Next Article in Journal
Comparative Analysis of the Placental Microbiome in Pregnancies with Late Fetal Growth Restriction versus Physiological Pregnancies
Next Article in Special Issue
Molecular Mechanisms of Dementia
Previous Article in Journal
Hairy Root Cultures as a Source of Phenolic Antioxidants: Simple Phenolics, Phenolic Acids, Phenylethanoids, and Hydroxycinnamates
Previous Article in Special Issue
Light, Water, and Melatonin: The Synergistic Regulation of Phase Separation in Dementia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cholinergic Antagonists and Behavioral Disturbances in Neurodegenerative Diseases

1
Department of Geriatric and Internal Medicine, Reims University Hospitals, Maison Blanche Hospital, 51092 Reims, France
2
UR 3797 Vieillissement, Fragilité (VieFra), Faculty of Medicine, University of Reims Champagne-Ardenne, 51687 Reims, France
3
Cognition Health and Society Laboratory (C2S-EA 6291), Faculty of Medicine, University of Reims Champagne-Ardenne, 51687 Reims, France
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(8), 6921; https://doi.org/10.3390/ijms24086921
Submission received: 14 February 2023 / Revised: 26 March 2023 / Accepted: 4 April 2023 / Published: 7 April 2023
(This article belongs to the Special Issue Molecular Mechanisms of Dementia)

Abstract

:
Cholinergic antagonists interfere with synaptic transmission in the central nervous system and are involved in pathological processes in patients with neurocognitive disorders (NCD), such as behavioral and psychological symptoms of dementia (BPSD). In this commentary, we will briefly review the current knowledge on the impact of cholinergic burden on BPSD in persons with NCD, including the main pathophysiological mechanisms. Given the lack of clear consensus regarding symptomatic management of BPSD, special attention must be paid to this preventable, iatrogenic condition in patients with NCD, and de-prescription of cholinergic antagonists should be considered in patients with BPSD.

1. Background

Acetylcholine (ACh) is the main neurotransmitter of the central nervous system. It was the first neurotransmitter to be discovered, and the scientists who first described it, Henry Dale and Otto Loewi, were awarded the Nobel Prize for the Physiology and Medicine for their discovery [1]. A cholinergic deficit in Alzheimer’s disease (AD) was described for the first time by several groups simultaneously, namely Davies and Maloney [2], Perry et al. [3] and Bowen et al. [4,5]. The cholinergic deficit observed during AD is thought to be partially linked to loss of neurons at the level of the central cholinergic nuclei; especially the nucleus basalis of Meynert [6,7]. The cholinergic hypothesis is one of the explanations for the cognitive decline observed in AD [8,9].
ACh binds to pre- or postsynaptic receptors of two different classes. The first class of receptors is composed of ligand-gated cation channels, termed nicotinic ACh receptors (nAChR). nAChR have been found in various areas of the cerebral cortex, notably the hippocampal regions, the substantia nigra, and the ventral tegmental area [10]. nAChR are homo- or hetero-pentameric transmembrane proteins comprising five subunits. The most commonly found representatives in the central nervous system are the α7 and α4β2 receptors [11,12]. Nicotinic receptors are located both pre- and post-synaptically. In the pre-synaptic context, they promote the release of glutamate destined for dopaminergic neurons [13]. At the level of the GABAergic neurons, nAChR are located post-synaptically [14]. The second class of ACh receptors, termed muscarinic receptors, signal via heterotrimeric GTP binding proteins (G proteins). Muscarinic receptors are classified into five main subtypes, namely M1, M2, M3, M4, and M5. M1 receptors are predominant, and account for 35 to 60% of all muscarinic receptors in the human brain [15]. M2 receptors are present in the presynaptic neurons of the thalamic region, the nucleus basalis of Meynert, and limbic structures such as the hippocampus and amygdala. M3 and M4 receptors are more rarely found in the brain; M3 receptors are mainly located in the hippocampus and striatum, and are implicated in the regulation of insulin secretion [16]. In a murine model, M4 receptors are inhibitory autoreceptors in glutamatergic neurons, and they regulate the metabolism of dopamine [17]. It is thought that M5 receptors are expressed at low levels in the brain [18].
The cholinergic deficit that is present from the early stages in certain neurocognitive disorders (NCD) can be aggravated iatrogenically by medical therapy. Medications with anticholinergic properties are reversible and competitive inhibitors of ACh receptors. Cholinergic antagonists act by blocking nicotinic or muscarinic receptors in the central and peripheral nervous system. They prevent ACh from binding to its dedicated receptors, thereby reducing cholinergic transmission. The majority of drugs with anticholinergic properties are not receptor-specific. Consequently, they may act on either type of the receptor, be it muscarinic or nicotinic.
The term “anticholinergic burden” (AB) refers to the cumulative effect of taking one or more medications with anticholinergic properties [19,20]. Often, the AB is the result of taking several drugs with low anticholinergic load or activity. For some medications, the anticholinergic properties are known to prescribers, and indeed constitute the desired effect, such as treatments for stress urinary incontinence. Conversely, for other medications, the anticholinergic effects maybe unknown and harmful [19,20], and may be responsible for falls, cognitive and functional decline, and delirium.
Drugs with anticholinergic effects refer to drugs that bind exclusively to muscarinic receptors (M1, M2, M3, M4, and M5). The antagonism of these receptors corresponds to their primary mechanism of action (e.g., oxybutynin, trihexyphenidyl, and ipratropium bromide), and to drugs whose anticholinergic activity is not connected with their primary therapeutic purpose and mechanism of action (e.g., antidepressants, antipsychotics, and antihistamines) [21]. Blockades of nicotinic receptor sites attributed to these drugs is negligible [22].
Most drugs with anticholinergic activity are nonselective for receptor binding and are not tissue-selective. The distribution of muscarinic receptors across many physiological systems leads to a wide range of peripheral (e.g., dry mouth, dry eyes, urinary retention, constipation, and tachycardia) and central (e.g., cognitive impairment, delirium, and confusion) adverse effects. Affinity for muscarinic receptors varies from one therapeutic class to another, and from one molecule to another within the same therapeutic class [23].
In a study by Golds et al. [24], the binding of certain antidepressants to Ach muscarinic receptors in rat brain was investigated. Most of atypical antidepressant drugs tested in this radio-ligand binding assay were muscarinic receptor antagonists. When the dissociation constants (Kd) for all the drugs tested were compared, they were found to fall approximately into different groups: Amitryptiline with the lowest Kd and therefore highest affinity of the drugs tested for the muscarinic receptor; drugs with dissociation constants in the 1 × 10−7 M to 7 × 10−7 M range: desipramine, doxepin, imipramine, maprotiline, mianserin. These drugs were 5 to 20 times less potent than amitryptiline for binding to the muscarinic receptor.
Measurement of anticholinergic activity is based on invasive techniques, either via blood tests [25], or the cerebrospinal fluid [26,27]. Non-invasive estimation of the AB can be performed using tools such as scales or indices [28], but the major drawback of this approach is the heterogeneity between instruments in terms of prediction of outcomes [29]. Given the different properties of the AB estimation tools (expert agreement, correlation with measures of serum anticholinergic activity, cross-sectional design of princeps studies), the scales are heterogeneous between them [30]. A drug can have an AB rated differently depending on the tool used as having weak or strong anticholinergic properties. Given this heterogeneity, it is difficult for the clinician to favor one scale over another. However, the choice of a scale by the clinician and/or researcher must be made according to the outcomes of interest: risk of fall, delirium, cognitive and functional decline, rehospitalization, or death [31,32,33,34,35,36,37,38].
Beyond its impact on cognitive function, the cholinergic deficit in NCD is thought to be implicated in certain behavioral and psychological symptoms of dementia (BPSD) [39,40,41]. These include notably irritability, depression, agitation, psychosis, sleep disorders, anxiety, apathy, dysphoria, aberrant motor behavior, hallucinations, and delusions [42]. BPSD affect 90% of subjects with AD, and are also common in other forms of NCD [43]. They are a frequent cause of hospitalization [44] and are associated with an increased risk of nursing home entry [45], caregiver burnout [46], and more rapid progression of NCD [47]. There is currently no consensus regarding the medical management of BPSD, and non-drug approaches should be preferred [48].
In this article, we review the established relations between exposure to anticholinergic drugs and the onset or persistence of BPSD during NCD. We outline the main pathophysiological mechanisms, and highlight the key approaches to therapeutic management, both current and future.

