1. Trang chủ
  2. » Y Tế - Sức Khỏe

Neurochemical Mechanisms in Disease P79 pptx

10 326 0

Đang tải... (xem toàn văn)

THÔNG TIN TÀI LIỆU

Nội dung

Nicotinic Receptors in Brain Diseases 765 variant allele of Chrna7 has been identified in mice. This allele is linked to vari- ability in α7 expression in the hippocampus (Stitzel et al., 1996), neuroanatomical distribution of α7 nAChRs in the hippocampus (Adams et al., 2001), developmental expression of α7 nAChRs in the hippocampus (Adams et al., 2006), and audi- tory gating deficits (Stevens et al., 2001). The fact that the allele of Chrna7 that leads to reduced α7 expression in the hippocampus also leads to impaired auditory gating is consistent with the role of Chrna7 in regulating the auditory gating phe- notype in schizophrenics. Recently, Liu et al. (2006) reported that α7nAChRsare involved in the normal development of the GABAergic system in the hippocam- pus. Thus, abnormal expression of α7 nAChRs during pre- and perinatal periods of development may have long-term consequences on brain function. Suggestive support for a developmental role of α7 nAChRs in impaired auditory gating comes from two recent studies that have shown that perinatal dietary supplementation with choline, an α7-selective agonist, permanently improves gating in two animal models of impaired auditory gating (Stevens et al., 2008a,b). 3.2 Autism A second disease where there appears to be altered expression of nAChRs is autism. Studies have shown that high-affinity nicotinic receptors as measured by [3H] epi- batidine, α4 RNA and anti-α4 antibodies, are reduced in various cortical regions in autistic subjects (Martin-Ruiz et al., 2004; Perry et al., 2001). Using both [3H] epi- batidine and anti-α4 antibodies, Lee et al. (2002) and Martin-Ruiz et al. (2004)also reported that α4 nAChRs are reduced in cerebellar regions in subjects with autism relative to normal controls. The α7 subunit was not found to be altered in expression in cortical regions of autistic patients but was found to be upregulated in cerebellum (Lee et al., 2002; Martin-Ruiz et al., 2004); the binding of [125I] α-bungarotoxin was increased in cerebellum although no change in α7RNAorα7 immunoreactiv- ity was detected. Finally, in a small sample, α7 and β2 but not α4 immunoreactivity was found to be decreased in the thalamus of individuals with autism (Ray et al., 2005). In addition to altered levels of nAChRs, there also appear to be increased num- bers and enlarged morphology of cholinergic neurons in the cortex of children with autism (Bauman and Kemper, 2005). Based on this observation, it has been hypothesized that the downregulation of nAChRs in the cortex and thalamus in autism is the result of a homeostatic response to hypercholinergic activity in the cortex (Lippiello, 2006). The potential hyperactivity in the cortex of individuals with autism may explain the low level of smoking associated with autism relative to both the general population and other mental diseases (Bejerot and Nylander, 2003; Poirier et al., 2002). Nonetheless, there currently are no pharmacological or animal model data to convincingly implicate nAChRs in the etiology of autism. Therefore, the relevance of the altered expression of nAChRs in this disease remains to be determined. 766 J.A. Stitzel 4 Genetic Variants of nAChR Subunit Genes and Brain Disease Each nAChR subunit is encoded by a different gene and any mutation in any of these genes that affects the expression or function of an nAChR could lead to disease or contribute to individual differences in risk for disease. In this section, one dis- ease directly caused by mutations in nAChR subunit genes is discussed. In addition, the potential role of genetic variability in nAChR subunit genes in altering risk for disease is summarized. 4.1 Autosomal Dominant Nocturnal Frontal Lobe Epilepsy (ADNFLE) Autosomal dominant nocturnal frontal lobe epilepsy (ADNFLE) is the only brain disease known to be caused by mutations in genes that code for nicotinic receptor subunits. ADNFLE is a rare, inherited form of epilepsy characterized by hyperki- netic or tonic seizures that tend to occur in clusters. Seizures also are of frontal lobe origin and tend to occur during periods of light sleep (Scheffer et al., 1995). To date, there have been ten mutations in genes that code for nAChR subunits that cause ADNFLE, four in CHRNA4 the gene that encodes the nAChR α4 subunit (Hirose et al., 1999; Leniger et al., 2003; McLellan et al., 2003; Phillips et al., 2000; Saenz et al., 1999; Steinlein et al., 1995, 1997, 2000), five in CHRNB2, the gene that codes the β2 nAChR subunit (Bertrand et al., 2005; De Fusco et al., 2000; Hoda et al., 2008; Phillips et al., 2001), and one in CHRNA2, the gene that encodes the α2 nAChR subunit (Aridon et al., 2006). For more details on these ADNFLE-causing mutations, please see the recent review by Steinlein and Bertrand (2008). A fifth mutation in CHRNA4 recently has been identified that may add to this long list of nAChR subunit gene mutations that cause ADNFLE (Chen et al., 2009b). There also is some debate as to whether the seizure disorder caused by the CHRNA2 mutation is ADNFLE or a related seizure disorder (Hoda et al., 2009). Regardless of whether the seizure disorder caused by the CHRNA2 mutation is ADNFLE or a related dis- ease, it still is an example of a mutation in an nAChR subunit gene that directly causes an inherited disease. Although mutations in CHRNA2, CHRNA4, and CHRNB2 have been shown t o cause ADNFLE or related seizure disorders, how these mutations cause epilepsy remains unknown. However, in vitro functional analysis indicates that a common feature of nAChRs possessing ADNFLE mutations is a gain of function, either by increased sensitivity to acetylcholine or reduced desensitization (Aridon et al., 2006; Bertrand et al., 2002; Hoda et al., 2008, 2009; Leniger et al., 2003; Moulard et al., 2001; Phillips et al., 2001). In addition, two recent studies found that smoking or nicotine treatment decreased seizure frequency in ADNFLE patients with nAChR mutations (Brodtkorb and Picard, 2006; Willoughby et al., 2003). The therapeutic effect of smoking/nicotine presumably was the result of decreasing or inhibiting the function of the hyperactive nAChRs via the well-characterized desensitizing effect of nicotine on α4β2 ∗ . A mechanism by which nAChR gain of function mutations Nicotinic Receptors in Brain Diseases 767 might lead to ADNFLE has been suggested by studies using two lines of mice a engineered to possess different ADNFLE mutations in Chrna4 (Klaassen et al., 2006). In these studies, it was found that nicotine was greater than 20 times more potent at activating inhibitory postsynaptic currents in cortical regions of mice with ADNFLE mutations than in their control littermates. In contrast, nicotine had no effect on excitatory postsynaptic currents. Based on these data and the observation that picrotoxin, a use-dependent GABA antagonist, transiently eliminated epilep- tiform activity in ADNFLE mice, the authors concluded that nAChR-mediated ADNFLE may be caused by hyperactive nAChRs in GABAergic neurons that leads to synchronization of cortical networks. An interesting feature of ADNFLE-causing nAChR mutants is their differen- tial sensitivity to the antiepileptic drug carbamazepine. Carbamazepine can inhibit nAChR function via open channel blockade and three of the known ADNFLE mutations, two in α4 and one in β2, have substantially increased sensitivity to inhi- bition by carbamazepine (Bertrand et al., 2005; Hogg and Bertrand, 2004; Picard et al., 1999). For individuals with any of these carbamazepine-sensitive mutations, carbamazepine has proven to be an effective treatment. In contrast, other ADNFLE- causing mutations in CHRNA4 and CHRNA2 either do not show altered sensitivity to carbamazepine or actually show a reduced sensitivity to inhibition by this drug (Bertrand et al., 2005; Hoda et al., 2009; Leniger et al., 2003). Individuals with these mutations apparently do not benefit from carbamazepine treatment. Thus knowing which nAChR mutation a patient carries can be a valuable aid in treatment selec- tion. However, it should be pointed out that mutations in nAChR subunit genes only account for a fraction of total ADNFLE cases and therefore, the predictive power of nAChR subunit gene mutation identification is restricted to a small percentage of ADNFLE patients. 4.2 Other Genetic Variants in nAChR Subunit Genes and Their Relation to Diseases of the Brain A significant number of polymorphisms and rare mutations in nAChR subunit genes have been implicated in various diseases of the brain through linkage and association studies. Diseases thought to be influenced by nAChR subunit gene vari- ants include schizophrenia, Alzheimer’s disease, non-ADNFLE epilepsies, various cognitive disorders including attention deficits, and drug addiction-related pheno- types. Because there have been several recent reviews on this topic (Portugal and Gould, 2008; Steinlein and Bertrand, 2008; Stitzel, 2008) it is not extensively reviewed here. However, at the time of these reviews, studies began appearing that implicated the gene cluster on chromosome 15q24 that contains CHRNA5, CHRNA3, and CHRNB4 in various aspects of addiction to nicotine, alcohol, and cocaine. This cluster of genes encodes the α5, α3, and β4 nAChR sub- units, respectively. Because this gene cluster repeatedly has been implicated in influencing individual variability in addiction-related measures over the past two years, it warrants some further discussion. The first two reports that implicated this nAChR gene cluster in addiction were published by Bierut et al. (2007) and 768 J.A. Stitzel Saccone et al. (2007). These two studies identified single nucleotide polymor- phisms (SNPs) in both CHRNA5 and CHRNA3 that were associated with nicotine dependence. Subsequent studies have confirmed the association between the CHRNA5 and CHRNA3 SNPs and nicotine dependence (Baker et al., 2009; Bierut et al., 2008; Caporaso et al., 2009; Chen et al., 2009a; Saccone et al., 2009; Spitz et al., 2008; Stevens et al., 2008; Thorgeirsson et al., 2008; Wang et al., 2009; Weiss et al., 2008) as well as implicated the gene cluster in individual differences in level of smok- ing (Berrettini et al., 2008; Le et al., 2008), subjective effects of smoking (Sherva et al., 2008), age of initiation of smoking (Schlaepfer et al., 2008), cocaine addic- tion (Grucza et al., 2008), and alcohol dependence (Joslyn et al., 2008; Schlaepfer et al., 2008; Wang et al., 2008). The same SNPS also have been associated with risk for lung cancer (Amos et al., 2008; Hung et al., 2008; Liu et al., 2008; Shiraishi et al., 2009; Spitz et al., 2008; Thorgeirsson et al., 2008) and chronic obstructive pulmonary disease (COPD) (Pillai et al., 2009; Young et al., 2008). Whether the association between the CHRNA5-CHRNA3 SNPs and lung cancer or COPD are due to an altered risk for smoking or represent an independent signal remains a matter of debate (Volkow et al., 2008). Although beyond the scope of this review (see Egleton et al. (2008), and Song and Spindel (2008) for recent reviews of this topic), nAChRs, including those that contain the α3 and/or α5 subunit are expressed in pulmonary epithelial cells and lung cancer cells so an independent role of SNPs in CHRNA3 and/or CHRNA5 on risk for these diseases certainly is feasible. Although the repeated associations between the CHRNA5–CHRNA3–CHRNB4 gene cluster and the mentioned addiction-related measures strongly suggest that there is one or more polymorphism in the cluster that alters risk for drug use and abuse, the identity of the causative SNP or SNPs is not known. However, a strong candidate is an amino-acid-altering SNP in CHRNA5 that changes a highly conserved aspartic acid codon at amino acid position 398 in the α5 s ub- unit to an asparagine codon. Preliminary in vitro data indicate that the amino acid change associated with increased risk for nicotine dependence (asparagine at position 398) reduces the function of α4β2α5 nAChRs (Bierut et al., 2008). Whether this functional effect of the polymorphism is responsible for altered lia- bility to nicotine dependence and if so, by what mechanism does the change in function of α4β2α5 nAChRs alter addiction risk, are questions that remain to be answered. 5 Diseases Where nAChRs Are Implicated by Therapeutic Effects of Nicotine A putative role for nAChRs in schizophrenia was suggested by the high rate of smoking in schizophrenic patients and the observation that nicotine normalizes neu- rophysiological deficits associated with the disease. As described elsewhere in this review, subsequent studies provided strong evidence for a role of nAChRs in the Nicotinic Receptors in Brain Diseases 769 etiology of this disease. However, there are some diseases where nicotine has been shown to have therapeutic value although a specific role of nAChRs has yet to be established. Two examples of such diseases are discussed here. 5.1 Tourette Syndrome Tourette syndrome is a neurological disorder characterized by repetitive, stereo- typed, involuntary movements and vocalizations called tics (NINDS, 2008). In cases where the tics interfere with normal functioning, therapeutics such as haloperidol often are used. The first evidence for the role of nicotinic receptors in Tourette syndrome came from a study by Sanberg et al. (1988) that reported that nicotine gum in combination with haloperidol improved symptoms in two patients where haloperidol alone was without effect. Follow-up studies have confirmed that nico- tine gum potentiates the therapeutic effects of haloperidol in Tourette syndrome (McConville et al., 1991, 1992; Sanberg et al., 1989). In addition, the use of a trans- dermal nicotine patch rather than nicotine gum has been shown to have long-lasting potentiation of the effects of neuroleptics on tic frequency and severity (Dursun et al., 1994; Silver et al., 1996, 2001). Dursun et al. (1994) also r eported that nico- tine alone improved Tourette syndrome symptoms. In studies where it has been assessed, combined nicotine/neuroleptic treatment also improved measures of atten- tion in Tourette syndrome patients relative to neuroleptic treatment alone (Dursun et al., 1994; Howson et al., 2004). Although the mechanism through which nico- tine improves symptoms of Tourette syndrome is not known, a relatively recent study demonstrated that nicotine normalized deficits in inhibitory function of motor cortex in Tourette syndrome patients ( Orth et al., 2005). Nonetheless, there are no pharmacological data to suggest which nAChR subtypes might be responsible for the therapeutic effects of nicotine in this disease and no postmortem data to eval- uate whether there might be abnormalities in nAChR expression that may directly contribute to the disease. 5.2 Down Syndrome Down syndrome is a genetic disease caused by the inheritance of an extra copy (tri- somy) of chromosome 21. In addition to some common physical features and health problems, most subjects with Down syndrome also have mild to moderate mental retardation. Postmortem brain tissue of Down syndrome patients exhibits amyloid plaques (Burger and Vogel, 1973; Ellis et al., 1974) and cholinergic deficits (Yates et al., 1980) similar to those observed in postmortem brain tissue from Alzheimer patients. In addition, studies with primary cultures from Down syndrome patient brain or cell lines derived from a mouse model of Down syndrome (trisomy 16) suggest that there are cholinergic deficiencies in trisomy 21/16 neurons (Allen et al., 2000; Cardenas et al., 2002; Fiedler et al., 1994). Based on the apparent cholinergic deficits in Down syndrome, Lubec and colleagues (Bernert et al., 2001; Seidl et al., 770 J.A. Stitzel 2000) examined whether transdermal nicotine could improve some of the cognitive deficits associated with Down Syndrome. In both published studies, nicotine was found to improve cognitive performance in the Down syndrome subjects. However, despite the cholinergic deficits and presumably related therapeutic effect of nicotine in Down syndrome, a specific contribution of nAChRs remains to be established for this disease. For example, neither Lee et al. (2002) nor Ray et al. (2005) found any deficits in [3H] epibatidine or [125I] α bungarotoxin binding in postmortem brain of Down syndrome patients. These findings contradict the observation by Engidawork et al. (2001) that the expression of α3 and α7 subunits is altered in Down syndrome. This apparent discrepancy likely is due to the fact that Engidawork et al. (2001)used immunohistochemical methods to detect nAChR subunits. The use of antibodies for standard immunohistochemical detection of nAChR subunits has come under recent scrutiny (Jones and Wonnacott, 2005; Moser et al., 2007). Another mechanism proposed for the therapeutic effect of nicotine in Down syndrome is that the high levels of β amyloid present in the Down syndrome brain are inhibiting the function of α7 nAChRs essentially as described above in Alzheimer’s disease (Deutsch et al., 2003). However, a recent report found no cor- relation between β amyloid levels and dementia in older Down syndrome subjects (Jones et al., 2009). Therefore, despite the therapeutic effect of nicotine in Down syndrome, the specific role of nAChRs remains elusive. 6 Conclusions The research summarized in this review suggests that nAChRs contribute to a wide range of neuropathologies. In many cases the combined therapeutic effect of nico- tine and/or nicotinic drug in addition to detectable differences in nAChR expression provides compelling evidence for a contribution of nAChRs to neuropathology. However, in the diseases that fall into this category, including Alzheimer’s and Parkinson’s disease, schizophrenia, and autism ( among others), the mechanism responsible for the altered expression of the nAChRs is not known. Moreover, whether the altered expression of nAChRs and these diseases is causal or casual remains to be established. In the case of ADNFLE, identified mutations in nAChR subunit genes clearly define a role of nAChRs in diseases of the brain and ani- mal models provide a plausible mechanism. In contrast, the contribution of genetic variants in genes that code for nAChR subunits in diseases other than ADNFLE is only beginning to emerge. Not surprisingly, very little is known regarding the biological mechanisms responsible for the associations between nAChR subunit gene variation and diseases such as nicotine dependence. Finally, there are several diseases such as Tourette syndrome and Down syndrome where nicotine has ther- apeutic effects despite the lack of any detectable alterations in nAChR expression or function. In summary, there is substantial evidence that nAChRs contribute to a wide assortment of brain disease. Nonetheless, much work remains to be done to establish the mechanisms through which nAChRs contribute to the etiology of disease. Nicotinic Receptors in Brain Diseases 771 Acknowedgments This work was supported by grants from the NIH (CA089392, DA022462, MH068582). References Abood LG, Reynolds DT, Bidlack JM (1980) Stereospecific 3H-nicotine binding to intact and solubilized rat brain membranes and evidence for its noncholinergic nature. Life Sci 27: 1307–1314 Abood LG, Reynolds DT, Booth H, Bidlack JM (1981) Sites and mechanisms for nicotine’s action in the brain. Neurosci Biobehav Rev 5:479–486 Adams CE, Stitzel JA, Collins AC, Freedman R (2001) Alpha7-nicotinic receptor expression and the anatomical organization of hippocampal interneurons. Brain Res 922:180–190 Adams CE, Yonchek JC, Stitzel JA (2006) Development of hippocampal alpha7 nicotinic receptors in C3H and DBA/2 congenic mice. Brain Res 1122:27–35 Adams CE, Yonchek JC, Zheng L, Collins AC, Stevens KE (2008) Altered hippocampal circuit function in C3H alpha7 null mutant heterozygous mice. Brain Res 1194:138–145 Adler LE, Hoffer LJ, Griffith J, Waldo MC, Freedman R (1992) Normalization by nicotine of deficient auditory sensory gating in the relatives of schizophrenics. Biol Psychiatry 32: 607–616 Allen DD, Martin J, Arriagada C, Cardenas AM, Rapoport SI, Caviedes R, Caviedes P (2000) Impaired cholinergic function in cell lines derived from the cerebral cortex of normal and trisomy 16 mice. Eur J Neurosci 12:3259–3264 Amos CI, Wu X, Broderick P, Gorlov IP, Gu J, Eisen T, Dong Q, Zhang Q, Gu X, Vijayakrishnan J, Sullivan K, Matakidou A, Wang Y, Mills G, Doheny K, Tsai YY, Chen WV, Shete S, Spitz MR, Houlston RS (2008) Genome-wide association scan of tag SNPs identifies a susceptibility locus for lung cancer at 15q25.1. Nat Genet 40:616–622 Aridon P, Marini C, Di Resta C, Brilli E, De Fusco M, Politi F, Parrini E, Manfredi I, Pisano T, Pruna D, Curia G, Cianchetti C, Pasqualetti M, Becchetti A, Guerrini R, Casari G (2006) Increased sensitivity of the neuronal nicotinic receptor alpha 2 subunit causes familial epilepsy with nocturnal wandering and ictal fear. Am J Hum Genet 79:342–350 Aubert I, Araujo DM, Cecyre D, Robitaille Y, Gauthier S, Quirion R (1992) Comparative alter- ations of nicotinic and muscarinic binding sites in Alzheimer’s and Parkinson’s diseases. J Neurochem 58:529–541 Austin J (2005) Schizophrenia: an update and review. J Genet Couns 14:329–340 Baker N, Adler LE, Franks RD, Waldo M, Berry S, Nagamoto H, Muckle A, Freedman R (1987) Neurophysiological assessment of sensory gating in psychiatric inpatients: comparison between schizophrenia and other diagnoses. Biol Psychiatry 22:603–617 Baker TB, Weiss RB, Bolt D, von Niederhausern A, Fiore MC, Dunn DM, Piper ME, Matsunami N, Smith SS, Coon H, McMahon WM, Scholand MB, Singh N, Hoidal JR, Kim SY, Leppert MF, Cannon DS (2009) Human neuronal acetylcholine receptor A5-A3-B4 haplotypes are associated with multiple nicotine dependence phenotypes. Nicotine Tob Res 11(7):785–796 Banerjee C, Nyengaard JR, Wevers A, de Vos RA, Jansen Steur EN, Lindstrom J, Pilz K, Nowacki S, Bloch W, Schroder H (2000) Cellular expression of alpha7 nicotinic acetylcholine recep- tor protein in the temporal cortex in Alzheimer’s and Parkinson’s disease – a stereological approach. Neurobiol Dis 7:666–672 Bauman ML, Kemper TL (2005) Neuroanatomic observations of the brain in autism: a review and future directions. Int J Dev Neurosci 23:183–187 Bednar I, Paterson D, Marutle A, Pham TM, Svedberg M, Hellstrom-Lindahl E, Mousavi M, Court J, Morris C, Perry E, Mohammed A, Zhang X, Nordberg A (2002) Selective nicotinic receptor consequences in APP(SWE) transgenic mice. Mol Cell Neurosci 20:354–365 Bejerot S, Nylander L (2003) Low prevalence of smoking in patients with autism spectrum disorders. Psychiatry Res 119:177–182 772 J.A. Stitzel Benwell ME, Balfour DJ, Anderson JM (1988) Evidence that tobacco smoking increases the density of (-)-[3H]nicotine binding sites in human brain. J Neurochem 50:1243–1247 Bernert G, Sustrova M, Sovcikova E, Seidl R, Lubec G (2001) Effects of a single transdermal nicotine dose on cognitive performance in adults with Down syndrome. J Neural Transm Suppl (61):237–245 Berrettini W, Yuan X, Tozzi F, Song K, Francks C, Chilcoat H, Waterworth D, Muglia P, Mooser V (2008) Alpha-5/alpha-3 nicotinic receptor subunit alleles increase risk for heavy smoking. Mol Psychiatry 13:368–373 Bertrand D, Elmslie F, Hughes E, Trounce J, Sander T, Bertrand S, Steinlein OK (2005) The CHRNB2 mutation I312M is associated with epilepsy and distinct memory deficits. Neurobiol Dis 20:799–804 Bertrand D, Picard F, Le Hellard S, Weiland S, Favre I, Phillips H, Bertrand S, Berkovic SF, Malafosse A, Mulley J (2002) How mutations in the nAChRs can cause ADNFLE epilepsy. Epilepsia 43(Suppl 5):112–122 Bierut LJ, Madden PA, Breslau N, Johnson EO, Hatsukami D, Pomerleau OF, Swan GE, Rutter J, Bertelsen S, Fox L, Fugman D, Goate AM, Hinrichs AL, Konvicka K, Martin NG, Montgomery GW, Saccone NL, Saccone SF, Wang JC, Chase GA, Rice JP, Ballinger DG (2007) Novel genes identified in a high-density genome wide association study for nicotine dependence. Hum Mol Genet 16:24–35 Bierut LJ, Stitzel JA, Wang JC, Hinrichs AL, Grucza RA, Xuei X, Saccone NL, Saccone SF, Bertelsen S, Fox L, Horton WJ, Breslau N, Budde J, Cloninger CR, Dick DM, Foroud T, Hatsukami D, Hesselbrock V, Johnson EO, Kramer J, Kuperman S, Madden PA, Mayo K, Nurnberger J Jr, Pomerleau O, Porjesz B, Reyes O, Schuckit M, Swan G, Tischfield JA, Edenberg HJ, Rice JP, Goate AM (2008) Variants in nicotinic receptors and risk for nicotine dependence. Am J Psychiatry 165:1163–1171 Bohr IJ, Ray MA, McIntosh JM, Chalon S, Guilloteau D, McKeith IG, Perry RH, Clementi F, Perry EK, Court JA, Piggott MA (2005) Cholinergic nicotinic receptor involvement in movement disorders associated with Lewy body diseases. An autoradiography study using [(125)I]alpha- conotoxinMII in the striatum and thalamus. Exp Neurol 191:292–300 Bordia T, Grady SR, McIntosh JM, Quik M (2007) Nigrostriatal damage preferentially decreases a subpopulation of alpha6beta2 ∗ nAChRs in mouse, monkey, and Parkinson’s disease striatum. Mol Pharmacol 72:52–61 Boutros NN, Belger A, Campbell D, D’Souza C, Krystal J (1999) Comparison of four components of sensory gating in schizophrenia and normal subjects: a preliminary report. Psychiatry Res 88:119–130 Boyd RT (1997) The molecular biology of neuronal nicotinic acetylcholine receptors. Crit Rev Toxicol 27:299–318 Braff DL, Geyer MA, Swerdlow NR (2001) Human studies of prepulse inhibition of startle: nor- mal subjects, patient groups, and pharmacological studies. Psychopharmacology (Berl) 156: 234–258 Breese CR, Lee MJ, Adams CE, Sullivan B, Logel J, Gillen KM, Marks MJ, Collins AC, Leonard S (2000) Abnormal regulation of high affinity nicotinic receptors in subjects with schizophrenia. Neuropsychopharmacology 23:351–364 Breese CR, Marks MJ, Logel J, Adams CE, Sullivan B, Collins AC, Leonard S (1997) Effect of smoking history on [3H]nicotine binding in human postmortem brain. J Pharmacol Exp Ther 282:7–13 Brodtkorb E, Picard F (2006) Tobacco habits modulate autosomal dominant nocturnal frontal lobe epilepsy. Epilepsy Behav 9:515–520 Burger PC, Vogel FS (1973) The development of the pathologic changes of Alzheimer’s disease and senile dementia in patients with Down’s syndrome. Am J Pathol 73:457–476 Burghaus L, Schutz U, Krempel U, Lindstrom J, Schroder H (2003) Loss of nicotinic acetylcholine receptor subunits alpha4 and alpha7 in the cerebral cortex of Parkinson patients. Parkinsonism Relat Disord 9:243–246 Nicotinic Receptors in Brain Diseases 773 Burghaus L, Schutz U, Krempel U, de Vos RA, Jansen Steur EN, Wevers A, Lindstrom J, Schroder H (2000) Quantitative assessment of nicotinic acetylcholine receptor proteins in the cerebral cortex of Alzheimer patients. Brain Res Mol Brain Res 76:385–388 Caporaso N, Gu F, Chatterjee N, Sheng-Chih J, Yu K, Yeager M, Chen C, Jacobs K, Wheeler W, Landi MT, Ziegler RG, Hunter DJ, Chanock S, Hankinson S, Kraft P, Bergen AW (2009) Genome-wide and candidate gene association study of cigarette smoking behaviors. PLoS ONE 4:e4653 Cardenas AM, Arriagada C, Allen DD, Caviedes R, Cortes JF, Martin J, Couve E, Rapoport SI, Shimahara T, Caviedes P (2002) Cell lines derived from hippocampal neurons of the normal and trisomy 16 mouse fetus (a model for Down syndrome) exhibit neuronal markers, cholinergic function, and functional neurotransmitter receptors. Exp Neurol 177:159–170 Champtiaux N, Han ZY, Bessis A, Rossi FM, Zoli M, Marubio L, McIntosh JM, Changeux JP (2002) Distribution and pharmacology of alpha 6-containing nicotinic acetylcholine receptors analyzed with mutant mice. J Neurosci 22:1208–1217 Chen X, Chen J, Williamson VS, An SS, Hettema JM, Aggen SH, Neale MC, Kendler KS (2009a) Variants in nicotinic acetylcholine receptors alpha5 and alpha3 increase risks to nicotine dependence. Am J Med Genet B Neuropsychiatr Genet 150B(7):926–933 Chen D, Patrick JW (1997) The alpha-bungarotoxin-binding nicotinic acetylcholine receptor from rat brain contains only the alpha7 subunit. J Biol Chem 272:24024–24029 Chen Y, Wu L, Fang Y, He Z, Peng B, Shen Y, Xu Q (2009b) A novel mutation of the nicotinic acetylcholine receptor gene CHRNA4 in sporadic nocturnal frontal lobe epilepsy. Epilepsy Res 83:152–156 Chin JH, Tse FW, Harris K, Jhamandas JH (2006) Beta-amyloid enhances intracellular calcium rises mediated by repeated activation of intracellular calcium stores and nicotinic receptors in acutely dissociated rat basal forebrain neurons. Brain Cell Biol 35:173–186 Clarke PB, Schwartz RD, Paul SM, Pert CB, Pert A (1985) Nicotinic binding in rat brain: autora- diographic comparison of [3H]acetylcholine [3H]nicotine, and [125I]-alpha-bungarotoxin. J Neurosci 5:1307–1315 Clementz BA, Geyer MA, Braff DL (1998) Poor P50 suppression among schizophrenia patients and their first-degree biological relatives. Am J Psychiatry 155:1691–1694 Counts SE, He B, Che S, Ikonomovic MD, Dekosky ST, Ginsberg SD, Mufson EJ (2007) Alpha7 nicotinic receptor up-regulation in cholinergic basal forebrain neurons in Alzheimer disease. Arch Neurol 64:1771–1776 Court JA, Piggott MA, Lloyd S, Cookson N, Ballard CG, McKeith IG, Perry RH, Perry EK (2000) Nicotine binding in human striatum: elevation in schizophrenia and reductions in dementia with Lewy bodies, Parkinson’s disease and Alzheimer’s disease and in relation to neuroleptic medication. Neuroscience 98:79–87 Court J, Spurden D, Lloyd S, McKeith I, Ballard C, Cairns N, Kerwin R, Perry R, Perry E (1999) Neuronal nicotinic receptors in dementia with Lewy bodies and schizophrenia: alpha-bungarotoxin and nicotine binding in the thalamus. J Neurochem 73:1590–1597 Couturier S, Bertrand D, Matter JM, Hernandez MC, Bertrand S, Millar N, Valera S, Barkas T, Ballivet M (1990) A neuronal nicotinic acetylcholine receptor subunit (alpha 7) is devel- opmentally regulated and forms a homo-oligomeric channel blocked by alpha-BTX. Neuron 5:847–856 Cui C, Booker TK, Allen RS, Grady SR, Whiteaker P, Marks MJ, Salminen O, Tritto T, Butt CM, Allen WR, Stitzel JA, McIntosh JM, Boulter J, Collins AC, Heinemann SF (2003) The beta3 nicotinic receptor subunit: a component of alpha-conotoxin MII-binding nicotinic acetylcholine receptors that modulate dopamine release and related behaviors. J Neurosci 23:11045–11053 Dalack GW, Healy DJ, Meador-Woodruff JH (1998) Nicotine dependence in schizophrenia: clinical phenomena and laboratory findings. Am J Psychiatry 155:1490–1501 De Fusco M, Becchetti A, Patrignani A, Annesi G, Gambardella A, Quattrone A, Ballabio A, Wanke E, Casari G (2000) The nicotinic receptor beta 2 subunit is mutant in nocturnal frontal lobe epilepsy. Nat Genet 26:275–276 774 J.A. Stitzel Deutsch SI, Rosse RB, Mastropaolo J, Chilton M (2003) Progressive worsening of adaptive func- tions in Down syndrome may be mediated by the complexing of soluble Abeta peptides with the alpha 7 nicotinic acetylcholine receptor: therapeutic implications. Clin Neuropharmacol 26:277–283 Dineley KT, Westerman M, Bui D, Bell K, Ashe KH, Sweatt JD (2001) Beta-amyloid activates the mitogen-activated protein kinase cascade via hippocampal alpha7 nicotinic acetylcholine receptors: in vitro and in vivo mechanisms related to Alzheimer’s disease. J Neurosci 21: 4125–4133 Dineley KT, Xia X, Bui D, Sweatt JD, Zheng H (2002) Accelerated plaque accumulation, asso- ciative l earning deficits, and up-regulation of alpha 7 nicotinic receptor protein in transgenic mice co-expressing mutant human presenilin 1 and amyloid precursor proteins. J Biol Chem 277:22768–22780 Durany N, Zochling R, Boissl KW, Paulus W, Ransmayr G, Tatschner T, Danielczyk W, Jellinger K, Deckert J, Riederer P (2000) Human post-mortem striatal alpha4beta2 nicotinic acetylcholine receptor density in schizophrenia and Parkinson’s syndrome. Neurosci Lett 287:109–112 Dursun SM, Reveley MA, Bird R, Stirton F (1994) Longlasting improvement of Tourette’s syndrome with transdermal nicotine. Lancet 344:1577 Egleton RD, Brown KC, Dasgupta P (2008) Nicotinic acetylcholine receptors in cancer: multiple roles in proliferation and inhibition of apoptosis. Trends Pharmacol Sci 29:151–158 Ellis WG, McCulloch JR, Corley CL (1974) Presenile dementia in Down’s syndrome. Ultrastructural identity with Alzheimer’s disease. Neurology 24:101–106 Ellis JR, Nathan PJ, Villemagne VL, Mulligan RS, Saunder T, Young K, Smith CL, Welch J, Woodward M, Wesnes KA, Savage G, Rowe CC (2009) Galantamine-induced improvements in cognitive function are not related to alterations in alpha(4)beta (2) nicotinic recep- tors in early Alzheimer’s disease as measured in vivo by 2-[(18)F]Fluoro-A-85380 PET. Psychopharmacology (Berl) 202:79–91 Ellis JR, Villemagne VL, Nathan PJ, Mulligan RS, Gong SJ, Chan JG, Sachinidis J, O’Keefe GJ, Pathmaraj K, Wesnes KA, Savage G, Rowe CC (2008) Relationship between nicotinic receptors and cognitive function in early Alzheimer’s disease: a 2-[18F]fluoro-A-85380 PET study. Neurobiol Learn Mem 90:404–412 Engidawork E, Gulesserian T, Balic N, Cairns N, Lubec G (2001) Changes in nicotinic acetyl- choline receptor subunits expression in brain of patients with Down syndrome and Alzheimer’s disease. J Neural Transm Suppl (61):211–222 Erwin RJ, Turetsky BI, Moberg P, Gur RC, Gur RE (1998) P50 abnormalities in schizophre- nia: relationship to clinical and neuropsychological indices of attention. Schizophr Res 33: 157–167 Fiedler JL, Epstein CJ, Rapoport SI, Caviedes R, Caviedes P (1994) Regional alteration of cholin- ergic function in central neurons of trisomy 16 mouse fetuses, an animal model of human trisomy 21 (Down syndrome). Brain Res 658:27–32 Flores CM, Rogers SW, Pabreza LA, Wolfe BB, Kellar KJ (1992) A subtype of nicotinic cholin- ergic receptor in rat brain is composed of alpha 4 and beta 2 subunits and is up-regulated by chronic nicotine treatment. Mol Pharmacol 41:31–37 Freedman R, Hall M, Adler LE, Leonard S (1995) Evidence in postmortem brain tissue for decreased numbers of hippocampal nicotinic receptors in schizophrenia. Biol Psychiatry 38:22–33 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 (2008) Initial phase 2 trial of a nicotinic agonist in schizophrenia. Am J Psychiatry 165:1040–1047 Fu W, Jhamandas JH (2003) Beta-amyloid peptide activates non-alpha7 nicotinic acetylcholine receptors in rat basal forebrain neurons. J Neurophysiol 90:3130–3136 Fucile S (2004) Ca2+ permeability of nicotinic acetylcholine receptors. Cell Calcium 35:1–8 Fucile S, Renzi M, Lax P, Eusebi F (2003) Fractional Ca(2+) current through human neuronal alpha7 nicotinic acetylcholine receptors. Cell Calcium 34:205–209 . Perry EK (2000) Nicotine binding in human striatum: elevation in schizophrenia and reductions in dementia with Lewy bodies, Parkinson’s disease and Alzheimer’s disease and in relation to neuroleptic medication in nAChR expression provides compelling evidence for a contribution of nAChRs to neuropathology. However, in the diseases that fall into this category, including Alzheimer’s and Parkinson’s disease, . assortment of brain disease. Nonetheless, much work remains to be done to establish the mechanisms through which nAChRs contribute to the etiology of disease. Nicotinic Receptors in Brain Diseases 771 Acknowedgments

Ngày đăng: 07/07/2014, 09:20

TỪ KHÓA LIÊN QUAN