1,360
11
Essay, 28 pages (7000 words)

Monoaminergic system modulation in depression and alzheimer’s disease: a new standpoint?

Introduction

Many pathologies have been indicated as comorbid with Alzheimer’s diseases (AD) and in particular neuropsychiatric disorders such as depression ( Ownby et al., 2006 ; Sun et al., 2008 ). Indeed, depression is common in pre-clinical AD and may represent an early manifestation of this disease before the appearance of cognitive impairments ( Geerlings et al., 2000 ; Visser et al., 2000 ). In this regard, much evidence endorses a strong relationship between depression and AD, so much that this mental illness has been proposed as a risk factor for AD or as a prodromic AD phase ( Modrego and Ferrandez, 2004 ). The amyloid cascade hypothesis postulates that neurodegeneration in AD is related to abnormal accumulation of amyloid beta (Aβ) plaques in various areas of the brain. However, soluble forms of this peptide have been implicated in the development of early memory deficits as well as of neuropsychiatric symptoms ( Rowan et al., 2005 ). Indeed, significant cognitive deficits have been directly attributed to soluble Aβ fragments ( Mattson, 2004 ; Cleary et al., 2005 ), and increased levels of soluble Aβ oligomers have been linked to synaptic dysfunction ( Hardy and Selkoe, 2002 ; Selkoe and Schenk, 2003 ). Meanwhile, it has been reported that in depressed patients, Aβ peptide levels are increased ( Pomara and Sidtis, 2010 ). In good agreement, we have previously demonstrated that Aβ, intracerebroventricularly (icv) injected in rats 7 days earlier, evokes a depressive-like profile accompanied by lower cortical serotonin (5-HT) and neurotrophin content ( Colaianna et al., 2010 ). Furthermore, we later reported that such impairment was associated with altered stress response and increased noradrenaline (NA) levels ( Morgese et al., 2014 , 2015 ). In addition, in the same model, cognitive impairment was demonstrated either acutely, such as 2 h after Aβ administration, or more enduringly, i. e., 7 days after the peptide central release ( Morgese et al., 2014 ; Tucci et al., 2014 ; Mhillaj et al., 2018 ). Although the role of dopamine (DA) was less studied concerning depression and AD, recently, its role has been brought to the fore ( Nobili et al., 2017 ) but is still in need of further evaluation.

The present review is aimed at summarizing the main knowledge related to biological and pathological substrates, such as monoamines and related molecules, commonly involved in AD and depressive pathology, with the scope of shedding light on possible therapeutic approaches.

Monoamine System in Depression and Alzheimer’s Disease

Serotonergic System

The treatment of affective disorders is mainly based on the enhancement of the noradrenergic and serotonergic systems through selective or nonselective reuptake inhibitors. Such a pharmacological schedule sinks the roots on the catecholaminergic theory of affective disorders stating the crucial role of lower central NA and 5-HT availability in the insurgence of depression ( Mann et al., 1986 ; Schildkraut, 1995 ; Mann, 1999 ). Alterations in these neurotransmitter systems have also been linked to neurodegenerative disorders such as AD. Impairment of the serotonergic system has been reported in the very early stages of AD ( Versijpt et al., 2003 ; Egashira et al., 2005 ; Kepe et al., 2006 ), and substantial disruption of the serotonergic system in AD has been postulated according to both clinical and postmortem studies ( Morgan et al., 1987 ; Lanctot et al., 2001 ). In this regard, Aβ in its soluble forms, either monomeric or oligomeric, has been associated with the modulation of these systems. In particular, we have previously found that soluble Aβ injected icv in rats caused a significant reduction in 5-HT at the prefrontal cortex level, without interfering with the physiological functioning of other areas such as the striatum or the nucleus accumbens ( Colaianna et al., 2010 ). These results indicated that the prefrontal cortex is an area highly sensitive to Aβ effects, and this area is also crucially involved in the etiopathogenesis of depressive phenomena. Indeed, impairment of 5-HT neurotransmission in the prefrontal area is central to both depressive disorders ( Krishnan and Nestler, 2008 ) and several neurodegenerative diseases ( Mattson, 2004 ; Egashira et al., 2005 ). Furthermore, we have more recently individuated the vulnerability of the hippocampal area to the action of exogenous Aβ icv injected. Indeed, we have found that this peptide can reduce 5-HT levels in the hippocampus, and this event is associated with a proinflammatory state and higher rate of activated microglia ( Mhillaj et al., 2018 ). In addition, the treatment with a selective COX-2 inhibitor, such as celecoxib, was able to prevent the reduction in 5-HT levels, thus preventing the Aβ-induced depressive-like behavior and restoring Aβ plasma levels to control ( Mhillaj et al., 2018 ; Morgese et al., 2018a ). Accordingly, we have recently demonstrated that environmental factors, such as modified dietary factors, can lead to serotonergic impairment associated with increased levels of Aβ. In particular, we found that deficiency in polyunsaturated fatty acids of the omega 3 family, thus corresponding to a condition linked to a pseudoinflammatory state ( Solbrig et al., 2010 ; Graeber et al., 2011 ), led to a depressive-like phenotype characterized by reduced 5-HT content and higher Aβ levels ( Morgese et al., 2017 ). Accordingly, an anti-inflammatory diet, such as a diet enriched in omega 3 fatty acids, was able to prevent the reduction in 5-HT caused by Aβ injection, preventing the depressive phenomenon ( Bove et al., 2018 ; Morgese et al., 2018b ). Likewise, depressed patients showed higher risk for the development of AD ( Kessing and Andersen, 2004 ). On the other hand, postmortem studies performed in AD patients revealed low 5-HT and relative receptor content ( Reynolds et al., 1995 ). An in vitro model of familiar AD confirmed these observations, since cells overexpressing APP gene with the Swedish mutations associated with familial AD, indicated an altered sensitivity of the serotonergic system and 5-HT 1B receptor subtype in particular ( Tajeddinn et al., 2016 ). Furthermore, in a double transgenic model of early AD, fluoxetine, an antidepressant drug acting as serotonin-selective re-uptake inhibitors (SSRIs), ameliorated the impairment of spatial learning by preventing neuronal loss ( Ma et al., 2017 ) and delayed the cognitive decline associated with synaptic changes ( Zhou et al., 2018 ). Accordingly, clinical evidence revealed that SSRIs significantly improve depressant symptoms and daily activities in AD patients ( Werner and Covenas, 2015 ). This point is very intriguing considering that cognitive decline is recognized also as a clinical feature of depressive state. Interestingly, serotonergic system activation was reported to negatively modulate interstitial Aβ content. Indeed, in transgenic animal models of AD, the enhancing of 5-HT signaling, through the administration of SSRI antidepressants, rapidly reduced Aβ production in vivo via activation of extracellular regulated kinase (ERK) and the α-secretase-mediated pathway ( Cirrito et al., 2011 ; Fisher et al., 2016 ). Indeed, the sequential proteolytic cleavage of amyloid precursor protein (APP) can also occur via α-secretase, leading to the production of α-CTF later transformed by γ-secretase into AICD and p3 peptides ( Chow et al., 2010 ). This pathway is recognized as the non-amyloidogenic pathway since APP is cleaved by α-secretase in the Aβ region, yielding to lower Aβ production ( Chow et al., 2010 ). This pathway has been described as neurotrophic and neuroprotective ( Chow et al., 2010 ); therefore, therapeutic strategies steered at pushing APP processing toward α-secretase-mediated derivatives are under the spotlight. Furthermore, a PET imaging study carried out in cognitively normal individuals evidenced lower Aβ accumulation in consequence to increased 5-HT signaling ( Sheline et al., 2014 ), and retrospective analysis on patients under antidepressants further confirmed this finding ( Vlassenko et al., 2011 ). In this regard, we have recently demonstrated that fluoxetine treatment not only could restore 5-HT content in animals centrally injected with Aβ characterized by depressive-like phenotype but also reduced Aβ plasma levels ( Schiavone et al., 2017 ). In further agreement, activation of serotonergic receptors, such as 5-HT 4 , 5-HT 6 , and 5-HT 7 , corresponded to lower Aβ content, whereas the opposite effect was retrieved after simultaneous pharmacological blockade of 5-HT 4 and 5-HT 7 ( Cho and Hu, 2007 ; Fisher et al., 2016 ). 5-HT 4 partial agonists have been proposed as fast-acting antidepressants ( Lucas et al., 2007 ; Vidal et al., 2014 ) and have been shown to ameliorate cognitive deficit in anxiety/depressive models ( Darcet et al., 2016 ). In good agreement, pharmacological activation of 5-HT 4 receptors was shown to enhance short- and long-term memory function ( Meneses, 2007 ), endorsing the hypothesis of a putative role of these drugs for the amelioration of symptomatology of depression in AD. With regard to other receptor subtypes, it has been shown that APP can be released upon activation of 5-HT 2A and 5-HT 2C , and activation of 5-HT 2C receptor promotes the expression of neprilysin, a well-characterized Aβ degrading enzymes ( Tian et al., 2015 ). However, it should be considered that both 5-HT 2C agonists and antagonists have been evaluated as antidepressants ( Cryan and Lucki, 2000 ; Steardo et al., 2000 ; Cryan et al., 2005 ).

As regard to 5-HT 2A receptors, genetic polymorphisms have been described in AD patients affected by major depression ( Holmes et al., 2003 ) and, in AD patients, lower binding to these receptors has been identified ( Versijpt et al., 2003 ). In addition, intra-hippocampal injection of Aβ was associated with a significant reduction in 5-HT 2A expression ( Christensen et al., 2008 ). However, the effects of the activation of these receptors may vary depending on the cerebral pathway involved. Indeed, 5-HT 2A knocked down mice showed an altered phenotype with depressive-like symptoms ( Popa et al., 2005 ), and 5-HT 2A antagonists have been evaluated as antidepressants ( Zhang and Stackman, 2015 ); thus, a better understanding would help the developing of targeted compounds. On the other hand, 5-HT 6 receptors represent a novel therapeutic strategy in AD. Indeed, clinical trial for studying the efficacy and tolerability of the 5-HT 6 receptor antagonist, SB-742457, in subjects with mild-to-moderate and probable AD, revealed a safe profile and possible utility in improving cognitive symptoms of AD ( Maher-Edwards et al., 2010 ). However, antagonists of these receptor subtypes have been indicated as useful also in the treatment of non-cognitive symptoms associated with AD ( Garcia-Alloza et al., 2004 ). However, despite early positive findings, larger phase-III trials have failed to demonstrate any statistically significant impact on cognition for either idalopirdine or intepirdine, two 5-HT 6 antagonists, as adjunct to cholinesterase inhibitors. Paradoxically, 5-HT 6 receptor agonists also hold cognitive enhancing properties ( Khoury et al., 2018 ). Likewise, polymorphism of these receptors has been associated with altered response to antidepressant treatment in major depressive disorder ( Lee et al., 2005 ), although contrasting results have been reported ( Wu et al., 2001 ); hence, further research is warranted.

Noradrenergic System

The noradrenergic system is also implicated in the etiopathogenesis of both depression and AD. However, it has been recognized that the cause of depression is more complex than just an alteration in the levels of 5-HT and/or NA, being more directly caused by dysfunction in brain areas or neuronal systems modulated by monoamine systems ( Delgado and Moreno, 2000 ). It has been postulated that antidepressants, by enhancing neurotransmission in normal noradrenergic or serotonergic neurons, can restore lost functions in affected brain areas under monoamine control through a time-dependent process ( Delgado and Moreno, 2000 ). Indeed, noradrenergic and serotonergic systems are strictly interconnected and control each other via heteroreceptors. In particular, a negative feedback has been hypothesized considering that increased 5-HT levels correspond to NA release, which in turn inhibits further 5-HT release via α 2 AR activation ( Mongeau et al., 1997 ). This process is mediated through inhibitory α 2 receptors (α 2 AR) at 5-HT terminal levels and 5-HT 3 receptors at NA terminals. Interestingly, increased α 2 AR have been found in postmortem brains of depressed patients ( Meana et al., 1992 ; Ordway et al., 1994 ), and a theory of α 2 AR supersensitivity in depression was postulated early on Charney et al., 1981 . In this regard, increased α2-adrenoceptor density was retrieved in most regions of a rat model of depression, such as the flinders sensitive rat ( Lillethorup et al., 2015 ) and in patients with depressive disorders ( Cottingham and Wang, 2012 ). Interestingly, it has been postulated that tricyclic compounds can bind α2AR, thus functioning as arrestin-based ligands, and such an effect can explain their antidepressant property ( Cottingham et al., 2015 ). Βeta-arrestins are a small family of regulators of G protein-coupled receptors that regulate desensitization, internalization along, and initiation of their own signaling of such receptors ( Jiang et al., 2013 ). Long-term activation of these receptors causes endocytosis and downregulation through the recruitment of α 2 AR/arrestin complex ( Cottingham et al., 2015 ). The NA system is deeply affected also in neurodegeneration and in early AD ( Haglund et al., 2006 ). Indeed, α2A adrenergic receptors modulate APP endocytic sorting and promote Aβ generation through disrupting APP interaction with a vacuolar protein sorting (Vps10) family protein, a family of receptors that plays a decisive role in controlling the outcome of APP proteolytic processing ( Chen et al., 2014 ). In addition, this study pointed to the use of α2A antagonists as a new direction for AD treatment. In this light, another putative target for the generation of novel AD treatments is targeting β-arrestin. Indeed, increased β-arrestin 1 levels were shown in a transgenic animal model of AD as well as in postmortem study ( Liu et al., 2013 ). In keeping in mind a parallel route for depression and AD, β-arrestin signaling has also been associated with antidepressant properties of drugs ( Golan et al., 2013 ). Overexpression of β-arrestin 2 was associated with increased Aβ production. In particular, experimental conditions able to silence the β-arrestin 2 gene corresponded to Aβ rate of production by regulating γ-secretase activity ( Thathiah et al., 2013 ). Accordingly, Pontrello et al. found that the loss of dendritic spine in hippocampal neurons caused by Aβ was prevented by deleting β-arrestin-2 ( Pontrello et al., 2012 ). On the other hand, polymorphisms in the gene encoding for β2 adrenergic receptor have been associated with an increased risk of developing sporadic late onset AD ( Yu et al., 2008 ), while alterations in β adrenergic receptors were reported in depressed patients ( Mann et al., 1986 ). Indeed, much evidence indicates that activation of these receptors yield to antidepressant effects ( Overstreet et al., 2008 ; Gu et al., 2012 ). Nonetheless, Aβ interacts with the noradrenergic system directly binding to β-adrenergic receptors ( Igbavboa et al., 2006 ; Wang et al., 2011 ). Aβ may cause desensitization and subsequently internalization of β2 adrenergic receptors in prefrontal cortical neurons ( Wang et al., 2011 ). Furthermore, β2 adrenergic receptor activation mediates phosphorylation of tau after Aβ exposure both in vivo and in vitro ( Wang et al., 2013 ). On the other hand, we have found that central icv injection of Aβ increases noradrenergic tone after either 2 h or after 7 days from the central injection, probably reflecting a neuroprotective phenomenon ( Morgese et al., 2014 , 2015 ), considering that, NA is protective against neuroinflammatory processes. Accordingly, NA is able to modulate glial activation, and pharmacological strategies finalized to increase NA are considered a valid approach for neurodegenerative diseases ( Braun et al., 2014 ). In vitro studies have evidenced that neuroprotective effects of noradrenergic locus coeruleus (LC) afferents against Aβ rely on the stimulation of neurotrophic NGF and BDNF autocrine or paracrine loops via beta adrenoceptor activation of the cAMP response element binding protein pathway ( Counts and Mufson, 2010 ; Liu et al., 2015 ). After Aβ exposure, lower NA concentrations in LC projecting areas facilitate the inflammatory reaction of microglial cells, thus impairing microglial migration and phagocytosis, ultimately decreasing Aβ clearance ( Heneka et al., 2010 ). Accordingly, progression of AD is paralleled by the loss of noradrenergic function in LC ( Kelly et al., 2017 ), indicating the crucial role of this system in neurodegeneration.

Dopaminergic System

As regards the dopaminergic system, impairment of its neurotransmission has been implicated in many diseases including depression ( Schmidt et al., 2001 ), and several pre-clinical studies have indicated the involvement of dopaminergic, either D1, D2, or D3, in antidepressant effects ( Pytka et al., 2016 ). In good agreement, it has been shown that pure dopaminergic drugs, such as pramipexole, DA precursors, and DA reuptake inhibitors, show therapeutic efficacy in depression ( El Mansari et al., 2010 ; Belujon and Grace, 2017 ). In addition, neurodegenerative diseases associated with the loss of dopaminergic function, such as Parkinson’s or Huntington’s diseases, have high comorbidities with depression and anxiety ( Dale et al., 2016 ; Schrag and Taddei, 2017 ; Smeltere et al., 2017 ).

Concerning AD, it was shown that prefrontal cortical and hippocampal areas showed lower DA receptor expression ( Kemppainen et al., 2003 ; Kumar and Patel, 2007 ). Interestingly accumbal expression of D2-like receptors, dopaminergic transporter, and tyrosine hydroxylase enzyme was found altered in AD brains ( Rinne et al., 1986 ; Allard et al., 1990 ; Murray et al., 1995 ; Joyce et al., 1997 ). Imaging studies evidenced atrophy of this area in a cohort of AD patients ( Pievani et al., 2013 ). Aβ administration disrupts the cholinergic control of DA release, particularly in the nucleus accumbens ( Preda et al., 2008 ), but we also reported a blunting of DA release in the prefrontal cortex of rat after icv injection of the peptide ( Trabace et al., 2007 ). In addition, the increase in DAnergic tone has been proposed as a possible therapeutic strategy for AD, considering that dopaminergic dysfunction plays a pathogenic role in cognitive decline ( Martorana et al., 2009 , 2013 ; Koch et al., 2014 ; Martorana and Koch, 2014 ). Furthermore, selective DAnergic neuronal degeneration in ventral tegmental area was demonstrated in AD transgenic mice at pre-plaque stages, suggesting that lower hippocampal and accumbal DA outflow correlate to memory deficits and dysfunction of reward processing ( Nobili et al., 2017 ).

Conclusions

It has been reported that depressed individuals are nearly twice as likely to develop dementia, often in the form of AD, compared with non-depressed individuals. Unfortunately, few pharmacological tools are available for dementia; thus, the need for novel therapeutic strategies is very compelling. Future studies aimed at elucidating the mechanisms through which drugs modulating monoamine release may prove helpful in individuating novel strategy for slowing down cognitive impairment in pre-clinical AD phase, often associated with mood alterations, taking into account their effects on Aβ production/clearance, aggregation status, and neuroinflammatory-induced pathways. Furthermore, some of these molecules are already commercialized; thus, such a novel potential therapeutic approach for AD treatment may become rapidly clinically suitable.

Author Contributions

MM and LT helped in study design, drafting, revising, and accepting of the final version of the manuscript.

Funding

This work was supported by Intervento cofinanziato dal Fondo di Sviluppo e Coesione 2007-2013–APQ Ricerca Regione Puglia “ Programma regionale a sostegno della specializzazione intelligente e della sostenibilità sociale ed ambientale–FutureInResearch”, Italy to MM (code OC970P6) and by PRIN 2015 code 2015XSZ9A2_005 to LT.

Conflict of Interest Statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

Allard, P., Alafuzoff, I., Carlsson, A., Eriksson, K., Ericson, E., Gottfries, C. G., et al. (1990). Loss of dopamine uptake sites labeled with [3H]GBR-12935 in Alzheimer’s disease. Eur. Neurol. 30, 181–185.

Belujon, P., and Grace, A. A. (2017). Dopamine system dysregulation in major depressive disorders. Int. J. Neuropsychopharmacol. 20, 1036–1046. doi: 10. 1093/ijnp/pyx056

||

Bove, M., Mhillaj, E., Tucci, P., Giardino, I., Schiavone, S., Morgese, M. G., et al. (2018). Effects of n-3 PUFA enriched and n-3 PUFA deficient diets in naïve and Aβ-treated female rats. Biochem. Pharmacol. 155, 326–335. doi: 10. 1016/j. bcp. 2018. 07. 017

||

Braun, D., Madrigal, J. L., and Feinstein, D. L. (2014). Noradrenergic regulation of glial activation: molecular mechanisms and therapeutic implications. Curr. Neuropharmacol. 12, 342–352. doi: 10. 2174/1570159X12666140828220938

||

Charney, D. S., Menkes, D. B., and Heninger, G. R. (1981). Receptor sensitivity and the mechanism of action of antidepressant treatment. Implications for the etiology and therapy of depression. Arch. Gen. Psychiatry 38, 1160–1180. doi: 10. 1001/archpsyc. 1981. 01780350094011

||

Chen, Y., Peng, Y., Che, P., Gannon, M., Liu, Y., Li, L., et al. (2014). Alpha(2A) adrenergic receptor promotes amyloidogenesis through disrupting APP-SorLA interaction. Proc. Natl. Acad. Sci. USA 111, 17296–17301. doi: 10. 1073/pnas. 1409513111

|

Cho, S., and Hu, Y. (2007). Activation of 5-HT4 receptors inhibits secretion of beta-amyloid peptides and increases neuronal survival. Exp. Neurol. 203, 274–278. doi: 10. 1016/j. expneurol. 2006. 07. 021

||

Chow, V. W., Mattson, M. P., Wong, P. C., and Gleichmann, M. (2010). An overview of APP processing enzymes and products. NeuroMolecular Med. 12, 1–12. doi: 10. 1007/s12017-009-8104-z

|

Christensen, D. Z., Kraus, S. L., Flohr, A., Cotel, M. C., Wirths, O., and Bayer, T. A. (2008). Transient intraneuronal A beta rather than extracellular plaque pathology correlates with neuron loss in the frontal cortex of APP/PS1KI mice. Acta Neuropathol. 116, 647–655. doi: 10. 1007/s00401-008-0451-6

||

Cirrito, J. R., Disabato, B. M., Restivo, J. L., Verges, D. K., Goebel, W. D., Sathyan, A., et al. (2011). Serotonin signaling is associated with lower amyloid-beta levels and plaques in transgenic mice and humans. Proc. Natl. Acad. Sci. USA 108, 14968–14973. doi: 10. 1073/pnas. 1107411108

||

Cleary, J. P., Walsh, D. M., Hofmeister, J. J., Shankar, G. M., Kuskowski, M. A., Selkoe, D. J., et al. (2005). Natural oligomers of the amyloid-beta protein specifically disrupt cognitive function. Nat. Neurosci. 8, 79–84. doi: 10. 1038/nn1372

||

Colaianna, M., Tucci, P., Zotti, M., Morgese, M. G., Schiavone, S., Govoni, S., et al. (2010). Soluble beta amyloid(1-42): a critical player in producing behavioural and biochemical changes evoking depressive-related state? Br. J. Pharmacol. 159, 1704–1715. doi: 10. 1111/j. 1476-5381. 2010. 00669. x

||

Cottingham, C., Ferryman, C. J., and Wang, Q. (2015). Alpha2 adrenergic receptor trafficking as a therapeutic target in antidepressant drug action. Prog. Mol. Biol. Transl. Sci. 132, 207–225. doi: 10. 1016/bs. pmbts. 2015. 03. 007

||

Cottingham, C., and Wang, Q. (2012). Alpha2 adrenergic receptor dysregulation in depressive disorders: implications for the neurobiology of depression and antidepressant therapy. Neurosci. Biobehav. Rev. 36, 2214–2225. doi: 10. 1016/j. neubiorev. 2012. 07. 011

||

Counts, S. E., and Mufson, E. J. (2010). Noradrenaline activation of neurotrophic pathways protects against neuronal amyloid toxicity. J. Neurochem. 113, 649–660. doi: 10. 1111/j. 1471-4159. 2010. 06622. x

||

Cryan, J. F., and Lucki, I. (2000). Antidepressant-like behavioral effects mediated by 5-Hydroxytryptamine(2C) receptors. J. Pharmacol. Exp. Ther. 295, 1120–1126.

|

Cryan, J. F., Valentino, R. J., and Lucki, I. (2005). Assessing substrates underlying the behavioral effects of antidepressants using the modified rat forced swimming test. Neurosci. Biobehav. Rev. 29, 547–569. doi: 10. 1016/j. neubiorev. 2005. 03. 008

||

Dale, M., Maltby, J., Shimozaki, S., Cramp, R., and Rickards, H. REGISTRY Investigators of the European Huntington’s Disease Network (2016). Disease stage, but not sex, predicts depression and psychological distress in Huntington’s disease: a European population study. J. Psychosom. Res. 80, 17–22. doi: 10. 1016/j. jpsychores. 2015. 11. 003

||

Darcet, F., Gardier, A. M., David, D. J., and Guilloux, J. P. (2016). Chronic 5-HT4 receptor agonist treatment restores learning and memory deficits in a neuroendocrine mouse model of anxiety/depression. Neurosci. Lett. 616, 197–203. doi: 10. 1016/j. neulet. 2016. 01. 055

||

Delgado, P. L., and Moreno, F. A. (2000). Role of norepinephrine in depression. J. Clin. Psychiatry. 61(Suppl. 1), 5–12.

Egashira, N., Iwasaki, K., Takashima, A., Watanabe, T., Kawabe, H., Matsuda, T., et al. (2005). Altered depression-related behavior and neurochemical changes in serotonergic neurons in mutant R406W human tau transgenic mice. Brain Res. 1059, 7–12. doi: 10. 1016/j. brainres. 2005. 08. 004

||

El Mansari, M., Guiard, B. P., Chernoloz, O., Ghanbari, R., Katz, N., and Blier, P. (2010). Relevance of norepinephrine-dopamine interactions in the treatment of major depressive disorder. CNS Neurosci. Ther. 16, e1–e17. doi: 10. 1111/j. 1755-5949. 2010. 00146. x

||

Fisher, J. R., Wallace, C. E., Tripoli, D. L., Sheline, Y. I., and Cirrito, J. R. (2016). Redundant Gs-coupled serotonin receptors regulate amyloid-beta metabolism in vivo. Mol. Neurodegener. 11: 45. doi: 10. 1186/s13024-016-0112-5

||

Garcia-Alloza, M., Hirst, W. D., Chen, C. P., Lasheras, B., Francis, P. T., and Ramirez, M. J. (2004). Differential involvement of 5-HT(1B/1D) and 5-HT6 receptors in cognitive and non-cognitive symptoms in Alzheimer’s disease. Neuropsychopharmacology 29, 410–416. doi: 10. 1038/sj. npp. 1300330

||

Geerlings, M. I., Schoevers, R. A., Beekman, A. T., Jonker, C., Deeg, D. J., Schmand, B., et al. (2000). Depression and risk of cognitive decline and Alzheimer’s disease. Results of two prospective community-based studies in The Netherlands. Br. J. Psychiatry 176, 568–575. doi: 10. 1192/bjp. 176. 6. 568

||

Golan, M., Schreiber, G., and Avissar, S. (2013). Antidepressant-induced differential ubiquitination of beta-arrestins 1 and 2 in mononuclear leucocytes of patients with depression. Int. J. Neuropsychopharmacol. 16, 1745–1754. doi: 10. 1017/S1461145713000291

||

Graeber, M. B., Li, W., and Rodriguez, M. L. (2011). Role of microglia in CNS inflammation. FEBS Lett. 585, 3798–3805. doi: 10. 1016/j. febslet. 2011. 08. 033

|

Gu, Y., Schupf, N., Cosentino, S. A., Luchsinger, J. A., and Scarmeas, N. (2012). Nutrient intake and plasma beta-amyloid. Neurology 78, 1832–1840. doi: 10. 1212/WNL. 0b013e318258f7c2

|

Haglund, M., Sjobeck, M., and Englund, E. (2006). Locus ceruleus degeneration is ubiquitous in Alzheimer’s disease: possible implications for diagnosis and treatment. Neuropathology 26, 528–532. doi: 10. 1111/j. 1440-1789. 2006. 00725. x

||

Hardy, J., and Selkoe, D. J. (2002). The amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics. Science 297, 353–356. doi: 10. 1126/science. 1072994

||

Heneka, M. T., Nadrigny, F., Regen, T., Martinez-Hernandez, A., Dumitrescu-Ozimek, L., Terwel, D., et al. (2010). Locus ceruleus controls Alzheimer’s disease pathology by modulating microglial functions through norepinephrine. Proc. Natl. Acad. Sci. USA 107, 6058–6063. doi: 10. 1073/pnas. 0909586107

|

Holmes, C., Arranz, M., Collier, D., Powell, J., and Lovestone, S. (2003). Depression in Alzheimer’s disease: the effect of serotonin receptor gene variation. Am. J. Med. Genet. B Neuropsychiatr. Genet. 119B, 40–43. doi: 10. 1002/ajmg. b. 10068

||

Igbavboa, U., Johnson-Anuna, L. N., Rossello, X., Butterick, T. A., Sun, G. Y., and Wood, W. G. (2006). Amyloid beta-protein1-42 increases cAMP and apolipoprotein E levels which are inhibited by beta1 and beta2-adrenergic receptor antagonists in mouse primary astrocytes. Neuroscience 142, 655–660. doi: 10. 1016/j. neuroscience. 2006. 06. 056

||

Jiang, Z., Cowell, R. M., and Nakazawa, K. (2013). Convergence of genetic and environmental factors on parvalbumin-positive interneurons in schizophrenia. Front. Behav. Neurosci. 7: 116. doi: 10. 3389/fnbeh. 2013. 00116

|

Joyce, J. N., Smutzer, G., Whitty, C. J., Myers, A., and Bannon, M. J. (1997). Differential modification of dopamine transporter and tyrosine hydroxylase mRNAs in midbrain of subjects with Parkinson’s, Alzheimer’s with parkinsonism, and Alzheimer’s disease. Mov. Disord. 12, 885–897. doi: 10. 1002/mds. 870120609

||

Kelly, S. C., He, B., Perez, S. E., Ginsberg, S. D., Mufson, E. J., and Counts, S. E. (2017). Locus coeruleus cellular and molecular pathology during the progression of Alzheimer’s disease. Acta Neuropathol. Commun. 5: 8. doi: 10. 1186/s40478-017-0411-2

||

Kemppainen, N., Laine, M., Laakso, M. P., Kaasinen, V., Nagren, K., Vahlberg, T., et al. (2003). Hippocampal dopamine D2 receptors correlate with memory functions in Alzheimer’s disease. Eur. J. Neurosci. 18, 149–154. doi: 10. 1046/j. 1460-9568. 2003. 02716. x

||

Kepe, V., Barrio, J. R., Huang, S. C., Ercoli, L., Siddarth, P., Shoghi-Jadid, K., et al. (2006). Serotonin 1A receptors in the living brain of Alzheimer’s disease patients. Proc. Natl. Acad. Sci. USA 103, 702–707. doi: 10. 1073/pnas. 0510237103

|

Kessing, L. V., and Andersen, P. K. (2004). Does the risk of developing dementia increase with the number of episodes in patients with depressive disorder and in patients with bipolar disorder? J. Neurol. Neurosurg. Psychiatry 75, 1662–1666. doi: 10. 1136/jnnp. 2003. 031773

||

Khoury, R., Grysman, N., Gold, J., Patel, K., and Grossberg, G. T. (2018). The role of 5 HT6-receptor antagonists in Alzheimer’s disease: an update. Expert Opin. Investig. Drugs 27, 523–533. doi: 10. 1080/13543784. 2018. 1483334

|

Koch, G., Di Lorenzo, F., Bonni, S., Giacobbe, V., Bozzali, M., Caltagirone, C., et al. (2014). Dopaminergic modulation of cortical plasticity in Alzheimer’s disease patients. Neuropsychopharmacology 39, 2654–2661. doi: 10. 1038/npp. 2014. 119

||

Krishnan, V., and Nestler, E. J. (2008). The molecular neurobiology of depression. Nature 455, 894–902. doi: 10. 1038/nature07455

||

Kumar, U., and Patel, S. C. (2007). Immunohistochemical localization of dopamine receptor subtypes (D1R-D5R) in Alzheimer’s disease brain. Brain Res. 1131, 187–196. doi: 10. 1016/j. brainres. 2006. 10. 049

||

Lanctot, K. L., Herrmann, N., and Mazzotta, P. (2001). Role of serotonin in the behavioral and psychological symptoms of dementia. J. Neuropsychiatr. Clin. Neurosci. 13, 5–21. doi: 10. 1176/jnp. 13. 1. 5

||

Lee, S. H., Lee, K. J., Lee, H. J., Ham, B. J., Ryu, S. H., and Lee, M. S. (2005). Association between the 5-HT6 receptor C267T polymorphism and response to antidepressant treatment in major depressive disorder. Psychiatry Clin. Neurosci. 59, 140–145. doi: 10. 1111/j. 1440-1819. 2005. 01348. x

||

Lillethorup, T. P., Iversen, P., Wegener, G., Doudet, D. J., and Landau, A. M. (2015). Alpha2-adrenoceptor binding in Flinders-sensitive line compared with Flinders-resistant line and Sprague-Dawley rats. Acta Neuropsychiatr. 27, 345–352. doi: 10. 1017/neu. 2015. 24

||

Liu, X., Ye, K., and Weinshenker, D. (2015). Norepinephrine protects against amyloid-beta toxicity via TrkB. J. Alzheimers Dis. 44, 251–260. doi: 10. 3233/JAD-141062

||

Liu, X., Zhao, X., Zeng, X., Bossers, K., Swaab, D. F., Zhao, J., et al. (2013). Beta-arrestin1 regulates gamma-secretase complex assembly and modulates amyloid-beta pathology. Cell Res. 23, 351–365. doi: 10. 1038/cr. 2012. 167

||

Lucas, G., Rymar, V. V., Du, J., Mnie-Filali, O., Bisgaard, C., Manta, S., et al. (2007). Serotonin(4) (5-HT(4)) receptor agonists are putative antidepressants with a rapid onset of action. Neuron 55, 712–725. doi: 10. 1016/j. neuron. 2007. 07. 041

||

Ma, J., Gao, Y., Jiang, L., Chao, F. L., Huang, W., Zhou, C. N., et al. (2017). Fluoxetine attenuates the impairment of spatial learning ability and prevents neuron loss in middle-aged APPswe/PSEN1dE9 double transgenic Alzheimer’s disease mice. Oncotarget 8, 27676–27692. doi: 10. 18632/oncotarget. 15398

||

Maher-Edwards, G., Zvartau-Hind, M., Hunter, A. J., Gold, M., Hopton, G., Jacobs, G., et al. (2010). Double-blind, controlled phase II study of a 5-HT6 receptor antagonist, SB-742457, in Alzheimer’s disease. Curr. Alzheimer Res. 7, 374–385. doi: 10. 2174/156720510791383831

||

Mann, J. J. (1999). Role of the serotonergic system in the pathogenesis of major depression and suicidal behavior. Neuropsychopharmacology 21, 99S–105S. doi: 10. 1016/S0893-133X(99)00040-8

||

Mann, J. J., Mcbride, P. A., and Stanley, M. (1986). Postmortem serotonergic and adrenergic receptor binding to frontal cortex: correlations with suicide. Psychopharmacol. Bull. 22, 647–649.

|

Martorana, A., Di Lorenzo, F., Esposito, Z., Lo Giudice, T., Bernardi, G., Caltagirone, C., et al. (2013). Dopamine D(2)-agonist rotigotine effects on cortical excitability and central cholinergic transmission in Alzheimer’s disease patients. Neuropharmacology 64, 108–113. doi: 10. 1016/j. neuropharm. 2012. 07. 015

||

Martorana, A., and Koch, G. (2014). Is dopamine involved in Alzheimer’s disease? Front. Aging Neurosci. 6: 252. doi: 10. 3389/fnagi. 2014. 00252

|

Martorana, A., Mori, F., Esposito, Z., Kusayanagi, H., Monteleone, F., Codeca, C., et al. (2009). Dopamine modulates cholinergic cortical excitability in Alzheimer’s disease patients. Neuropsychopharmacology 34, 2323–2328. doi: 10. 1038/npp. 2009. 60

||

Mattson, M. P. (2004). Pathways towards and away from Alzheimer’s disease. Nature 430, 631–639. doi: 10. 1038/nature02621

||

Meana, J. J., Barturen, F., and Garcia-Sevilla, J. A. (1992). Alpha 2-adrenoceptors in the brain of suicide victims: increased receptor density associated with major depression. Biol. Psychiatry 31, 471–490. doi: 10. 1016/0006-3223(92)90259-3

||

Meneses, A. (2007). Stimulation of 5-HT1A, 5-HT1B, 5-HT2A/2C, 5-HT3 and 5-HT4 receptors or 5-HT uptake inhibition: short- and long-term memory. Behav. Brain Res. 184, 81–90. doi: 10. 1016/j. bbr. 2007. 06. 026

||

Mhillaj, E., Morgese, M. G., Tucci, P., Furiano, A., Luongo, L., Bove, M., et al. (2018). Celecoxib prevents cognitive impairment and neuroinflammation in soluble amyloid beta-treated rats. Neuroscience 372, 58–73. doi: 10. 1016/j. neuroscience. 2017. 12. 046

||

Modrego, P. J., and Ferrandez, J. (2004). Depression in patients with mild cognitive impairment increases the risk of developing dementia of Alzheimer type: a prospective cohort study. Arch. Neurol. 61, 1290–1293. doi: 10. 1001/archneur. 61. 8. 1290

||

Mongeau, R., Blier, P., and De Montigny, C. (1997). The serotonergic and noradrenergic systems of the hippocampus: their interactions and the effects of antidepressant treatments. Brain Res. Brain Res. Rev. 23, 145–195. doi: 10. 1016/S0165-0173(96)00017-3

||

Morgan, D. G., May, P. C., and Finch, C. E. (1987). Dopamine and serotonin systems in human and rodent brain: effects of age and neurodegenerative disease. J. Am. Geriatr. Soc. 35, 334–345.

Morgese, M. G., Colaianna, M., Mhillaj, E., Zotti, M., Schiavone, S., D’antonio, P., et al. (2015). Soluble beta amyloid evokes alteration in brain norepinephrine levels: role of nitric oxide and interleukin-1. Front. Neurosci. 9: 428. doi: 10. 3389/fnins. 2015. 00428

||

Morgese, M. G., Schiavone, S., Bove, M., Mhillaj, E., Tucci, P., and Trabace, L. (2018a). Sub-chronic celecoxib prevents soluble beta amyloid-induced depressive-like behaviour in rats. J. Affect. Disord. 238, 118–121. doi: 10. 1016/j. jad. 2018. 05. 030

|

Morgese, M. G., Schiavone, S., Mhillaj, E., Bove, M., Tucci, P., and Trabace, L. (2018b). N-3 PUFA diet enrichment prevents amyloid beta-induced depressive-like phenotype. Pharmacol. Res. 129, 526–534. doi: 10. 1016/j. phrs. 2017. 11. 034

|

Morgese, M. G., Tucci, P., Colaianna, M., Zotti, M., Cuomo, V., Schiavone, S., et al. (2014). Modulatory activity of soluble beta amyloid on HPA axis function in rats. Curr. Pharm. Des. 20, 2539–2546. doi: 10. 2174/13816128113199990500

||

Morgese, M. G., Tucci, P., Mhillaj, E., Bove, M., Schiavone, S., Trabace, L., et al. (2017). Lifelong nutritional omega-3 deficiency evokes depressive-like state through soluble beta amyloid. Mol. Neurobiol. 54, 2079–2089. doi: 10. 1007/s12035-016-9809-2

||

Murray, A. M., Weihmueller, F. B., Marshall, J. F., Hurtig, H. I., Gottleib, G. L., and Joyce, J. N. (1995). Damage to dopamine systems differs between Parkinson’s disease and Alzheimer’s disease with parkinsonism. Ann. Neurol. 37, 300–312. doi: 10. 1002/ana. 410370306

||

Nobili, A., Latagliata, E. C., Viscomi, M. T., Cavallucci, V., Cutuli, D., Giacovazzo, G., et al. (2017). Dopamine neuronal loss contributes to memory and reward dysfunction in a model of Alzheimer’s disease. Nat. Commun. 8: 14727. doi: 10. 1038/ncomms14727

|

Ordway, G. A., Widdowson, P. S., Smith, K. S., and Halaris, A. (1994). Agonist binding to alpha 2-adrenoceptors is elevated in the locus coeruleus from victims of suicide. J. Neurochem. 63, 617–624.

|

Overstreet, D. H., Stemmelin, J., and Griebel, G. (2008). Confirmation of antidepressant potential of the selective beta3 adrenoceptor agonist amibegron in an animal model of depression. Pharmacol. Biochem. Behav. 89, 623–626. doi: 10. 1016/j. pbb. 2008. 02. 020

||

Ownby, R. L., Crocco, E., Acevedo, A., John, V., and Loewenstein, D. (2006). Depression and risk for Alzheimer disease: systematic review, meta-analysis, and metaregression analysis. Arch. Gen. Psychiatry 63, 530–538. doi: 10. 1001/archpsyc. 63. 5. 530

||

Pievani, M., Bocchetta, M., Boccardi, M., Cavedo, E., Bonetti, M., Thompson, P. M., et al. (2013). Striatal morphology in early-onset and late-onset alzheimer’s disease: a preliminary study. Neurobiol. Aging 34, 1728–1739. doi: 10. 1016/j. neurobiolaging. 2013. 01. 016

||

Pomara, N., and Sidtis, J. J. (2010). Brain neurotoxic amyloid-beta peptides: their potential role in the pathophysiology of depression and as molecular therapeutic targets. Br. J. Pharmacol. 161, 768–770. doi: 10. 1111/j. 1476-5381. 2010. 00948. x

||

Pontrello, C. G., Sun, M. Y., Lin, A., Fiacco, T. A., Defea, K. A., and Ethell, I. M. (2012). Cofilin under control of beta-arrestin-2 in NMDA-dependent dendritic spine plasticity, long-term depression (LTD), and learning. Proc. Natl. Acad. Sci. USA 109, E442–E451. doi: 10. 1073/pnas. 1118803109

|

Popa, D., Lena, C., Fabre, V., Prenat, C., Gingrich, J., Escourrou, P., et al. (2005). Contribution of 5-HT2 receptor subtypes to sleep-wakefulness and respiratory control, and functional adaptations in knock-out mice lacking 5-HT2A receptors. J. Neurosci. 25, 11231–11238. doi: 10. 1523/JNEUROSCI. 1724-05. 2005

||

Preda, S., Govoni, S., Lanni, C., Racchi, M., Mura, E., Grilli, M., et al. (2008). Acute beta-amyloid administration disrupts the cholinergic control of dopamine release in the nucleus accumbens. Neuropsychopharmacology 33, 1062–1070. doi: 10. 1038/sj. npp. 1301485

||

Pytka, K., Podkowa, K., Rapacz, A., Podkowa, A., Zmudzka, E., Olczyk, A., et al. (2016). The role of serotonergic, adrenergic and dopaminergic receptors in antidepressant-like effect. Pharmacol. Rep. 68, 263–274. doi: 10. 1016/j. pharep. 2015. 08. 007

||

Reynolds, G. P., Mason, S. L., Meldrum, A., De Keczer, S., Parnes, H., Eglen, R. M., et al. (1995). 5-Hydroxytryptamine (5-HT)4 receptors in post mortem human brain tissue: distribution, pharmacology and effects of neurodegenerative diseases. Br. J. Pharmacol. 114, 993–998. doi: 10. 1111/j. 1476-5381. 1995. tb13303. x

||

Rinne, J. O., Sako, E., Paljarvi, L., Molsa, P. K., and Rinne, U. K. (1986). Brain dopamine D-1 receptors in senile dementia. J. Neurol. Sci. 73, 219–230.

Rowan, M. J., Klyubin, I., Wang, Q., and Anwyl, R. (2005). Synaptic plasticity disruption by amyloid beta protein: modulation by potential Alzheimer’s disease modifying therapies. Biochem. Soc. Trans. 33, 563–567. doi: 10. 1042/BST0330563

||

Schiavone, S., Tucci, P., Mhillaj, E., Bove, M., Trabace, L., and Morgese, M. G. (2017). Antidepressant drugs for beta amyloid-induced depression: a new standpoint? Prog. Neuro-Psychopharmacol. Biol. Psychiatry 78, 114–122. doi: 10. 1016/j. pnpbp. 2017. 05. 004

||

Schildkraut, J. J. (1995). The catecholamine hypothesis of affective disorders: a review of supporting evidence. 1965. J. Neuropsychiatr. Clin. Neurosci. 7, 524–533. discussion: 523-524. doi: 10. 1176/jnp. 7. 4. 524

|

Schmidt, K., Nolte-Zenker, B., Patzer, J., Bauer, M., Schmidt, L. G., and Heinz, A. (2001). Psychopathological correlates of reduced dopamine receptor sensitivity in depression, schizophrenia, and opiate and alcohol dependence. Pharmacopsychiatry 34, 66–72. doi: 10. 1055/s-2001-15184

||

Schrag, A., and Taddei, R. N. (2017). Depression and anxiety in Parkinson’s disease. Int. Rev. Neurobiol. 133, 623–655. doi: 10. 1016/bs. irn. 2017. 05. 024

|

Selkoe, D. J., and Schenk, D. (2003). Alzheimer’s disease: molecular understanding predicts amyloid-based therapeutics. Annu. Rev. Pharmacol. Toxicol. 43, 545–584. doi: 10. 1146/annurev. pharmtox. 43. 100901. 140248

||

Sheline, Y. I., West, T., Yarasheski, K., Swarm, R., Jasielec, M. S., Fisher, J. R., et al. (2014). An antidepressant decreases CSF Abeta production in healthy individuals and in transgenic AD mice. Sci. Transl. Med. 6: 236re234. doi: 10. 1126/scitranslmed. 3008169

||

Smeltere, L., Kuznecovs, V., and Erts, R. (2017). Depression and social phobia in essential tremor and Parkinson’s disease. Brain Behav. 7: e00781. doi: 10. 1002/brb3. 781

||

Solbrig, M. V., Fan, Y., Hermanowicz, N., Morgese, M. G., and Giuffrida, A. (2010). A synthetic cannabinoid agonist promotes oligodendrogliogenesis during viral encephalitis in rats. Exp. Neurol. 226, 231–241. doi: 10. 1016/j. expneurol. 2010. 09. 003

||

Steardo, L., Monteleone, P., Trabace, L., Cannizzaro, C., Maj, M., and Cuomo, V. (2000). Serotonergic modulation of rat pineal gland activity: in vivo evidence for a 5-Hydroxytryptamine(2C) receptor involvement. J. Pharmacol. Exp. Ther. 295, 266–273.

|

Sun, X., Steffens, D. C., Au, R., Folstein, M., Summergrad, P., Yee, J., et al. (2008). Amyloid-associated depression: a prodromal depression of Alzheimer disease? Arch. Gen. Psychiatry 65, 542–550. doi: 10. 1001/archpsyc. 65. 5. 542

||

Tajeddinn, W., Persson, T., Calvo-Garrido, J., Seed Ahmed, M., Maioli, S., Vijayaraghavan, S., et al. (2016). Pharmacological modulations of the serotonergic system in a cell-model of familial Alzheimer’s disease. J. Alzheimers Dis. 53, 349–361. doi: 10. 3233/JAD-160046

||

Thathiah, A., Horre, K., Snellinx, A., Vandewyer, E., Huang, Y., Ciesielska, M., et al. (2013). beta-arrestin 2 regulates Abeta generation and gamma-secretase activity in Alzheimer’s disease. Nat. Med. 19, 43–49. doi: 10. 1038/nm. 3023

||

Tian, X. L., Yu, L. H., Li, W. Q., Hu, Y., Yin, M., and Wang, Z. J. (2015). Activation of 5-HT(2C) receptor promotes the expression of neprilysin in U251 human glioma cells. Cell. Mol. Neurobiol. 35, 425–432. doi: 10. 1007/s10571-014-0138-6

||

Trabace, L., Kendrick, K. M., Castrignano, S., Colaianna, M., De Giorgi, A., Schiavone, S., et al. (2007). Soluble amyloid beta1-42 reduces dopamine levels in rat prefrontal cortex: relationship to nitric oxide. Neuroscience 147, 652–663. doi: 10. 1016/j. neuroscience. 2007. 04. 056

||

Tucci, P., Mhillaj, E., Morgese, M. G., Colaianna, M., Zotti, M., Schiavone, S., et al. (2014). Memantine prevents memory consolidation failure induced by soluble beta amyloid in rats. Front. Behav. Neurosci. 8: 332. doi: 10. 3389/fnbeh. 2014. 00332

||

Versijpt, J., Van Laere, K. J., Dumont, F., Decoo, D., Vandecapelle, M., Santens, P., et al. (2003). Imaging of the 5-HT2A system: age-, gender-, and Alzheimer’s disease-related findings. Neurobiol. Aging 24, 553–561. doi: 10. 1016/S0197-4580(02)00137-9

||

Vidal, R., Castro, E., Pilar-Cuellar, F., Pascual-Brazo, J., Diaz, A., Rojo, M. L., et al. (2014). Serotonin 5-HT4 receptors: a new strategy for developing fast acting antidepressants? Curr. Pharm. Des. 20, 3751–3762. doi: 10. 2174/13816128113196660734

||

Visser, P. J., Verhey, F. R., Ponds, R. W., Kester, A., and Jolles, J. (2000). Distinction between preclinical Alzheimer’s disease and depression. J. Am. Geriatr. Soc. 48, 479–484. doi: 10. 1111/j. 1532-5415. 2000. tb04992. x

|

Vlassenko, A. G., Mintun, M. A., Xiong, C., Sheline, Y. I., Goate, A. M., Benzinger, T. L., et al. (2011). Amyloid-beta plaque growth in cognitively normal adults: longitudinal [11C]Pittsburgh compound B data. Ann. Neurol. 70, 857–861. doi: 10. 1002/ana. 22608

||

Wang, D., Fu, Q., Zhou, Y., Xu, B., Shi, Q., Igwe, B., et al. (2013). Beta2 adrenergic receptor, protein kinase A (PKA) and c-Jun N-terminal kinase (JNK) signaling pathways mediate tau pathology in Alzheimer disease models. J. Biol. Chem. 288, 10298–10307. doi: 10. 1074/jbc. M112. 415141

||

Wang, D., Yuen, E. Y., Zhou, Y., Yan, Z., and Xiang, Y. K. (2011). Amyloid beta peptide-(1-42) induces internalization and degradation of beta2 adrenergic receptors in prefrontal cortical neurons. J. Biol. Chem. 286, 31852–31863. doi: 10. 1074/jbc. M111. 244335

||

Werner, F. M., and Covenas, R. (2015). Review: classical neurotransmitters and neuropeptides involved in generalized epilepsy in a multi-neurotransmitter system: how to improve the antiepileptic effect? Epilepsy Behav. 71, 124–129. doi: 10. 1016/j. yebeh. 2015. 01. 038

|

Wu, W. H., Huo, S. J., Cheng, C. Y., Hong, C. J., and Tsai, S. J. (2001). Association study of the 5-HT(6) receptor polymorphism (C267T) and symptomatology and antidepressant response in major depressive disorders. Neuropsychobiology 44, 172–175. doi: 10. 1159/000054938

||

Yu, J. T., Tan, L., Ou, J. R., Zhu, J. X., Liu, K., Song, J. H., et al. (2008). Polymorphisms at the beta2-adrenergic receptor gene influence Alzheimer’s disease susceptibility. Brain Res. 1210, 216–222. doi: 10. 1016/j. brainres. 2008. 03. 019

||

Zhang, G., and Stackman, R. W. Jr. (2015). The role of serotonin 5-HT2A receptors in memory and cognition. Front. Pharmacol. 6: 225. doi: 10. 3389/fphar. 2015. 00225

|

Zhou, C. N., Chao, F. L., Zhang, Y., Jiang, L., Zhang, L., Fan, J. H., et al. (2018). Fluoxetine delays the cognitive function decline and synaptic changes in a transgenic mouse model of early Alzheimer’s disease. J. Comp. Neurol. 527, 1378–1387. doi: 10. 1002/cne. 24616

|

Thank's for Your Vote!
Monoaminergic system modulation in depression and alzheimer’s disease: a new standpoint?. Page 1
Monoaminergic system modulation in depression and alzheimer’s disease: a new standpoint?. Page 2
Monoaminergic system modulation in depression and alzheimer’s disease: a new standpoint?. Page 3
Monoaminergic system modulation in depression and alzheimer’s disease: a new standpoint?. Page 4
Monoaminergic system modulation in depression and alzheimer’s disease: a new standpoint?. Page 5
Monoaminergic system modulation in depression and alzheimer’s disease: a new standpoint?. Page 6
Monoaminergic system modulation in depression and alzheimer’s disease: a new standpoint?. Page 7
Monoaminergic system modulation in depression and alzheimer’s disease: a new standpoint?. Page 8
Monoaminergic system modulation in depression and alzheimer’s disease: a new standpoint?. Page 9

This work, titled "Monoaminergic system modulation in depression and alzheimer’s disease: a new standpoint?" was written and willingly shared by a fellow student. This sample can be utilized as a research and reference resource to aid in the writing of your own work. Any use of the work that does not include an appropriate citation is banned.

If you are the owner of this work and don’t want it to be published on AssignBuster, request its removal.

Request Removal
Cite this Essay

References

AssignBuster. (2021) 'Monoaminergic system modulation in depression and alzheimer’s disease: a new standpoint'. 20 December.

Reference

AssignBuster. (2021, December 20). Monoaminergic system modulation in depression and alzheimer’s disease: a new standpoint? Retrieved from https://assignbuster.com/monoaminergic-system-modulation-in-depression-and-alzheimers-disease-a-new-standpoint/

References

AssignBuster. 2021. "Monoaminergic system modulation in depression and alzheimer’s disease: a new standpoint?" December 20, 2021. https://assignbuster.com/monoaminergic-system-modulation-in-depression-and-alzheimers-disease-a-new-standpoint/.

1. AssignBuster. "Monoaminergic system modulation in depression and alzheimer’s disease: a new standpoint?" December 20, 2021. https://assignbuster.com/monoaminergic-system-modulation-in-depression-and-alzheimers-disease-a-new-standpoint/.


Bibliography


AssignBuster. "Monoaminergic system modulation in depression and alzheimer’s disease: a new standpoint?" December 20, 2021. https://assignbuster.com/monoaminergic-system-modulation-in-depression-and-alzheimers-disease-a-new-standpoint/.

Work Cited

"Monoaminergic system modulation in depression and alzheimer’s disease: a new standpoint?" AssignBuster, 20 Dec. 2021, assignbuster.com/monoaminergic-system-modulation-in-depression-and-alzheimers-disease-a-new-standpoint/.

Get in Touch

Please, let us know if you have any ideas on improving Monoaminergic system modulation in depression and alzheimer’s disease: a new standpoint?, or our service. We will be happy to hear what you think: [email protected]