2. Anticholinergic Burden and Behavioral Disturbances in Neurodegenerative Diseases

2.1. Alzheimer’s Disease

AD is the leading cause of NCD worldwide. It is characterized neuropathologically by the extracellular accumulation of amyloid-beta protein [49] and by the intracellular accumulation oh hyperphosphorylated tau protein in neurofibrillary tangles [50]. In addition to the cognitive decline occurring during the disease course, AD patients frequently experience BPSD. The most common BPSD among patients with AD are agitation [51]; delusions, apathy, hallucinations [52]; depression [53]; anxiety [54] and aberrant motor behavior [55].
In a cross-sectional study with 473 patients mostly with AD, BPSD were more frequent and more severe in patients with a higher anticholinergic burden, as assessed by the Anticholinergic Cognitive Burden (ACB) [56] and Han’s score (Han et al. JAGS 2008). Overall, the more the number of anticholinergic properties increased, the more there was a negative impact on the NPI score. However, the types of BPSD were not specified in this publication [57].
In a longitudinal study with 230 patients with probable AD, exposure to anticholinergic drugs was associated with an increased risk of psychosis [58]. In this study, the five most frequently prescribed anticholinergic drugs included two psychotropic agents (olanzapine and amitriptyline) and three cardiovascular drugs (warfarin, digoxin, and furosemide).
In a study investigating the relationship between serum anticholinergic activity (SAA) and the severity of clinical symptoms of AD patients, BPSD, and particularly items of paranoid and delusional ideation and hallucinations, were found to be more frequent in those with positive SAA [59]. The authors postulated that there may be a certain level of endogenous SAA acting as a predisposing factor to BPSD among patients with AD, and that this could be aggravated by the additional anticholinergic burden deriving from prescribed drugs. In this study, most of the cholinergic antagonists to which patients with NCD were exposed belonged to the pharmacological class of psychotropic drugs.
Sunderland et al. showed that certain cholinergic antagonists (notably, intravenous scopolamine in their study) caused significant behavioral changes in patients with AD compared to age- and sex-matched controls on five domains of the Brief Psychiatric Rating Scale, namely thought disorder, hostility, anxiety/depression, mania, and activation [60].
In a sample of 125 elderly patients with NCD (mainly AD), reducing the anticholinergic burden by at least 20% yielded a significant decrease in the frequency × severity scores of the Neuropsychiatric Inventory–Nursing Home scale, and reduced the impact on the caregivers’ workload [61]. In a larger sample of 147 patients, the same group reported that using the Anticholinergic Drug Scale [62], a reduction of 2 points in patients with moderate-intensity BPSD was associated with a clinically significant reduction in the frequency and severity of symptoms, while a reduction of 3 points yielded a clinically significant improvement in the occupational disruptiveness score [63].
A prospective study by Liu et al. failed to establish a relationship between exposure to anticholinergic burden, and the occurrence of BPSD [64]. However, contrary to previous publications, BPSD were not evaluated using a validated instrument in this study, such as the Neuropsychiatric Inventory, but rather using an indirect estimation of BPSD via the prescription of antipsychotics, antidepressants, and sedative-hypnotics of estimated treatment duration ≥30 days during follow-up. Despite an average follow-up of 15 years in this cohort, this measurement bias hampers the interpretation of the findings.
In a study of 9 patients with AD who were taking medication for urinary incontinence, Jewart et al. reported that psychiatric symptoms, as measured by the NPI completed by the caregiver, did not differ between patients who were on medication and those were not [65]. Further, serum anticholinergic activity was not significantly associated with the Memory and Behavior Checklist score. However, the major limitation of this study is the small sample size, which limits the statistical power.
Accordingly, it is nowadays clear that exposure to an anticholinergic burden leads to more frequent BPSD among patients with AD, but conversely, reducing the anticholinergic burden makes it possible to attenuate BPSD in samples of subjects presenting predominantly AD. It therefore seems logical to assume that anticholinergic drugs may be implicated in the genesis and/or persistence of BPSD in AD.
These conclusions are based on the cholinergic hypothesis previously proposed by several authors [8,9]. Cummings and Kaufer [66] suggested that the cholinergic deficit was implicated in the genesis and persistence of BPSD such as delusions, depression, agitation, and apathy.
In a post-mortem study of 46 patients with NCD, of whom 36 had AD, Minger et al. showed that compared to 32 controls, cholinergic deficit in AD was associated with BPSD such as aggressive behavior and depression [67]. Similarly, Garcia-Alloza et al. investigated an imbalance between the cholinergic and serotonergic systems in a series of 22 autopsies from AD patients who had undergone prospective follow-up for cognitive impairment and BPSD prior to death. They reported that decreases in cholinergic function correlated with aggressive behavior [68].
In addition to a deficit of production of ACh due to loss of neurons at the level of the central cholinergic nuclei, genetic variation in the α7 nicotinic acetylcholine receptor gene has been shown to be associated with delusions in AD [69]. The specific implication of nicotinic receptors in the occurrence of delusions during NCD is garnering increasing attention [70]. Moreover, the blockage of these receptors by anticholinergics is thought to be involved in the onset of BPSD in patients with AD, including apathy, hallucinations, delusions, and disinhibition [66].
In patients with AD, Lai et al. used radioligand binding assays to investigate the metabolic anomalies and the density of muscarinic receptors in the frontal and temporal cortex, and correlate neurochemical findings with the presence of BPSD, notably delusions and hallucination [71]. Indeed, it is believed that the cholinergic deficit is most pronounced in the frontal and temporal cortex, and that there is increased M2 receptor binding. Moreover, in this study, authors found that M2 receptor density was significantly increased in the frontal cortex of patients with AD with hallucination and in the temporal cortex of those with delusions. These findings should be interpreted in perspective with those from an autopsy study by Wang et al., who showed a statistically significant decrease in messenger RNA (mRNA) encoding for the M1 receptor in the temporal and occipital cortex, with no change in other regions. There was also no change in the level of mRNA encoding the M2, M3, or M4 receptors in any of the brain regions studied [72]. Finally, another post-mortem study on brain specimens from patients with AD and significant psychotic symptoms, showed a loss of non-M2 muscarinic receptors in the orbitofrontal cortex, independently of the effects of dementia severity [73].

2.2. Lewy Body Dementia/Parkinson’s Disease Related Dementia

Synucleinopathies comprise several NCD characterized by the accumulation of aggregated forms of the α-synuclein in both neuronal and non-neuronal cells of the brain [74]. Lewy body diseases are represented by the accumulation of aggregated α-synuclein into Lewy bodies within vulnerable neurons, and Lewy neurites in neuronal processes. The Lewy body diseases comprise mainly dementia with Lewy bodies (LBD) and Parkinson’s disease (PD)-related dementia. The most common BPSD among patients with LBD are anxiety, depression, apathy, agitation, sleep disorders, and psychosis [75].
Few studies to date have investigated the impact of exposure to anticholinergics on BPSD in NCD with Lewy bodies. In a study by Jaïdi et al., five subjects with LBD were included, and a reduction in the anticholinergic burden made it possible to reduce BPSD in these subjects [61,63]. However, the very small number of subjects precludes any generalization of these results. Nevertheless, it would appear that in patients with Lewy body NCD, exposure to anticholinergics may aggravate BPSD.
Indeed, in subjects with PD (another form of synucleinopathy), there is a more marked loss of neurons in the central cholinergic nuclei than in AD, suggesting that the cholinergic deficit plays a more important role in NCD with Lewy bodies than in AD [6,7]. The cholinergic deficit is also associated with hallucinations in NCD with Lewy bodies, as a corollary of the neurodegeneration of a central cholinergic neuron, the pedunculopontine tegmental nucleus [76].
Hori et al. purported that the onset of visual hallucinations and delusions occurs earlier in NCD with Lewy bodies, because initiation of endogenous anticholinergic activity occurs earlier than in AD, under the influence of hormonal, inflammatory, and neurovegetative factors [77].
As in AD, the onset of visual hallucinations in LBD is associated with reduction in ACh-related activity in the temporal and parietal cortex [78]. In addition, there is a reduction in choline acetyl transferase (ChAT) activity in LBD, as well as an increase in M2 and M4 receptor binding [79].
According to the results of a study using in vivo SPECT imaging of muscarinic receptors using (R,R) 123I-QNB in patients with LBD or PDD, there is a significant increase in the binding of 123I-QNB in the occipital lobe (the left lobe in DLB, and the right lobe in PDD). (R,R) 123I-QNB is a ligand with selectivity for the M1 and M4 receptor subtypes. This mechanism appears to be involved in the occurrence of visual hallucinations and visuo-spatial impairment [80].
Moreover, it has been shown that there is a more marked reduction in the M1 subtype of muscarinic receptors in LBD than in AD, and blockade of these receptors is thought to be associated with complex visual hallucinations in NCD with Lewy bodies [81,82,83]. Shiozaki et al. demonstrated in necropsied brains from a series of 7 patients with LBD and 11 patients with AD that there was a higher proportion of M1 receptors in LBD patients, and M2 receptors in AD patients. The authors purported that the deregulation of the cholinergic system differs between the two diseases, and they further hypothesized that presynaptic cholinergic depletion due to a more marked loss of neurons in the central cholinergic nuclei than in AD causes the upregulation of M1 receptors in DLB [81].

2.3. Frontotemporal Dementia

Frontotemporal dementia refers to a group of NCD that encompasses progressive dysfunction in executive functioning, behavior disturbances, and language. As its name indicates, it is a cluster of syndromes that results from the degeneration of the frontal ant temporal lobes, and is subdivided into two categories that are unique with respect to their predominating presentations: behavioral subtype (bvFTD) and language subtype [84]. The most common BPSD found during the course of the bvFTD are disinhibition, apathy, loss of empathy or sympathy and preservative, stereotyped or compulsive behaviors [85].
To the best of our knowledge, no study to date has investigated the impact of anticholinergic burden on BPSD in the setting of bvFTD. Compared to AD, the cholinergic system is relatively well preserved in FTD. However, autopsy studies in subjects with FTD have shown a reduction in muscarinic receptors [86,87]. This suggests that exposure to anticholinergics may be harmful in patients with FTD.

2.4. Vascular Dementia

Vascular dementia (VaD) refers to a decline in mental ability, severe enough to interfere with daily life. The Vascular Impairment of Cognition Classification Consensus Study defines VaD as a clinical deficit in at least one cognitive domain that is of sufficient severity to cause a severe disruption of (instrumental) activities in daily living [88]. According to this classification, VaD is classified into four major subtypes: post-stroke dementia (defined as a dementia within 6 months after a stroke); subcortical ischemic VaD; multi-infarct (cortical) dementia; and mixed dementia. VaD is associated with irritability, apathy, depression, sleep/night-time behaviors, and agitation [89].
Again, no study to date has specifically evaluated the effects of anticholinergic burden on BPSD in a sufficiently large sample of patients with VaD. Of note, in the studies published by Jaïdi et al., 13.6% of subjects included had VaD and almost 25% had mixed NCD (AD and VaD) [61,63]. It was shown in these patients that a reduction in the anticholinergic burden led to an improvement in BPSD symptoms.
In VaD, cholinergic deficits may result from ischemia of the central cholinergic nuclei, which are very sensitive to ischemia since they are irrigated by perforating arteries. The cholinergic deficit observed in VaD may also arise from white matter lesions that alter cholinergic input to the cortex [90,91]. Accordingly, in patients with VaD, and independently of any AD-like pathology, cholinergic activity is diminished [27,92].
Table 1 presents the main clinical studies that have evaluated the relationship between BPSD and AB according to the type of NCD and Figure 1 shows the mechanism of action of cholinergic antagonists at the cholinergic synapse.
Alzheimer’s disease: the cholinergic deficit is most pronounced in the frontal and temporal cortex, and there is an increase in binding to the M2 receptor [67]. There is a loss of non-M2 muscarinic receptors in the orbitofrontal cortex, independently of the effects of dementia severity in AD patients with psychotic symptoms [69].
Lewy body dementia/Parkinson’s disease related dementia: There is increased binding in the occipital lobe with selectivity for the M1 and M4 receptor subtypes, and this mechanism appears to be involved in the occurrence of visual hallucinations and visuo-spatial impairment [76]. Furthermore, there is an increased binding in the occipital lobe with selectivity for the M1 and M4 receptor subtypes, and this mechanism appears to be involved in the occurrence of visual hallucinations and visuo-spatial impairment [77,78,79].
Fronto-temporal dementia and Vascular dementia: There are no studies investigating the behavioral outcomes of muscarnic receptor binding in FTD and VaD.

3. How Can We Lessen the Impact of AB on BPSD?

The iatrogenic effects arising from the anticholinergic burden are largely avoidable. Although the anticholinergic properties of some drugs are unknown or occult, they can be detected more easily by the systematic use of scales to estimate anticholinergic burden [28]. These scales present the notable advantage of being non-invasive. However, there is a certain degree of heterogeneity between the existing instruments [29]. Thus, the prescriber should assess the anticholinergic burden when initiating a treatment, or during periodic medication reviews (e.g., at the occasion of a hospitalization or admission to a nursing home). Within a deprescribing framework, in addition to the leading role of the prescriber, the pharmacist can also be a key contributor to reducing the anticholinergic burden (via such approaches as medication review, academic detailing, education of healthcare practitioners, and computerized clinical decision support systems), especially among older populations, as shown in several therapeutic trials [93]. Deprescription of anticholinergics seems to be feasible, even in a population of frail geriatric patients [94]. To be profitable, this strategy would be desirable to specifically target psychotropic drugs given the prevalence of their prescription in the elderly population on the one hand [95,96], and their significant anticholinergic burden on the other hand [97,98].
As far as possible, a cholinergic antagonist whose indication is indisputable should be replaced by another molecule of the same class using the anticholinergic load estimation scales. The Anticholinergic Risk Scale [99] seems to date to have the best correlation with pharmacokinetic parameters [23]. Moreover, in a situation of overuse, cholinergic antagonist cannot be renewed.
A trial is currently underway to compare the effectiveness of different types of reduction interventions (e.g., pharmacist-led versus GP-led; educational versus audit and feedback) in reducing overall anticholinergic burden; to establish optimal duration of anticholinergic reduction interventions; sustainability and lessons learned for upscaling; and to compare results according to differing anticholinergic scales used in medication reduction intervention trials [100].
Despite the prevalence of BPSD and its hazards in patients with NCD, there has not yet been medication that is clearly effective and approved by the Food and Drug Administration or the French High Health Authority. Again, and in the absence of approved therapy targeting BPSD with a satisfying benefit/risk balance, it seems essential to pay a special attention not to worsen a cholinergic deficit (suspected or proven). Every practitioner should avoid as far as possible cholinergic antagonists in patients with NCD whether they present with BPSD or not.
Cholinesterase inhibitors (CI) have shown to yield a clinically significant, albeit modest improvement in BPSD in both AD and NCD with Lewy bodies [101,102]. Nevertheless, in the case of FTD, CI have been shown to be inefficacious at best in improving BPSD [103], and may even aggravate symptoms [104]. Furthermore, memantine alone or in combination with CIs participates in the improvement of BPSD with a different effect size from one study to another [31,105,106,107,108].
Finally, muscarinic ACh receptors seem to be a promising therapeutic target in BPSD, in which a cholinergic deficit is implicated, particularly in AD and NCD with Lewy bodies. Indeed, subtype-selective allosteric modulators for muscarinic ACh receptors (coupled with G protein) have shown promising therapeutic potential and may yield a benefit in the management of BPSD [109,110].
Table 2 illustrates the different types of ACh receptors and therapeutic targets of potential interest.

4. Further Considerations/Conclusions

This brief review illustrates that to date, few clinical studies have investigated specifically the impact of the AB in BPSD. There is a strong presumption based on the imbalance between neurotransmitters, especially ACh, and receptor dysfunction, but an actual deleterious effect of anticholinergics on BPSD in NCDs other than AD has never been clearly demonstrated. Nevertheless, despite the current lack of robust evidence, special attention should be paid to avoid medication-induced iatrogenic effects, notably due to anticholinergic drugs, in patients presenting with NCD. This could be achieved by practicing a “zero exposure” policy in patients with NCD, based on the “safety-first” principle. Other prospective clinical studies are warranted to evaluate the impact of the anticholinergic burden and the effect of deprescription of anticholinergics on BPSD.

Author Contributions

Conceptualization, R.M. and Y.J.; methodology, R.M. and Y.J.; software, Y.J.; validation, R.M., J.L.N. and Y.J.; formal analysis, none.; investigation, none.; resources, S.L.-B.; data curation, none.; writing—original draft preparation, R.M., S.B., D.T., I.M. and Y.J.; writing—review and editing, R.M. and Y.J.; visualization, S.L.-B., Y.J. and R.M.; supervision, Y.J.; project administration, R.M.; funding acquisition, none. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Auterhoff, H. Nobel lectures-physiology or medicine 1942–1962. Herausgegeben von der Nobel-Stiftung. 840 Seiten. Elsevier Publishing Co, Amsterdam—London—New York 1964. Preis etwa DM 90,—. Arch. Pharm. Pharm. Med. Chem. 1965, 298, 77–78. [Google Scholar] [CrossRef]
  2. Davies, P.; Maloney, A.J. Selective loss of central cholinergic neurons in Alzheimer’s disease. Lancet 1976, 2, 1403. [Google Scholar] [CrossRef] [PubMed]
  3. Perry, E.K.; Perry, R.H.; Blessed, G.; Tomlinson, B.E. Necropsy evidence of central cholinergic deficits in senile dementia. Lancet 1977, 1, 189. [Google Scholar] [CrossRef]
  4. Spillane, J.A.; White, P.; Goodhardt, M.J.; Flack, R.H.A.; Bowen, D.M.; Davison, A.N. Selective vulnerability of neurones in organic dementia. Nature 1977, 266, 558–559. [Google Scholar] [CrossRef]
  5. White, P.; Goodhardt, M.J.; Keet, J.P.; Hiley, C.R.; Carrasco, L.H.; Williams, I.E.I.; Bowen, D.M. Neocortical Cholinergic Neurons in Elderly People. Lancet 1977, 309, 668–671. [Google Scholar] [CrossRef]
  6. Arendt, T.; Bigl, V.; Arendt, A.; Tennstedt, A. Loss of neurons in the nucleus basalis of Meynert in Alzheimer’s disease, paralysis agitans and Korsakoff’s Disease. Acta Neuropathol. 1983, 61, 101–108. [Google Scholar] [CrossRef]
  7. Whitehouse, P.J.; Price, D.L.; Struble, R.G.; Clark, A.W.; Coyle, J.T.; Delon, M.R. Alzheimer’s disease and senile dementia: Loss of neurons in the basal forebrain. Science 1982, 215, 1237–1239. [Google Scholar] [CrossRef] [PubMed]
  8. Drachman, D.A.; Leavitt, J. Human memory and the cholinergic system. A relationship to aging? Arch. Neurol. 1974, 30, 113–121. [Google Scholar] [CrossRef]
  9. Bartus, R.T.; Dean, R.L., 3rd; Beer, B.; Lippa, A.S. The cholinergic hypothesis of geriatric memory dysfunction. Science 1982, 217, 408–414. [Google Scholar] [CrossRef]
  10. Gotti, C.; Moretti, M.; Gaimarri, A.; Zanardi, A.; Clementi, F.; Zoli, M. Heterogeneity and complexity of native brain nicotinic receptors. Biochem. Pharmacol. 2007, 74, 1102–1111. [Google Scholar] [CrossRef]
  11. Leiser, S.C.; Bowlby, M.R.; Comery, T.A.; Dunlop, J. A cog in cognition: How the α7 nicotinic acetylcholine receptor is geared towards improving cognitive deficits. Pharmacol. Ther. 2009, 122, 302–311. [Google Scholar] [CrossRef]
  12. Picciotto, M.R.; Caldarone, B.J.; Brunzell, D.H.; Zachariou, V.; Stevens, T.R.; King, S.L. Neuronal nicotinic acetylcholine receptor subunit knockout mice: Physiological and behavioral phenotypes and possible clinical implications. Pharmacol. Ther. 2001, 92, 89–108. [Google Scholar] [CrossRef]
  13. Jones, B.E. Activity, modulation and role of basal forebrain cholinergic neurons innervating the cerebral cortex. Prog. Brain Res. 2004, 145, 157–169. [Google Scholar] [PubMed]
  14. Bürli, T.; Baer, K.; Ewers, H.; Sidler, C.; Fuhrer, C.; Fritschy, J.-M. Single Particle Tracking of α7 Nicotinic AChR in Hippocampal Neurons Reveals Regulated Confinement at Glutamatergic and GABAergic Perisynaptic Sites. PLoS ONE 2010, 5, e11507. [Google Scholar] [CrossRef] [PubMed]
  15. Volpicelli, L.A.; Levey, A.I. Muscarinic acetylcholine receptor subtypes in cerebral cortex and hippocampus. Prog. Brain Res. 2004, 145, 59–66. [Google Scholar] [PubMed]
  16. Gautam, D.; Han, S.-J.; Hamdan, F.F.; Jeon, J.; Li, B.; Li, J.H.; Cui, Y.; Mears, D.; Lu, H.; Deng, C.; et al. A critical role for β cell M3 muscarinic acetylcholine receptors in regulating insulin release and blood glucose homeostasis in vivo. Cell Metab. 2006, 3, 449–461. [Google Scholar] [CrossRef] [Green Version]
  17. Wess, J.; Eglen, R.M.; Gautam, D. Muscarinic acetylcholine receptors: Mutant mice provide new insights for drug development. Nat. Rev. Drug Discov. 2007, 6, 721–733. [Google Scholar] [CrossRef]
  18. Levey, A.I. Muscarinic acetylcholine receptor expression in memory circuits: Implications for treatment of Alzheimer disease. Proc. Natl. Acad. Sci. USA 1996, 93, 13541–13546. [Google Scholar] [CrossRef] [Green Version]
  19. Tune, L.E. Anticholinergic effects of medication in elderly patients. J. Clin. Psychiatry 2001, 62 (Suppl. S21), 11–14. [Google Scholar]
  20. Tune, L.; Carr, S.; Hoag, E.; Cooper, T. Anticholinergic effects of drugs commonly prescribed for the elderly: Potential means for assessing risk of delirium. Am. J. Psychiatry 1992, 149, 1393–1394. [Google Scholar]
  21. de Leon, J. Paying attention to pharmacokinetic and pharmacodynamic mechanisms to progress in the area of anticholinergic use in geriatric patients. Curr. Drug Metab. 2011, 12, 635–646. [Google Scholar] [CrossRef]
  22. Bostock, C.V.; Soiza, R.L.; Mangoni, A.A. Association between prescribing of antimuscarinic drugs and antimuscarinic adverse effects in older people. Expert Rev. Clin. Pharmacol. 2010, 3, 441–452. [Google Scholar] [CrossRef] [PubMed]
  23. Lavrador, M.; Cabral, A.C.; Veríssimo, M.T.; Fernandez-Llimos, F.; Figueiredo, I.V.; Castel-Branco, M.M. A Universal Pharmacological-Based List of Drugs with Anticholinergic Activity. Pharmaceutics 2023, 15, 230. [Google Scholar] [CrossRef] [PubMed]
  24. Golds, P.R.; Przyslo, F.R.; Strange, P.G. The binding of some antidepressant drugs to brain muscarinic acetylcholine receptors. Br. J. Pharmacol. 1980, 68, 541–549. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Tune, L.; Coyle, J.T. Serum Levels of Anticholinergic Drugs in Treatment of Acute Extrapyramidal Side Effects. Arch. Gen. Psychiatry 1980, 37, 293–297. [Google Scholar] [CrossRef]
  26. Tohgi, H.; Abe, T.; Hashiguchi, K.; Saheki, M.; Takahashi, S. Remarkable reduction in acetylcholine concentration in the cerebrospinal fluid from patients with Alzheimer type dementia. Neurosci. Lett. 1994, 177, 139–142. [Google Scholar] [CrossRef]
  27. Tohgi, H.; Abe, T.; Kimura, M.; Saheki, M.; Takahashi, S. Cerebrospinal fluid acetylcholine and choline in vascular dementia of Binswanger and multiple small infarct types as compared with Alzheimer-type dementia. J. Neural Transm. (Vienna) 1996, 103, 1211–1220. [Google Scholar] [CrossRef]
  28. Welsh, T.J.; van der Wardt, V.; Ojo, G.; Gordon, A.L.; Gladman, J.R.F. Anticholinergic Drug Burden Tools/Scales and Adverse Outcomes in Different Clinical Settings: A Systematic Review of Reviews. Drugs Aging 2018, 35, 523–538. [Google Scholar] [CrossRef]
  29. Lisibach, A.; Benelli, V.; Ceppi, M.G.; Waldner-Knogler, K.; Csajka, C.; Lutters, M. Quality of anticholinergic burden scales and their impact on clinical outcomes: A systematic review. Eur. J. Clin. Pharmacol. 2021, 77, 147–162. [Google Scholar] [CrossRef]
  30. Salahudeen, M.S.; Duffull, S.B.; Nishtala, P.S. Anticholinergic burden quantified by anticholinergic risk scales and adverse outcomes in older people: A systematic review. BMC Geriatr. 2015, 15, 31. [Google Scholar] [CrossRef] [Green Version]
  31. Clerici, F.; Vanacore, N.; Elia, A.; Spila-Alegiani, S.; Pomati, S.; Da Cas, R.; Raschetti, R.; Mariani, C. Memantine effects on behaviour in moderately severe to severe Alzheimer’s disease: A post-marketing surveillance study. Neurol. Sci. 2012, 33, 23–31. [Google Scholar] [CrossRef] [PubMed]
  32. Pasina, L.; Djade, C.D.; Lucca, U.; Nobili, A.; Tettamanti, M.; Franchi, C.; Salerno, F.; Corrao, S.; Marengoni, A.; Iorio, A.; et al. Association of anticholinergic burden with cognitive and functional status in a cohort of hospitalized elderly: Comparison of the anticholinergic cognitive burden scale and anticholinergic risk scale: Results from the REPOSI study. Drug Aging 2013, 30, 103–112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Vetrano, D.L.; La Carpia, D.; Grande, G.; Casucci, P.; Bacelli, T.; Bernabei, R.; Onder, G.; Agabiti, N.; Bartolini, C.; Bernabei, R.; et al. Anticholinergic Medication Burden and 5-Year Risk of Hospitalization and Death in Nursing Home Elderly Residents With Coronary Artery Disease. J. Am. Med. Dir. Assoc. 2016, 17, 1056–1059. [Google Scholar] [CrossRef] [PubMed]
  34. Kalisch Ellett, L.M.; Pratt, N.L.; Ramsay, E.N.; Barratt, J.D.; Roughead, E.E. Multiple Anticholinergic Medication Use and Risk of Hospital Admission for Confusion or Dementia. J. Am. Geriatr. Soc. 2014, 62, 1916–1922. [Google Scholar] [CrossRef] [PubMed]
  35. Watanabe, S.; Fukatsu, T.; Kanemoto, K. Risk of hospitalization associated with anticholinergic medication for patients with dementia. Psychogeriatrics 2018, 18, 57–63. [Google Scholar] [CrossRef] [PubMed]
  36. Tan, E.C.K.; Eriksdotter, M.; Garcia-Ptacek, S.; Fastbom, J.; Johnell, K. Anticholinergic Burden and Risk of Stroke and Death in People with Different Types of Dementia. J. Alzheimers Dis. 2018, 65, 589–596. [Google Scholar] [CrossRef] [Green Version]
  37. Lattanzio, F.; Corica, F.; Schepisi, R.; Amantea, D.; Bruno, F.; Cozza, A.; Onder, G.; Volpato, S.; Cherubini, A.; Ruggiero, C.; et al. Anticholinergic burden and 1-year mortality among older patients discharged from acute care hospital. J. Am. Med. Dir. Assoc. 2018, 18, 705–713. [Google Scholar] [CrossRef]
  38. Cross, A.J.; George, J.; Woodward, M.C.; Ames, D.; Brodaty, H.; Wolfe, R.; Connors, M.H.; Elliott, R.A. Potentially Inappropriate Medication, Anticholinergic Burden, and Mortality in People Attending Memory Clinics. J. Alzheimers Dis. 2017, 60, 349–358. [Google Scholar] [CrossRef]
  39. Pinto, T.; Lanctôt, K.L.; Herrmann, N. Revisiting the cholinergic hypothesis of behavioral and psychological symptoms in dementia of the Alzheimer’s type. Ageing Res. Rev. 2011, 10, 404–412. [Google Scholar] [CrossRef]
  40. Francis, P.T.; Perry, E.K. Cholinergic and other neurotransmitter mechanisms in Parkinson’s disease, Parkinson’s disease dementia, and dementia with Lewy bodies. Mov. Diord. 2007, 22 (Suppl. S17), S351–S357. [Google Scholar] [CrossRef]
  41. Cummings, J.L.; Back, C. The cholinergic hypothesis of neuropsychiatric symptoms in Alzheimer’s disease. Am. J. Psychiatry 1998, 6 (Suppl. S1), S64–S78. [Google Scholar] [CrossRef]
  42. Steinberg, M.; Shao, H.; Zandi, P.; Lyketsos, C.G.; Welsh-Bohmer, K.A.; Norton, M.C.; Breitner, J.C.; Steffens, D.C.; Tschanz, J.T.; Cache County, I. Point and 5-year period prevalence of neuropsychiatric symptoms in dementia: The Cache County Study. Int. J. Geriatr. Psychiatry 2008, 23, 170–177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Finkel, S.I.; Costa e Silva, J.; Cohen, G.; Miller, S.; Sartorius, N. Behavioral and psychological signs and symptoms of dementia: A consensus statement on current knowledge and implications for research and treatment. Int. Psychogeriatr. 1996, 8 (Suppl. S3), 497–500. [Google Scholar] [CrossRef] [PubMed]
  44. McKeith, I.; Cummings, J. Behavioural changes and psychological symptoms in dementia disorders. Lancet Neurol. 2005, 4, 735–742. [Google Scholar] [CrossRef] [PubMed]
  45. Chan, D.C.; Kasper, J.D.; Black, B.S.; Rabins, P.V. Presence of behavioral and psychological symptoms predicts nursing home placement in community-dwelling elders with cognitive impairment in univariate but not multivariate analysis. J. Gerontol. A Biol. Sci. Med. Sci. 2003, 58, 548–554. [Google Scholar] [CrossRef] [Green Version]
  46. van der Lee, J.; Bakker, T.J.E.M.; Duivenvoorden, H.J.; Dröes, R.-M. Multivariate models of subjective caregiver burden in dementia: A systematic review. Ageing Res. Rev. 2014, 15, 76–93. [Google Scholar] [CrossRef] [PubMed]
  47. Peters, M.E.; Rosenberg, P.B.; Steinberg, M.; Norton, M.C.; Welsh-Bohmer, K.A.; Hayden, K.M.; Breitner, J.; Tschanz, J.T.; Lyketsos, C.G. Neuropsychiatric Symptoms as Risk Factors for Progression From CIND to Dementia: The Cache County Study. Am. J. Geritar. Psychiatry 2013, 21, 1116–1124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Kales, H.C.; Lyketsos, C.G.; Miller, E.M.; Ballard, C. Management of behavioral and psychological symptoms in people with Alzheimer’s disease: An international Delphi consensus. Intl. Psychogeriatr. 2019, 31, 83–90. [Google Scholar] [CrossRef] [PubMed]
  49. Hardy, J.; Allsop, D. Amyloid deposition as the central event in the aetiology of Alzheimer’s disease. Trends Pharmacol. Sci. 1991, 12, 383–388. [Google Scholar] [CrossRef]
  50. Bancher, C.; Brunner, C.; Lassmann, H.; Budka, H.; Jellinger, K.; Wiche, G.; Seitelberger, F.; Grundke-Iqbal, I.; Iqbal, K.; Wisniewski, H.M. Accumulation of abnormally phosphorylated τ precedes the formation of neurofibrillary tangles in Alzheimer’s disease. Brain Res. 1989, 477, 90–99. [Google Scholar] [CrossRef]
  51. Halpern, R.; Seare, J.; Tong, J.; Hartry, A.; Olaoye, A.; Aigbogun, M.S. Using electronic health records to estimate the prevalence of agitation in Alzheimer disease/dementia. Int. J. Geriatr. Psychiatry 2019, 34, 420–431. [Google Scholar] [CrossRef]
  52. Zhao, Q.F.; Tan, L.; Wang, H.F.; Jiang, T.; Tan, M.S.; Tan, L.; Xu, W.; Li, J.Q.; Wang, J.; Lai, T.J.; et al. The prevalence of neuropsychiatric symptoms in Alzheimer’s disease: Systematic review and meta-analysis. J. Affect. Disord. 2016, 190, 264–271. [Google Scholar] [CrossRef]
  53. Kaup, A.R.; Byers, A.L.; Falvey, C.; Simonsick, E.M.; Satterfield, S.; Ayonayon, H.N.; Smagula, S.F.; Rubin, S.M.; Yaffe, K. Trajectories of Depressive Symptoms in Older Adults and Risk of Dementia. JAMA Psychiatry 2016, 73, 525–531. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Johansson, M.; Stomrud, E.; Lindberg, O.; Westman, E.; Johansson, P.M.; van Westen, D.; Mattsson, N.; Hansson, O. Apathy and anxiety are early markers of Alzheimer’s disease. Neurobiol. Aging 2020, 85, 74–82. [Google Scholar] [CrossRef] [PubMed]
  55. Vik-Mo, A.O.; Giil, L.M.; Borda, M.G.; Ballard, C.; Aarsland, D. The individual course of neuropsychiatric symptoms in people with Alzheimer’s and Lewy body dementia: 12-year longitudinal cohort study. Br. J. Psychiatry 2020, 216, 43–48. [Google Scholar] [CrossRef] [PubMed]
  56. Boustani, M.; Campbell, N.; Munger, S.; Maidment, I.; Fox, C. Impact of anticholinergics on the aging brain: A review and practical application. Aging Health 2008, 4, 411–420. [Google Scholar] [CrossRef]
  57. Dauphinot, V.; Mouchoux, C.; Veillard, S.; Delphin-Combe, F.; Krolak-Salmon, P. Anticholinergic drugs and functional, cognitive impairment and behavioral disturbances in patients from a memory clinic with subjective cognitive decline or neurocognitive disorders. Alzheimers Res. Ther. 2017, 9, 58. [Google Scholar] [CrossRef] [Green Version]
  58. Cancelli, I.; Valentinis, L.; Merlino, G.; Valente, M.; Gigli, G.L. Drugs with Anticholinergic Properties as a Risk Factor for Psychosis in Patients Affected by Alzheimer’s Disease. Clin. Pharmacol. Ther. 2008, 84, 63–68. [Google Scholar] [CrossRef]
  59. Hori, K.; Konishi, K.; Watanabe, K.; Uchida, H.; Tsuboi, T.; Moriyasu, M.; Tominaga, I.; Hachisu, M. Influence of anticholinergic activity in serum on clinical symptoms of Alzheimer’s disease. Neuropsychobiology 2011, 63, 147–153. [Google Scholar] [CrossRef]
  60. Sunderland, T.; Tariot, P.N.; Cohen, R.M.; Weingartner, H.; Mueller, E.A., III; Murphy, D.L. Anticholinergic Sensitivity in Patients With Dementia of the Alzheimer Type and Age-Matched Controls: A Dose-Response Study. Arch. Gen. Psychiatry 1987, 44, 418–426. [Google Scholar] [CrossRef]
  61. Jaïdi, Y.; Nonnonhou, V.; Kanagaratnam, L.; Bertholon, L.A.; Badr, S.; Noël, V.; Novella, J.-L.; Mahmoudi, R. Reduction of the Anticholinergic Burden Makes It Possible to Decrease Behavioral and Psychological Symptoms of Dementia. Am. J. Geriatr. Psychiatry 2018, 26, 280–288. [Google Scholar] [CrossRef]
  62. Carnahan, R.M.; Lund, B.C.; Perry, P.J.; Pollock, B.G.; Culp, K.R. The Anticholinergic Drug Scale as a measure of drug-related anticholinergic burden: Associations with serum anticholinergic activity. J. Clin. Pharmacol. 2006, 46, 1481–1486. [Google Scholar] [CrossRef]
  63. Jaïdi, Y.; Guilloteau, A.; Nonnonhou, V.; Bertholon, L.-A.; Badr, S.; Morrone, I.; Novella, J.-L.; Mahmoudi, R. Threshold for a Reduction in Anticholinergic Burden to Decrease Behavioral and Psychological Symptoms of Dementia. J. Am. Med. Dir. Assoc. 2019, 20, 159–164.e3. [Google Scholar] [CrossRef] [PubMed]
  64. Liu, Y.-P.; Chien, W.-C.; Chung, C.-H.; Chang, H.-A.; Kao, Y.-C.; Tzeng, N.-S. Are Anticholinergic Medications Associated With Increased Risk of Dementia and Behavioral and Psychological Symptoms of Dementia? A Nationwide 15-Year Follow-Up Cohort Study in Taiwan. Front. Pharmacol. 2020, 11, 30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Jewart, R.D.; Green, J.; Lu, C.J.; Cellar, J.; Tune, L.E. Cognitive, behavioral, and physiological changes in Alzheimer disease patients as a function of incontinence medications. Am. J. Geriatr. Psychiatry 2005, 13, 324–328. [Google Scholar] [CrossRef] [PubMed]
  66. Cummings, J.L.; Kaufer, D. Neuropsychiatric aspects of Alzheimer’s disease. The cholinergic hypothesis revisited. Neurology 1996, 47, 876–883. [Google Scholar] [CrossRef]
  67. Minger, S.L.; Esiri, M.M.; McDonald, B.; Keene, J.; Carter, J.; Hope, T.; Francis, P.T. Cholinergic deficits contribute to behavioral disturbance in patients with dementia. Neurology 2000, 55, 1460–1467. [Google Scholar] [CrossRef]
  68. Garcia-Alloza, M.; Gil-Bea, F.J.; Diez-Ariza, M.; Chen, C.P.; Francis, P.T.; Lasheras, B.; Ramirez, M.J. Cholinergic-serotonergic imbalance contributes to cognitive and behavioral symptoms in Alzheimer’s disease. Neuropsychologia 2005, 43, 442–449. [Google Scholar] [CrossRef] [Green Version]
  69. Carson, R.; Craig, D.; Hart, D.; Todd, S.; McGuinness, B.; Johnston, J.A.; O’Neill, F.A.; Ritchie, C.W.; Passmore, A.P. Genetic Variation in the α7 Nicotinic Acetylcholine Receptor is Associated with Delusional Symptoms in Alzheimer’s Disease. Neuromolecular Med. 2008, 10, 377–384. [Google Scholar] [CrossRef]
  70. Caton, M.; Ochoa, E.L.M.; Barrantes, F.J. The role of nicotinic cholinergic neurotransmission in delusional thinking. NPJ Schizophr. 2020, 6, 16. [Google Scholar] [CrossRef]
  71. Lai, M.K.; Lai, O.F.; Keene, J.; Esiri, M.M.; Francis, P.T.; Hope, T.; Chen, C.P. Psychosis of Alzheimer’s disease is associated with elevated muscarinic M2 binding in the cortex. Neurology 2001, 57, 805–811. [Google Scholar] [CrossRef]
  72. Wang, S.-Z.; Zhu, S.-Z.; Mash, D.C.; El-Fakahany, E.E. Comparison of the concentration of messenger RNA encoding four muscarinic receptor subtypes in control and Alzheimer brains. Brain Res. Mol. Brain Res. 1992, 16, 64–70. [Google Scholar] [CrossRef] [PubMed]
  73. Tsang, S.W.; Francis, P.T.; Esiri, M.M.; Wong, P.T.; Chen, C.P.; Lai, M.K. Loss of [3H]4-DAMP binding to muscarinic receptors in the orbitofrontal cortex of Alzheimer’s disease patients with psychosis. Psychopharmacology 2008, 198, 251–259. [Google Scholar] [CrossRef] [PubMed]
  74. McCann, H.; Stevens, C.H.; Cartwright, H.; Halliday, G.M. α-Synucleinopathy phenotypes. Park. Relat. Disord. 2014, 20 (Suppl. S1), S62–S67. [Google Scholar] [CrossRef] [Green Version]
  75. Borroni, B.; Agosti, C.; Padovani, A. Behavioral and psychological symptoms in dementia with Lewy-bodies (DLB): Frequency and relationship with disease severity and motor impairment. Arch. Gerontol. Geriatr. 2008, 46, 101–106. [Google Scholar] [CrossRef] [PubMed]
  76. Manford, M.; Andermann, F. Complex visual hallucinations. Clinical and neurobiological insights. Brain 1998, 121, 1819–1840. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Hori, K.; Konishi, K.; Hosoi, M.; Tomioka, H.; Tani, M.; Kitajima, Y.; Hachisu, M. Mini Review: Anticholinergic Activity as a Behavioral Pathology of Lewy Body Disease and Proposal of the Concept of “Anticholinergic Spectrum Disorders”. Park. Dis. 2016, 2016, 5380202. [Google Scholar] [CrossRef] [Green Version]
  78. Perry, E.; Walker, M.; Grace, J.; Perry, R. Acetylcholine in mind: A neurotransmitter correlate of consciousness? Trends Neurosci. 1999, 22, 273–280. [Google Scholar] [CrossRef]
  79. Teaktong, T.; Piggott, M.A.; McKeith, I.G.; Perry, R.H.; Ballard, C.G.; Perry, E.K. Muscarinic M2 and M4 receptors in anterior cingulate cortex: Relation to neuropsychiatric symptoms in dementia with Lewy bodies. Behav. Brain Res. 2005, 161, 299–305. [Google Scholar] [CrossRef]
  80. Colloby, S.J.; Pakrasi, S.; Firbank, M.J.; Perry, E.K.; Piggott, M.A.; Owens, J.; Wyper, D.J.; McKeith, I.G.; Burn, D.J.; Williams, E.D.; et al. In vivo SPECT imaging of muscarinic acetylcholine receptors using (R,R) 123I-QNB in dementia with Lewy bodies and Parkinson’s disease dementia. NeuroImage 2006, 33, 423–429. [Google Scholar] [CrossRef]
  81. Shiozaki, K.; Iseki, E.; Uchiyama, H.; Watanabe, Y.; Haga, T.; Kameyama, K.; Ikeda, T.; Yamamoto, T.; Kosaka, K. Alterations of muscarinic acetylcholine receptor subtypes in diffuse lewy body disease: Relation to Alzheimer’s disease. J. Neurol. Neurosurg. Psychiatry 1999, 67, 209–213. [Google Scholar] [CrossRef] [PubMed]
  82. Perry, E.K.; Perry, R.H. Acetylcholine and hallucinations: Disease-related compared to drug-induced alterations in human consciousness. Brain Cogn. 1995, 28, 240–258. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Ballard, C.; Piggott, M.; Johnson, M.; Cairns, N.; Perry, R.; McKeith, I.; Jaros, E.; O’Brien, J.; Holmes, C.; Perry, E. Delusions associated with elevated muscarinic binding in dementia with Lewy bodies. Ann. Neurol. 2000, 48, 868–876. [Google Scholar] [CrossRef]
  84. Kurz, A.; Kurz, C.; Ellis, K.; Lautenschlager, N.T. What is frontotemporal dementia? Maturitas 2014, 79, 216–219. [Google Scholar] [CrossRef] [PubMed]
  85. Johnen, A.; Bertoux, M. Psychological and Cognitive Markers of Behavioral Variant Frontotemporal Dementia-A Clinical Neuropsychologist’s View on Diagnostic Criteria and Beyond. Front. Neurol. 2019, 10, 594. [Google Scholar] [CrossRef] [Green Version]
  86. Hansen, L.A.; Deteresa, R.; Tobias, H.; Alford, M.; Terry, R.D. Neocortical morphometry and cholinergic neurochemistry in Pick’s disease. Am. J. Pathol. 1988, 131, 507–518. [Google Scholar]
  87. Yates, C.M.; Simpson, J.; Maloney, A.F.; Gordon, A. Neurochemical observations in a case of Pick’s disease. J. Neurol. Sci. 1980, 48, 257–263. [Google Scholar] [CrossRef]
  88. Skrobot, O.A.; O’Brien, J.; Black, S.; Chen, C.; DeCarli, C.; Erkinjuntti, T.; Ford, G.A.; Kalaria, R.N.; Pantoni, L.; Pasquier, F.; et al. The Vascular Impairment of Cognition Classification Consensus Study. Alzheimers Dement. 2017, 13, 624–633. [Google Scholar] [CrossRef] [Green Version]
  89. Sep, Y.C.P.; Leeuwis, A.E.; Exalto, L.G.; Boomsma, J.M.; Prins, N.D.; Verwer, J.H.; Scheltens, P.; van der Flier, W.M.; Biessels, G.J. Neuropsychiatric Symptoms as Predictor of Poor Clinical Outcome in Patients With Vascular Cognitive Impairment. Am. J. Geriatr. Psychiatry 2022, 30, 813–824. [Google Scholar] [CrossRef]
  90. Swartz, R.H.; Sahlas, D.J.; Black, S.E. Strategic involvement of cholinergic pathways and executive dysfunction: Does location of white matter signal hyperintensities matter? J. Stroke Cerebrovasc. Dis. 2003, 12, 29–36. [Google Scholar] [CrossRef]
  91. Mesulam, M.; Siddique, T.; Cohen, B. Cholinergic denervation in a pure multi-infarct state: Observations on CADASIL. Neurology 2003, 60, 1183–1185. [Google Scholar] [CrossRef]
  92. Wallin, A.; Sjögren, M.; Blennow, K.; Davidsson, P. Decreased Cerebrospinal Fluid Acetylcholinesterase in Patients with Subcortical Ischemic Vascular Dementia. Dement. Geriatr. Cogn. Disord. 2003, 16, 200–207. [Google Scholar] [CrossRef]
  93. Nakham, A.; Myint, P.K.; Bond, C.M.; Newlands, R.; Loke, Y.K.; Cruickshank, M. Interventions to Reduce Anticholinergic Burden in Adults Aged 65 and Older: A Systematic Review. J. Am. Med. Dir. Assoc. 2020, 21, 172–180.e5. [Google Scholar] [CrossRef] [PubMed]
  94. Ailabouni, N.; Mangin, D.; Nishtala, P.S. DEFEAT-polypharmacy: Deprescribing anticholinergic and sedative medicines feasibility trial in residential aged care facilities. Int. J. Clin. Pharm. 2019, 41, 167–178. [Google Scholar] [CrossRef] [PubMed]
  95. Jester, D.J.; Molinari, V.; Zgibor, J.C.; Volicer, L. Prevalence of psychotropic polypharmacy in nursing home residents with dementia: A meta-analysis. Int. Psychogeriatr. 2021, 33, 1083–1098. [Google Scholar] [CrossRef] [PubMed]
  96. Maust, D.T.; Strominger, J.; Kim, H.M.; Langa, K.M.; Bynum, J.P.W.; Chang, C.-H.; Kales, H.C.; Zivin, K.; Solway, E.; Marcus, S.C. Prevalence of Central Nervous System–Active Polypharmacy Among Older Adults With Dementia in the US. JAMA 2021, 325, 952–961. [Google Scholar] [CrossRef]
  97. Toto, S.; Hefner, G.; Hahn, M.; Hiemke, C.; Roll, S.C.; Wolff, J.; Klimke, A. Current use of anticholinergic medications in a large naturalistic sample of psychiatric patients. J. Neural. Transm. (Vienna) 2021, 128, 263–272. [Google Scholar] [CrossRef]
  98. López-Álvarez, J.; Sevilla-Llewellyn-Jones, J.; Agüera-Ortiz, L. Anticholinergic Drugs in Geriatric Psychopharmacology. Front. Neurosci. 2019, 13, 1309. [Google Scholar] [CrossRef] [Green Version]
  99. Rudolph, J.L.; Salow, M.J.; Angelini, M.C.; McGlinchey, R.E. The anticholinergic risk scale and anticholinergic adverse effects in older persons. Arch. Inter. Med. 2008, 168, 508–513. [Google Scholar] [CrossRef] [Green Version]
  100. Taylor-Rowan, M.; Kraia, O.; Kolliopoulou, C.; Noel-Storr, A.H.; Alharthi, A.A.; Cross, A.J.; Stewart, C.; Myint, P.K.; McCleery, J.; Quinn, T.J. Anticholinergic burden for prediction of cognitive decline or neuropsychiatric symptoms in older adults with mild cognitive impairment or dementia. Cochrane Database Syst. Rev. 2022, 8, CD015196. [Google Scholar]
  101. Hampel, H.; Mesulam, M.M.; Cuello, A.C.; Farlow, M.R.; Giacobini, E.; Grossberg, G.T.; Khachaturian, A.S.; Vergallo, A.; Cavedo, E.; Snyder, P.J.; et al. The cholinergic system in the pathophysiology and treatment of Alzheimer’s disease. Brain. 2018, 141, 1917–1933. [Google Scholar] [CrossRef]
  102. Campbell, N.; Ayub, A.; Boustani, M.A.; Fox, C.; Farlow, M.; Maidment, I.; Howards, R. Impact of cholinesterase inhibitors on behavioral and psychological symptoms of Alzheimer’s disease: A meta-analysis. Clin. Interv. Aging 2008, 3, 719–728. [Google Scholar]
  103. Kertesz, A.; Morlog, D.; Light, M.; Blair, M.; Davidson, W.; Jesso, S.; Brashear, R. Galantamine in frontotemporal dementia and primary progressive aphasia. Dement. Geriatr. Cogn. Disord. 2008, 25, 178–185. [Google Scholar] [CrossRef]
  104. Mendez, M.F.; Shapira, J.S.; McMurtray, A.; Licht, E. Preliminary findings: Behavioral worsening on donepezil in patients with frontotemporal dementia. Am. J. Geriatr. Psychiatry 2007, 15, 84–87. [Google Scholar] [CrossRef]
  105. Grossberg, G.T.; Pejović, V.; Miller, M.L.; Graham, S.M. Memantine therapy of behavioral symptoms in community-dwelling patients with moderate to severe Alzheimer’s disease. Dement. Geriatr. Cogn. Disord. 2009, 27, 164–172. [Google Scholar] [CrossRef] [PubMed]
  106. Chen, R.; Chan, P.T.; Chu, H.; Lin, Y.C.; Chang, P.C.; Chen, C.Y.; Chou, K.R. Treatment effects between monotherapy of donepezil versus combination with memantine for Alzheimer disease: A meta-analysis. PLoS ONE 2017, 12, e0183586. [Google Scholar] [CrossRef]
  107. Maidment, I.D.; Fox, C.G.; Boustani, M.; Rodriguez, J.; Brown, R.C.; Katona, C.L. Efficacy of memantine on behavioral and psychological symptoms related to dementia: A systematic meta-analysis. Ann. Pharmacother. 2008, 42, 32–38. [Google Scholar] [CrossRef] [PubMed]
  108. Ishikawa, I.; Shinno, H.; Ando, N.; Mori, T.; Nakamura, Y. The effect of memantine on sleep architecture and psychiatric symptoms in patients with Alzheimer’s disease. Acta Neuropsychiatr. 2016, 28, 157–164. [Google Scholar] [CrossRef] [PubMed]
  109. Moran, S.P.; Maksymetz, J.; Conn, P.J. Targeting Muscarinic Acetylcholine Receptors for the Treatment of Psychiatric and Neurological Disorders. Trends Pharmacol. Sci. 2019, 40, 1006–1020. [Google Scholar] [CrossRef]
  110. Scarpa, M.; Hesse, S.; Bradley, S.J. Chapter Ten—M1 muscarinic acetylcholine receptors: A therapeutic strategy for symptomatic and disease-modifying effects in Alzheimer’s disease? Adv. Pharmacol. 2020, 88, 277–310. [Google Scholar]
  111. Levey, A.I.; Kitt, C.A.; Simonds, W.F.; Price, D.L.; Brann, M.R. Identification and localization of muscarinic acetylcholine receptor proteins in brain with subtype-specific antibodies. J. Neurosci. 1991, 11, 3218–3226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Medeiros, R.; Kitazawa, M.; Caccamo, A.; Baglietto-Vargas, D.; Estrada-Hernandez, T.; Cribbs, D.H.; Fisher, A.; LaFerla, F.M. Loss of muscarinic M1 receptor exacerbates Alzheimer’s disease-like pathology and cognitive decline. Am. J. Pathol. 2011, 179, 980–991. [Google Scholar] [CrossRef] [PubMed]
  113. Bodick, N.C.; Offen, W.W.; Shannon, H.E.; Satterwhite, J.; Lucas, R.; van Lier, R.; Paul, S.M. The selective muscarinic agonist xanomeline improves both the cognitive deficits and behavioral symptoms of Alzheimer disease. Alzheimer Dis. Assoc. Disord. 1997, 11 (Suppl. S4), S16–S22. [Google Scholar]
  114. Nathan, P.J.; Watson, J.; Lund, J.; Davies, C.H.; Peters, G.; Dodds, C.M.; Swirski, B.; Lawrence, P.; Bentley, G.D.; O’Neill, B.V.; et al. The potent M1 receptor allosteric agonist GSK1034702 improves episodic memory in humans in the nicotine abstinence model of cognitive dysfunction. Int. J. Neuropsychopharmacol. 2013, 16, 721–731. [Google Scholar] [CrossRef]
  115. Shirey, J.K.; Brady, A.E.; Jones, P.J.; Davis, A.A.; Bridges, T.M.; Kennedy, J.P.; Jadhav, S.B.; Menon, U.N.; Xiang, Z.; Watson, M.L.; et al. A selective allosteric potentiator of the M1 muscarinic acetylcholine receptor increases activity of medial prefrontal cortical neurons and restores impairments in reversal learning. J. Neurosci. 2009, 29, 14271–14286. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Davoren, J.E.; Lee, C.W.; Garnsey, M.; Brodney, M.A.; Cordes, J.; Dlugolenski, K.; Edgerton, J.R.; Harris, A.R.; Helal, C.J.; Jenkinson, S.; et al. Discovery of the Potent and Selective M1 PAM-Agonist N-[(3R,4S)-3-Hydroxytetrahydro-2H-pyran-4-yl]-5-methyl-4-[4-(1,3-thiazol-4-yl)benzyl]pyridine-2-carboxamide (PF-06767832): Evaluation of Efficacy and Cholinergic Side Effects. J. Med. Chem. 2016, 59, 6313–6328. [Google Scholar] [CrossRef]
  117. Rouse, S.T.; Thomas, T.M.; Levey, A.I. Muscarinic acetylcholine receptor subtype, m2: Diverse functional implications of differential synaptic localization. Life Sci. 1997, 60, 1031–1038. [Google Scholar] [CrossRef]
  118. Clader, J.W.; Wang, Y. Muscarinic receptor agonists and antagonists in the treatment of Alzheimer’s disease. Curr. Pharm. Des. 2005, 11, 3353–3361. [Google Scholar] [CrossRef]
  119. Verma, S.; Kumar, A.; Tripathi, T.; Kumar, A. Muscarinic and nicotinic acetylcholine receptor agonists: Current scenario in Alzheimer’s disease therapy. J. Pharm. Pharmacol. 2018, 70, 985–993. [Google Scholar] [CrossRef] [Green Version]
  120. Poulin, B.; Butcher, A.; McWilliams, P.; Bourgognon, J.-M.; Pawlak, R.; Kong, K.C.; Bottrill, A.; Mistry, S.; Wess, J.; Rosethorne, E.M.; et al. The M3-muscarinic receptor regulates learning and memory in a receptor phosphorylation/arrestin-dependent manner. Proc. Natl. Acad. Sci. USA 2010, 107, 9440. [Google Scholar] [CrossRef] [Green Version]
  121. Mao, L.M.; He, N.; Jin, D.Z.; Wang, J.Q. Regulation of Phosphorylation of AMPA Glutamate Receptors by Muscarinic M4 Receptors in the Striatum In vivo. Neuroscience 2018, 375, 84–93. [Google Scholar] [CrossRef]
  122. Tzavara, E.T.; Bymaster, F.P.; Davis, R.J.; Wade, M.R.; Perry, K.W.; Wess, J.; McKinzie, D.L.; Felder, C.; Nomikos, G.G. M4 muscarinic receptors regulate the dynamics of cholinergic and dopaminergic neurotransmission: Relevance to the pathophysiology and treatment of related CNS pathologies. FASEB J. 2004, 18, 1410–1412. [Google Scholar] [CrossRef] [PubMed]
  123. Chan, W.Y.; McKinzie, D.L.; Bose, S.; Mitchell, S.N.; Witkin, J.M.; Thompson, R.C.; Christopoulos, A.; Lazareno, S.; Birdsall, N.J.M.; Bymaster, F.P.; et al. Allosteric modulation of the muscarinic M4 receptor as an approach to treating schizophrenia. Proc. Natl. Acad. Sci. USA 2008, 105, 10978. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Nelson, A.B.; Bussert, T.G.; Kreitzer, A.C.; Seal, R.P. Striatal Cholinergic Neurotransmission Requires VGLUT3. J. Neurosci. 2014, 34, 8772. [Google Scholar] [CrossRef] [Green Version]
  125. English, D.F.; Ibanez-Sandoval, O.; Stark, E.; Tecuapetla, F.; Buzsáki, G.; Deisseroth, K.; Tepper, J.M.; Koos, T. GABAergic circuits mediate the reinforcement-related signals of striatal cholinergic interneurons. Nat. Neurosci. 2011, 15, 123–130. [Google Scholar] [CrossRef] [Green Version]
  126. Court, J.A.; Ballard, C.G.; Piggott, M.A.; Johnson, M.; O’Brien, J.T.; Holmes, C.; Cairns, N.; Lantos, P.; Perry, R.H.; Jaros, E.; et al. Visual hallucinations are associated with lower alpha bungarotoxin binding in dementia with Lewy bodies. Pharmacol. Biochem Behavr. 2001, 70, 571–579. [Google Scholar] [CrossRef]
  127. Van Kampen, M.; Selbach, K.; Schneider, R.; Schiegel, E.; Boess, F.; Schreiber, R. AR-R 17779 improves social recognition in rats by activation of nicotinic alpha7 receptors. Psychopharmacology 2004, 172, 375–383. [Google Scholar] [CrossRef]
  128. Boess, F.G.; De Vry, J.; Erb, C.; Flessner, T.; Hendrix, M.; Luithle, J.; Methfessel, C.; Riedl, B.; Schnizler, K.; van der Staay, F.J.; et al. The novel alpha7 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. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Cholinergic synapse. Mechanisms of action of cholinergic antagonists. 1. Acetylcholinesterase hydrolyzes ACh to inactive choline and acetate. 2. Cholinergic antagonists compete with Ach for binding to post-synaptic receptors. Non-competitive antagonism: the cholinergic agonist and antagonist can bind simultaneously, but the binding of the cholinergic antagonist reduces or inhibits the action of the agonist. Competitive antagonism: binding of the antagonist to the receptor prevents the binding of the agonist to that same receptor. 3.Presynaptic muscarinic autoreceptor performing a negative feedback loop in signal transduction.
Figure 1. Cholinergic synapse. Mechanisms of action of cholinergic antagonists. 1. Acetylcholinesterase hydrolyzes ACh to inactive choline and acetate. 2. Cholinergic antagonists compete with Ach for binding to post-synaptic receptors. Non-competitive antagonism: the cholinergic agonist and antagonist can bind simultaneously, but the binding of the cholinergic antagonist reduces or inhibits the action of the agonist. Competitive antagonism: binding of the antagonist to the receptor prevents the binding of the agonist to that same receptor. 3.Presynaptic muscarinic autoreceptor performing a negative feedback loop in signal transduction.
Ijms 24 06921 g001
Table 1. Summary of the main studies that have investigated the association between BPSD and AB, according to the type of NCD.
Table 1. Summary of the main studies that have investigated the association between BPSD and AB, according to the type of NCD.
Author, YearType of StudyAnticholinergic Burden Assessment Tool, if UsedOutcome
Alzheimer’s diseaseDauphinot, 2017 [57]Cross-sectionalADS, ACB, ARS, Chew’s score, Han’s scoreHigher NPI was associated with the Han’s score
Cancelli, 2008 [58]Cross-sectionalnoneAD patients exposed to AB were more likely to have psychosis
Hori, 2011 [59]Cross-sectionalSAAElevated SAA was linked to higher BEHAVE-AD *
Sunderland, 1987 [60]Cross-sectionalnoneAfter IV scopolamine 0.25 mg, AD patients showed anxiety, depression, and agitation
Jaïdi, 2018 [61]Cross-sectionalADS, ACB, ARSReducing AB by at least 20% enabled a significant decrease in BPSD
Liu, 2020 [64] ACBAnticholinergic usage was not associated with BPSD
Jewart, 2005 [65]Cross-sectionalSAANo significant relationship was found
Lewy body dementia
Parkinson Disease dementia
Jaïdi, 2018 [61] *Cross-sectionalADS, ACB, ARSReducing AB by at least 20% enabled a significant decrease in BPSD
Fronto-temporal dementiaxxxx
Vascular dementiaJaïdi, 2018 [61] ***Cross-sectional Reducing AB by at least 20% enabled a significant decrease in BPSD
ADS: Anticholinergic Drug Scale. ACB: anticholinergic cognitive burden. ARS: Anticholinergic Risk Scale. NPI: NeuriPscyhiatric Inventory. AD: Alzheimer’s disease. AB: anticholinergic burden. SAA: serum anticholinergic activity. BEHAVE-AD: Behavioral Pathology in Alzheimer’s Disease Rating Scale. AB: anticholinergic burden. * In particular paranoid and delusional ideations and diurnal rhythm disturbances. *** 13.6% of subjects included had VaD and almost 25% had mixed NCD (AD and VaD).
Table 2. Types of ACh receptors and potential therapeutic targets.
Table 2. Types of ACh receptors and potential therapeutic targets.
ReceptorCentral LocationMechanism of ActionInvolvement in Cognition/BPSDTherapeutic Target
Muscarinic
MCortex, hippocampus, striatum, and thalamus, post-synaptic [111]Coupled with protein Gq (activates phospholipase C)Memory
Learning [112]
M1 agonists:
-Xanomeline LY593093 improved cognitive function and reduced BPSD such as agitation, delusions, and hallucinations in patients with AD [113]
-GSK1034702 is an allosteric M1 agonist in clinical development for the treatment of cognitive disorders in AD (NCT00743405) [114]
-Heptares announced positive phase I results with the molecule HTL9936 (NCT02291783)
Positive allosteric M1 modulators:
-Benzyl quinolone carboxylic acid, BQCA, improved memory performance in patients with AD [115]
-PF-06767832: poor gastro-intestinal and cardiovascular tolerance [116]
M2Brainstem, thalamus, cortex, hippocampus and striatum [117]Coupled with protein Gi/o (inhibits adenylyl cyclase and modules ion channels)Memory
Learning
Delusions
Hallucinations [71]
Selective and active M2 antagonists M2 currently being optimized [118] in view of their poor tolerance [119]
M3Cortex and hippocampus [111]Coupled with protein Gq (activates phospholipase C)Memory
Learning
[120]
M4Striatum, cortex,
Hippocampus [111]
Coupled with protein Gi/o (inhibits adenylyl cyclase and modules ion channels)Regulation of ACh turnover [121,122]
M4 Agonists:
-Heptares announced the development of selective M4 and mixed M1/M4 agonists
Positive allosteric M4 Modulators:
-LY2033298 [123] but has not yet entered clinical development [116]
M5Locus niger [111]Coupled with protein Gq (activates phospholipase C)Delusions
Hallucinations [73]
NicotinicGlutamateric and dopaminergic neurons; GABAergic and cholinergic interneurons [124,125]Ionotropic
Delusions [70]
Hallucinations [126]
-AR-R17779 improved social recognition by activation of α7 nicotinic receptor [127]
-N-[128]oct-3-yl-7-[128]-1-benzofuran-2-carboxamide improves working memory and recognition [128]
BPSD: behavioral and psychological symptoms of dementia. AD: Alzheimer’s disease.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Mahmoudi, R.; Novella, J.L.; Laurent-Badr, S.; Boulahrouz, S.; Tran, D.; Morrone, I.; Jaïdi, Y. Cholinergic Antagonists and Behavioral Disturbances in Neurodegenerative Diseases. Int. J. Mol. Sci. 2023, 24, 6921. https://doi.org/10.3390/ijms24086921

AMA Style

Mahmoudi R, Novella JL, Laurent-Badr S, Boulahrouz S, Tran D, Morrone I, Jaïdi Y. Cholinergic Antagonists and Behavioral Disturbances in Neurodegenerative Diseases. International Journal of Molecular Sciences. 2023; 24(8):6921. https://doi.org/10.3390/ijms24086921

Chicago/Turabian Style

Mahmoudi, Rachid, Jean Luc Novella, Sarah Laurent-Badr, Sarah Boulahrouz, David Tran, Isabella Morrone, and Yacine Jaïdi. 2023. "Cholinergic Antagonists and Behavioral Disturbances in Neurodegenerative Diseases" International Journal of Molecular Sciences 24, no. 8: 6921. https://doi.org/10.3390/ijms24086921

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop