric disorders. In: Bloom FE, Kupfer DJ, editors. Psychopharmacology: The Fourth Generation of Progress. Raven Press; New York: 1995. pp. 787–798. [Google Scholar]
  46. Goff DC, Coyle JT. The emerging role of glutamate in the pathophysiology and treatment of schizophrenia. Am J Psychiatry. 2001;158:1367–1377. doi: 10.1176/appi.ajp.158.9.1367. [DOI] [PubMed] [Google Scholar]
  47. Gold JM, Harvey PD. Cognitive deficits in schizophrenia. Psychiatr Clin North Am. 1993;16:295–312. [PubMed] [Google Scholar]
  48. Goldberg SR, Henningfield JE. Reinforcing effects of nicotine in humans and experimental animals responding under intermittent schedules of i. v. drug injection. Pharmacol Biochem Behav. 1988;30:227–234. doi: 10.1016/0091-3057(88)90450-9. [DOI] [PubMed] [Google Scholar]
  49. Grant BF, Hasin DS, Chou SP, Stinson FS, Dawson DA. Nicotine dependence and psychiatric disorders in the United States: results from the National Epidemiologic Survey on Alcohol and Related Conditions. Arch Gen Psychiatry. 2004;61:1107–1115. doi: 10.1001/archpsyc.61.11.1107. [DOI] [PubMed] [Google Scholar]
  50. Greco B, Invernizzi RW, Carli M. Phencyclidine-induced impairment in attention and response control depends on the background genotype of mice: reversal by the mGLU2/3 receptor agonist LY379268. Psychopharmacology (Berl) 2005;179:68–76. doi: 10.1007/s00213-004-2127-9. [DOI] [PubMed] [Google Scholar]
  51. Grilly DM. A verification of psychostimulant-induced improvement in sustained attention in rats: effects of d-amphetamine, nicotine, and pemoline. Exp Clin Psychopharmacol. 2000;8:14–21. doi: 10.1037//1064-1297.8.1.14. [DOI] [PubMed] [Google Scholar]
  52. Grinevich VP, Papke RL, Lippiello PM, Bencherif M. Atypical antipsychotics as noncompetitive inhibitors of α4β2 and α7 neuronal nicotinic receptors. Neuropharmacology. 2009;57:183–191. doi: 10.1016/j.neuropharm.2009.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Grottick AJ, Higgins GA. Effect of subtype selective nicotinic compounds on attention as assessed by the five-choice serial reaction time task. Behav Brain Res. 2000;117:197–208. doi: 10.1016/s0166-4328(00)00305-3. [DOI] [PubMed] [Google Scholar]
  54. Guan ZZ, Zhang X, Blennow K, Nordberg A. Decreased protein level of nicotinic receptor α7 subunit in the frontal cortex from schizophrenic brain. Neuroreport. 1999;10:1779–1782. doi: 10.1097/00001756-199906030-00028. [DOI] [PubMed] [Google Scholar]
  55. Hahn B, Sharples CG, Wonnacott S, Shoaib M, Stolerman IP. Attentional effects of nicotinic agonists in rats. Neuropharmacology. 2003;44:1054–1067. doi: 10.1016/s0028-3908(03)00099-6. [DOI] [PubMed] [Google Scholar]
  56. Hahn B, Shoaib M, Stolerman IP. Nicotine-induced enhancement of attention in the five-choice serial reaction time task: the influence of task demands. Psychopharmacology (Berl) 2002;162:129–137. doi: 10.1007/s00213-002-1005-6. [DOI] [PubMed] [Google Scholar]
  57. Handelmann GE, Contreras PC, O’Donohue TL. Selective memory impairment by phencyclidine in rats. Eur J Pharmacol. 1987;140:69–73. doi: 10.1016/0014-2999(87)90635-2. [DOI] [PubMed] [Google Scholar]
  58. Harris JG, Kongs S, Allensworth D, Martin L, Tregellas J, Sullivan B, Zerbe G, Freedman R. Effects of nicotine on cognitive deficits in schizophrenia. Neuropsychopharmacology. 2004;29:1378–1385. doi: 10.1038/sj.npp.1300450. [DOI] [PubMed] [Google Scholar]
  59. Hashimoto K, Ishima T, Fujita Y, Matsuo M, Kobashi T, Takahagi M, Tsukada H, Iyo M. Phencyclidine-induced cognitive deficits in mice are improved by subsequent subchronic administration of the novel selective α7 nicotinic receptor agonist SSR180711. Biol Psychiatry. 2008;63:92–97. doi: 10.1016/j.biopsych.2007.04.034. [DOI] [PubMed] [Google Scholar]
  60. Hasselmo ME, Sarter M. Modes and models of forebrain cholinergic neuromodulation of cognition. Neuropsychopharmacology. 2010 doi: 10.1038/npp.2010.104. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Hauser TA, Kucinski A, Jordan KG, Gatto GJ, Wersinger SR, Hesse RA, Stachowiak EK, Stachowiak MK, Papke RL, Lippiello PM, Bencherif M. TC-5619: an alpha7 neuronal nicotinic receptor-selective agonist that demonstrates efficacy in animal models of the positive and negative symptoms and cognitive dysfunction of schizophrenia. Biochem Pharmacol. 2009;78:803–812. doi: 10.1016/j.bcp.2009.05.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Heishman SJ, Snyder FR, Henningfield JE. Performance, subjective, and physiological effects of nicotine in non-smokers. Drug Alcohol Depend. 1993;34:11–18. doi: 10.1016/0376-8716(93)90041-n. [DOI] [PubMed] [Google Scholar]
  63. Hennekens CH. Increasing global burden of cardiovascular disease in general populations and patients with schizophrenia. J Clin Psychiatry. 2007;68(Suppl 4):4–7. [PubMed] [Google Scholar]
  64. Howe WM, Ji J, Parikh V, Williams S, Mocaer E, Trocme-Thibierge C, Sarter M. Enhancement of attentional performance by selective stimulation of α4β2(*) nAChRs: underlying cholinergic mechanisms. Neuropsychopharmacology. 2010;35:1391–1401. doi: 10.1038/npp.2010.9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Hoyle E, Genn RF, Fernandes C, Stolerman IP. Impaired performance of alpha7 nicotinic receptor knockout mice in the five-choice serial reaction time task. Psychopharmacology (Berl) 2006;189:211–223. doi: 10.1007/s00213-006-0549-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Hughes JR, Hatsukami DK, Mitchell JE, Dahlgren LA. Prevalence of smoking among psychiatric outpatients. Am J Psychiatry. 1986;143:993–997. doi: 10.1176/ajp.143.8.993. [DOI] [PubMed] [Google Scholar]
  67. Jones CK, Brady AE, Davis AA, Xiang Z, Bubser M, Tantawy MN, Kane AS, Bridges TM, Kennedy JP, Bradley SR, Peterson TE, Ansari MS, Baldwin RM, Kessler RM, Deutch AY, Lah JJ, Levey AI, Lindsley CW, Conn PJ. 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]
  68. Karson CN, Casanova MF, Kleinman JE, Griffin WS. Choline acetyltransferase in schizophrenia. Am J Psychiatry. 1993;150:454–459. doi: 10.1176/ajp.150.3.454. [DOI] [PubMed] [Google Scholar]
  69. Kasper S, Resinger E. Cognitive effects and antipsychotic treatment. Psychoneuroendocrinology. 2003;28(Suppl 1):27–38. doi: 10.1016/s0306-4530(02)00115-4. [DOI] [PubMed] [Google Scholar]
  70. Kesner RP, Dakis M. Phencyclidine disrupts acquisition and retention performance within a spatial continuous recognition memory task. Pharmacol Biochem Behav. 1993;44:419–424. doi: 10.1016/0091-3057(93)90484-b. [DOI] [PubMed] [Google Scholar]
  71. Khantzian EJ. The self-medication hypothesis of addictive disorders: focus on heroin and cocaine dependence. Am J Psychiatry. 1985;142:1259–1264. doi: 10.1176/ajp.142.11.1259. [DOI] [PubMed] [Google Scholar]
  72. Kim SH, Price MT, Olney JW, Farber NB. Excessive cerebrocortical release of acetylcholine induced by NMDA antagonists is reduced by GABAergic and α2-adrenergic agonists. Mol Psychiatry. 1999;4:344–352. doi: 10.1038/sj.mp.4000529. [DOI] [PubMed] [Google Scholar]
  73. Konradi C, Heckers S. Molecular aspects of glutamate dysregulation: implications for schizophrenia and its treatment. Pharmacol Ther. 2003;97:153–179. doi: 10.1016/s0163-7258(02)00328-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Krystal JH, Bennett A, Abi-Saab D, Belger A, Karper LP, D’Souza DC, Lipschitz D, Abi-Dargham A, Charney DS. Dissociation of ketamine effects on rule acquisition and rule implementation: possible relevance to NMDA receptor contributions to executive cognitive functions. Biol Psychiatry. 2000;47:137–143. doi: 10.1016/s0006-3223(99)00097-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Krystal JH, Karper LP, Seibyl JP, Freeman GK, Delaney R, Bremner JD, Heninger GR, Bowers MB, Jr, Charney DS. Subanesthetic effects of the noncompetitive NMDA antagonist, ketamine, in humans: psychotomimetic, perceptual, cognitive, and neuroendocrine responses. Arch Gen Psychiatry. 1994;51:199–214. doi: 10.1001/archpsyc.1994.03950030035004. [DOI] [PubMed] [Google Scholar]
  76. Leonard S, Adler LE, Benhammou K, Berger R, Breese CR, Drebing C, Gault J, Lee MJ, Logel J, Olincy A, Ross RG, Stevens K, Sullivan B, Vianzon R, Virnich DE, Waldo M, Walton K, Freedman R. Smoking and mental illness. Pharmacol Biochem Behav. 2001;70:561–570. doi: 10.1016/s0091-3057(01)00677-3. [DOI] [PubMed] [Google Scholar]
  77. Leonard S, Gault J, Adams C, Breese CR, Rollins Y, Adler LE, Olincy A, Freedman R. Nicotinic receptors, smoking and schizophrenia. Restor Neurol Neurosci. 1998;12:195–201. [PubMed] [Google Scholar]
  78. Levin ED. Psychopharmacological effects in the radial-arm maze. Neurosci Biobehav Rev. 1988;12:169–175. doi: 10.1016/s0149-7634(88)80008-3. [DOI] [PubMed] [Google Scholar]
  79. Levin ED, Bettegowda C, Weaver T, Christopher NC. Nicotine-dizocilpine interactions and working and reference memory performance of rats in the radial-arm maze. Pharmacol Biochem Behav. 1998;61:335–340. doi: 10.1016/s0091-3057(98)00109-9. [DOI] [PubMed] [Google Scholar]
  80. Levin ED, Bradley A, Addy N, Sigurani N. Hippocampal α7 and α4β2 nicotinic receptors and working memory. Neuroscience. 2002;109:757–765. doi: 10.1016/s0306-4522(01)00538-3. [DOI] [PubMed] [Google Scholar]
  81. Levin ED, Briggs SJ, Christopher NC, Rose JE. Chronic nicotinic stimulation and blockade effects on working memory. Behav Pharmacol. 1993;4:179–182. [PubMed] [Google Scholar]
  82. Levin ED, Christopher NC, Weaver T, Moore J, Brucato F. Ventral hippocampal ibotenic acid lesions block chronic nicotine-induced spatial working memory improvement in rats. Brain Res Cogn Brain Res. 1999;7:405–410. doi: 10.1016/s0926-6410(98)00044-5. [DOI] [PubMed] [Google Scholar]
  83. Levin ED, Lee C, Rose JE, Reyes A, Ellison G, Jarvik M, Gritz E. Chronic nicotine and withdrawal effects on radial-arm maze performance in rats. Behav Neural Biol. 1990;53:269–276. doi: 10.1016/0163-1047(90)90509-5. [DOI] [PubMed] [Google Scholar]
  84. Levin ED, Petro A, Beatty A. Olanzapine interactions with nicotine and mecamylamine in rats: effects on memory function. Neurotoxicol Teratol. 2005;27:459–464. doi: 10.1016/j.ntt.2005.01.011. [DOI] [PubMed] [Google Scholar]
  85. 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]
  86. Levin ED, Rose JE. Anticholinergic sensitivity following chronic nicotine administration as measured by radial-arm maze performance in rats. Behav Pharmacol. 1990;1:511–520. [PubMed] [Google Scholar]
  87. Levin ED, Rose JE. Nicotinic and muscarinic interactions and choice accuracy in the radial-arm maze. Brain Res Bull. 1991;27:125–128. doi: 10.1016/0361-9230(91)90293-s. [DOI] [PubMed] [Google Scholar]
  88. Levin ED, Torry D. Acute and chronic nicotine effects on working memory in aged rats. Psychopharmacology (Berl) 1996;123:88–97. doi: 10.1007/BF02246285. [DOI] [PubMed] [Google Scholar]
  89. Liljequist R, Mattila MJ. Effect of physostigmine and scopolamine on the memory functions of chess players. Med Biol. 1979;57:402–405. [PubMed] [Google Scholar]
  90. Lysakowski A, Wainer BH, Bruce G, Hersh LB. An atlas of the regional and laminar distribution of choline acetyltransferase immunoreactivity in rat cerebral cortex. Neuroscience. 1989;28:291–336. doi: 10.1016/0306-4522(89)90180-2. [DOI] [PubMed] [Google Scholar]
  91. Maehara S, Hikichi H, Satow A, Okuda S, Ohta H. Antipsychotic property of a muscarinic receptor agonist in animal models for schizophrenia. Pharmacol Biochem Behav. 2008;91:140–149. doi: 10.1016/j.pbb.2008.06.023. [DOI] [PubMed] [Google Scholar]
  92. Malhotra AK, Pinals DA, Adler CM, Elman I, Clifton A, Pickar D, Breier A. Ketamine-induced exacerbation of psychotic symptoms and cognitive impairment in neuroleptic-free schizophrenics. Neuropsychopharmacology. 1997;17:141–150. doi: 10.1016/S0893-133X(97)00036-5. [DOI] [PubMed] [Google Scholar]
  93. Malhotra AK, Pinals DA, Weingartner H, Sirocco K, Missar CD, Pickar D, Breier A. NMDA receptor function and human cognition: the effects of ketamine in healthy volunteers. Neuropsychopharmacology. 1996;14:301–307. doi: 10.1016/0893-133X(95)00137-3. [DOI] [PubMed] [Google Scholar]
  94. Marder SR, Fenton W. Measurement and Treatment Research to Improve Cognition in Schizophrenia: NIMH MATRICS initiative to support the development of agents for improving cognition in schizophrenia. Schizophr Res. 2004;72:5–9. doi: 10.1016/j.schres.2004.09.010. [DOI] [PubMed] [Google Scholar]
  95. Margutti P, Delunardo F, Ortona E. Autoantibodies associated with psychiatric disorders. Curr Neurovasc Res. 2006;3:149–157. doi: 10.2174/156720206776875894. [DOI] [PubMed] [Google Scholar]
  96. Markou A, Chiamulera C, Geyer MA, Tricklebank M, Steckler T. Removing obstacles in neuroscience drug discovery: the future path for animal models. Neuropsychopharmacology. 2009;34:74–89. doi: 10.1038/npp.2008.173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Markou A, Kosten TR, Koob GF. Neurobiological similarities in depression and drug dependence: a self-medication hypothesis. Neuropsychopharmacology. 1998;18:135–174. doi: 10.1016/S0893-133X(97)00113-9. [DOI] [PubMed] [Google Scholar]
  98. Masuda R, Fukuda M, Ono T, Endo S. Neuronal responses at the sight of objects in monkey basal forebrain subregions during operant visual tasks. Neurobiol Learn Mem. 1997;67:181–196. doi: 10.1006/nlme.1996.3756. [DOI] [PubMed] [Google Scholar]
  99. Mathew SV, Law AJ, Lipska BK, Davila-Garcia MI, Zamora ED, Mitkus SN, Vakkalanka R, Straub RE, Weinberger DR, Kleinman JE, Hyde TM. α7 nicotinic acetylcholine receptor mRNA expression and binding in postmortem human brain are associated with genetic variation in neuregulin 1. Hum Mol Genet. 2007;16:2921–2932. doi: 10.1093/hmg/ddm253. [DOI] [PubMed] [Google Scholar]
  100. McGaughy J, Sarter M. Behavioral vigilance in rats: task validation and effects of age, amphetamine, and benzodiazepine receptor ligands. Psychopharmacology (Berl) 1995;117:340–357. doi: 10.1007/BF02246109. [DOI] [PubMed] [Google Scholar]
  101. McGeer PL, McGeer EG. Possible changes in striatal and limbic cholinergic systems in schizophrenia. Arch Gen Psychiatry. 1977;34:1319–1323. doi: 10.1001/archpsyc.1977.01770230061003. [DOI] [PubMed] [Google Scholar]
  102. Mesulam MM. The cholinergic innervation of the human cerebral cortex. Prog Brain Res. 2004;145:67–78. doi: 10.1016/S0079-6123(03)45004-8. [DOI] [PubMed] [Google Scholar]
  103. Mirza NR, Bright JL. Nicotine-induced enhancements in the five-choice serial reaction time task in rats are strain-dependent. Psychopharmacology (Berl) 2001;154:8–12. doi: 10.1007/s002130000605. [DOI] [PubMed] [Google Scholar]
  104. Morgan CJ, Mofeez A, Brandner B, Bromley L, Curran HV. Acute effects of ketamine on memory systems and psychotic symptoms in healthy volunteers. Neuropsychopharmacology. 2004;29:208–218. doi: 10.1038/sj.npp.1300342. [DOI] [PubMed] [Google Scholar]
  105. Muir JL, Everitt BJ, Robbins TW. Reversal of visual attentional dysfunction following lesions of the cholinergic basal forebrain by physostigmine and nicotine but not by the 5-HT3 receptor antagonist, ondansetron. Psychopharmacology (Berl) 1995;118:82–92. doi: 10.1007/BF02245253. [DOI] [PubMed] [Google Scholar]
  106. Nelsen JM, Goldstein L. Improvement of performance on an attention task with chronic nicotine treatment in rats. Psychopharmacologia. 1972;26:347–360. doi: 10.1007/BF00421900. [DOI] [PubMed] [Google Scholar]
  107. Nelson CL, Burk JA, Bruno JP, Sarter M. Effects of acute and repeated systemic administration of ketamine on prefrontal acetylcholine release and sustained attention performance in rats. Psychopharmacology (Berl) 2002;161:168–179. doi: 10.1007/s00213-002-1004-7. [DOI] [PubMed] [Google Scholar]
  108. Nuechterlein KH, Barch DM, Gold JM, Goldberg TE, Green MF, Heaton RK. Identification of separable cognitive factors in schizophrenia. Schizophr Res. 2004;72:29–39. doi: 10.1016/j.schres.2004.09.007. [DOI] [PubMed] [Google Scholar]
  109. Nuotto E. Psychomotor, physiological and cognitive effects of scopolamine and ephedrine in healthy man. Eur J Clin Pharmacol. 1983;24:603–609. doi: 10.1007/BF00542208. [DOI] [PubMed] [Google Scholar]
  110. Olincy A, Harris JG, Johnson LL, Pender V, Kongs S, Allensworth D, Ellis J, Zerbe GO, Leonard S, Stevens KE, Stevens JO, Martin L, Adler LE, Soti F, Kem WR, Freedman R. Proof-of-concept trial of an α7 nicotinic agonist in schizophrenia. Arch Gen Psychiatry. 2006;63:630–638. doi: 10.1001/archpsyc.63.6.630. [DOI] [PubMed] [Google Scholar]
  111. Olincy A, Stevens KE. Treating schizophrenia symptoms with an α7 nicotinic agonist, from mice to men. Biochem Pharmacol. 2007;74:1192–1201. doi: 10.1016/j.bcp.2007.07.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Pepeu G, Giovannini MG. Changes in acetylcholine extracellular levels during cognitive processes. Learn Mem. 2004;11:21–27. doi: 10.1101/lm.68104. [DOI] [PubMed] [Google Scholar]
  113. Rezvani AH, Bushnell PJ, Levin ED. Effects of nicotine and mecamylamine on choice accuracy in an operant visual signal detection task in female rats. Psychopharmacology (Berl) 2002;164:369–375. doi: 10.1007/s00213-002-1221-0. [DOI] [PubMed] [Google Scholar]
  114. Rezvani AH, Kholdebarin E, Dawson E, Levin ED. Nicotine and clozapine effects on attentional performance impaired by the NMDA antagonist dizocilpine in female rats. Int J Neuropsychopharmacol. 2008;11:63–70. doi: 10.1017/S1461145706007528. [DOI] [PubMed] [Google Scholar]
  115. Robbins TW. The 5-choice serial reaction time task: behavioural pharmacology and functional neurochemistry. Psychopharmacology (Berl) 2002;163:362–380. doi: 10.1007/s00213-002-1154-7. [DOI] [PubMed] [Google Scholar]
  116. Rollema H, Hajos M, Seymour PA, Kozak R, Majchrzak MJ, Guanowsky V, Horner WE, Chapin DS, Hoffmann WE, Johnson DE, McLean S, Freeman J, Williams KE. 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]
  117. Rye DB, Wainer BH, Mesulam MM, Mufson EJ, Saper CB. Cortical projections arising from the basal forebrain: a study of cholinergic and noncholinergic components employing combined retrograde tracing and immunohistochemical localization of choline acetyltransferase. Neuroscience. 1984;13:627–643. doi: 10.1016/0306-4522(84)90083-6. [DOI] [PubMed] [Google Scholar]
  118. Sacco KA, Termine A, Seyal A, Dudas MM, Vessicchio JC, Krishnan-Sarin S, Jatlow PI, Wexler BE, George TP. Effects of cigarette smoking on spatial working memory and attentional deficits in schizophrenia: involvement of nicotinic receptor mechanisms. Arch Gen Psychiatry. 2005;62:649–659. doi: 10.1001/archpsyc.62.6.649. [DOI] [PubMed] [Google Scholar]
  119. Sarter M, Nelson CL, Bruno JP. Cortical cholinergic transmission and cortical information processing in schizophrenia. Schizophr Bull. 2005;31:117–138. doi: 10.1093/schbul/sbi006. [DOI] [PubMed] [Google Scholar]
  120. Sato K, Wu J, Kikuchi T, Wang Y, Watanabe I, Okumura F. Differential effects of ketamine and pentobarbitone on acetylcholine release from the rat hippocampus and striatum. Br J Anaesth. 1996;77:381–384. doi: 10.1093/bja/77.3.381. [DOI] [PubMed] [Google Scholar]
  121. Schwartz RD, Kellar KJ. Nicotinic cholinergic receptor binding sites in the brain: regulation in vivo. Science. 1983;220:214–216. doi: 10.1126/science.6828889. [DOI] [PubMed] [Google Scholar]
  122. Sellin AK, Shad M, Tamminga C. Muscarinic agonists for the treatment of cognition in schizophrenia. CNS Spectr. 2008;13:985–996. doi: 10.1017/s1092852900014048. [DOI] [PubMed] [Google Scholar]
  123. Semenova S, Stolerman IP, Markou A. Chronic nicotine administration improves attention while nicotine withdrawal induces performance deficits in the 5-choice serial reaction time task in rats. Pharmacol Biochem Behav. 2007;87:360–368. doi: 10.1016/j.pbb.2007.05.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Shanmugam G, Bhutani S, Khan DA, Brown ES. Psychiatric considerations in pulmonary disease. Psychiatr Clin North Am. 2007;30:761–780. doi: 10.1016/j.psc.2007.07.008. [DOI] [PubMed] [Google Scholar]
  125. Singhal SK, Zhang L, Morales M, Oz M. Antipsychotic clozapine inhibits the function of α7-nicotinic acetylcholine receptors. Neuropharmacology. 2007;52:387–394. doi: 10.1016/j.neuropharm.2006.08.023. [DOI] [PubMed] [Google Scholar]
  126. Smith RC, Warner-Cohen J, Matute M, Butler E, Kelly E, Vaidhyanathaswamy S, Khan A. Effects of nicotine nasal spray on cognitive function in schizophrenia. Neuropsychopharmacology. 2006;31:637–643. doi: 10.1038/sj.npp.1300881. [DOI] [PubMed] [Google Scholar]
  127. Spring B, Pingitore R, McChargue DE. Reward value of cigarette smoking for comparably heavy smoking schizophrenic, depressed, and nonpatient smokers. Am J Psychiatry. 2003;160:316–322. doi: 10.1176/appi.ajp.160.2.316. [DOI] [PubMed] [Google Scholar]
  128. Steinpreis RE. The behavioral and neurochemical effects of phencyclidine in humans and animals: some implications for modeling psychosis. Behav Brain Res. 1996;74:45–55. doi: 10.1016/0166-4328(95)00162-x. [DOI] [PubMed] [Google Scholar]
  129. Stolerman IP, Mirza NR, Hahn B, Shoaib M. Nicotine in an animal model of attention. Eur J Pharmacol. 2000;393:147–154. doi: 10.1016/s0014-2999(99)00886-9. [DOI] [PubMed] [Google Scholar]
  130. Taly A, Corringer PJ, Guedin D, Lestage P, Changeux JP. Nicotinic receptors: allosteric transitions and therapeutic targets in the nervous system. Nat Rev Drug Discov. 2009;8:733–750. doi: 10.1038/nrd2927. [DOI] [PubMed] [Google Scholar]
  131. Terry AV, Jr, Gearhart DA, Mahadik SP, Warsi S, Davis LW, Waller JL. Chronic exposure to typical or atypical antipsychotics in rodents: temporal effects on central α7 nicotinic acetylcholine receptors. Neuroscience. 2005;136:519–529. doi: 10.1016/j.neuroscience.2005.08.006. [DOI] [PubMed] [Google Scholar]
  132. Terry AV, Jr, Gearhart DA, Mahadik SP, Warsi S, Waller JL. Chronic treatment with first or second generation antipsychotics in rodents: effects on high affinity nicotinic and muscarinic acetylcholine receptors in the brain. Neuroscience. 2006;140:1277–1287. doi: 10.1016/j.neuroscience.2006.03.011. [DOI] [PubMed] [Google Scholar]
  133. Terry AV, Jr, Hill WD, Parikh V, Waller JL, Evans DR, Mahadik SP. Differential effects of haloperidol, risperidone, and clozapine exposure on cholinergic markers and spatial learning performance in rats. Neuropsychopharmacology. 2003;28:300–309. doi: 10.1038/sj.npp.1300039. [DOI] [PubMed] [Google Scholar]
  134. Theberge J, Al-Semaan Y, Drost DJ, Malla AK, Neufeld RW, Bartha R, Manchanda R, Menon R, Densmore M, Schaefer B, Williamson PC. Duration of untreated psychosis vs. N-acetylaspartate and choline in first episode schizophrenia: a 1H magnetic resonance spectroscopy study at 4.0 Tesla. Psychiatry Res. 2004;131:107–114. doi: 10.1016/j.pscychresns.2004.04.002. [DOI] [PubMed] [Google Scholar]
  135. Tidey JW, Rohsenow DJ, Kaplan GB, Swift RM. Cigarette smoking topography in smokers with schizophrenia and matched non-psychiatric controls. Drug Alcohol Depend. 2005;80:259–265. doi: 10.1016/j.drugalcdep.2005.04.002. [DOI] [PubMed] [Google Scholar]
  136. Timofeeva OA, Levin ED. Idazoxan blocks the nicotine-induced reversal of the memory impairment caused by the NMDA glutamate receptor antagonist dizocilpine. Pharmacol Biochem Behav. 2008;90:372–381. doi: 10.1016/j.pbb.2008.03.011. [DOI] [PubMed] [Google Scholar]
  137. Turchi J, Holley LA, Sarter M. Effects of nicotinic acetylcholine receptor ligands on behavioral vigilance in rats. Psychopharmacology (Berl) 1995;118:195–205. doi: 10.1007/BF02245840. [DOI] [PubMed] [Google Scholar]
  138. 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 α7 receptors. Psychopharmacology (Berl) 2004;172:375–383. doi: 10.1007/s00213-003-1668-7. [DOI] [PubMed] [Google Scholar]
  139. Velligan DI, Miller AL. Cognitive dysfunction in schizophrenia and its importance to outcome: the place of atypical antipsychotics in treatment. J Clin Psychiatry. 1999;60(Suppl 23):25–28. [PubMed] [Google Scholar]
  140. Weinberger AH, Sacco KA, Creeden CL, Vessicchio JC, Jatlow PI, George TP. Effects of acute abstinence, reinstatement, and mecamylamine on biochemical and behavioral measures of cigarette smoking in schizophrenia. Schizophr Res. 2007;91:217–225. doi: 10.1016/j.schres.2006.12.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Wilens TE, Biederman J, Spencer TJ, Bostic J, Prince J, Monuteaux MC, Soriano J, Fine C, Abrams A, Rater M, Polisner D. A pilot controlled clinical trial of ABT-418, a cholinergic agonist, in the treatment of adults with attention deficit hyperactivity disorder. Am J Psychiatry. 1999;156:1931–1937. doi: 10.1176/ajp.156.12.1931. [DOI] [PubMed] [Google Scholar]
  142. Wilens TE, Verlinden MH, Adler LA, Wozniak PJ, West SA. ABT-089, a neuronal nicotinic receptor partial agonist, for the treatment of attention-deficit/hyperactivity disorder in adults: results of a pilot study. Biol Psychiatry. 2006;59:1065–1070. doi: 10.1016/j.biopsych.2005.10.029. [DOI] [PubMed] [Google Scholar]
  143. Williams JM, Ziedonis DM, Abanyie F, Steinberg ML, Foulds J, Benowitz NL. Increased nicotine and cotinine levels in smokers with schizophrenia and schizoaffective disorder is not a metabolic effect. Schizophr Res. 2005;79:323–335. doi: 10.1016/j.schres.2005.04.016. [DOI] [PubMed] [Google Scholar]
  144. Yates SL, Bencherif M, Fluhler EN, Lippiello PM. Up-regulation of nicotinic acetylcholine receptors following chronic exposure of rats to mainstream cigarette smoke or α4β2 receptors to nicotine. Biochem Pharmacol. 1995;50:2001–2008. doi: 10.1016/0006-2952(95)02100-0. [DOI] [PubMed] [Google Scholar]
  145. Young JW, Crawford N, Kelly JS, Kerr LE, Marston HM, Spratt C, Finlayson K, Sharkey J. Impaired attention is central to the cognitive deficits observed in alpha 7 deficient mice. Eur Neuropsychopharmacol. 2007;17:145–155. doi: 10.1016/j.euroneuro.2006.03.008. [DOI] [PubMed] [Google Scholar]
  146. Young JW, Finlayson K, Spratt C, Marston HM, Crawford N, Kelly JS, Sharkey J. Nicotine improves sustained attention in mice: evidence for involvement of the α7 nicotinic acetylcholine receptor. Neuropsychopharmacology. 2004;29:891–900. doi: 10.1038/sj.npp.1300393. [DOI] [PubMed] [Google Scholar]
  147. Young JW, Powell SB, Risbrough V, Marston HM, Geyer MA. Using the MATRICS to guide development of a preclinical cognitive test battery for research in schizophrenia. Pharmacol Ther. 2009;122:150–202. doi: 10.1016/j.pharmthera.2009.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES