Next Article in Journal
Angiotensin II Infusion Leads to Aortic Dissection in LRP8 Deficient Mice
Next Article in Special Issue
Neuroprotective Effect of Cudrania tricuspidata Fruit Extracts on Scopolamine-Induced Learning and Memory Impairment
Previous Article in Journal
A Long-Day Photoperiod and 6-Benzyladenine Promote Runner Formation through Upregulation of Soluble Sugar Content in Strawberry
Previous Article in Special Issue
Acetylcholinesterase Inhibition of Diversely Functionalized Quinolinones for Alzheimer’s Disease Therapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Allosterism of Nicotinic Acetylcholine Receptors: Therapeutic Potential for Neuroinflammation Underlying Brain Trauma and Degenerative Disorders

1
Department of Pharmacology and Toxicology, The State University of New York at Buffalo, Buffalo, NY 14203, USA
2
Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
3
Department of Biochemistry and Neurology, Hunter James Kelly Research Institute, The State University of New York at Buffalo, Buffalo, NY 14203, USA
4
Department of Biology & Wildlife, University of Alaska Fairbanks, Fairbanks, AK 99775, USA
5
Department of Biomedical and Pharmaceutical Sciences, Idaho State University College of Pharmacy, Pocatello, ID 83209, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this article.
Int. J. Mol. Sci. 2020, 21(14), 4918; https://doi.org/10.3390/ijms21144918
Submission received: 30 June 2020 / Revised: 6 July 2020 / Accepted: 10 July 2020 / Published: 12 July 2020

Abstract

:
Inflammation is a key physiological phenomenon that can be pervasive when dysregulated. Persistent chronic inflammation precedes several pathophysiological conditions forming one of the critical cellular homeostatic checkpoints. With a steady global surge in inflammatory diseases, it is imperative to delineate underlying mechanisms and design suitable drug molecules targeting the cellular partners that mediate and regulate inflammation. Nicotinic acetylcholine receptors have a confirmed role in influencing inflammatory pathways and have been a subject of scientific scrutiny underlying drug development in recent years. Drugs designed to target allosteric sites on the nicotinic acetylcholine receptors present a unique opportunity to unravel the role of the cholinergic system in regulating and restoring inflammatory homeostasis. Such a therapeutic approach holds promise in treating several inflammatory conditions and diseases with inflammation as an underlying pathology. Here, we briefly describe the potential of cholinergic allosterism and some allosteric modulators as a promising therapeutic option for the treatment of neuroinflammation.

1. Introduction

Inflammation is the body’s defense mechanism that is pivotal for maintaining health status. Inflammation encompasses immune responses to noxious and harmful stimuli [1,2,3,4,5]. At the tissue level, inflammation involves the recruitment of plasma proteins, fluid and leukocytes into the damaged area resulting in vasodilation, a phenomenon characterized by a surge in blood flow, edema and increased vascular permeability [6]. Any damage signals are identified through transmembrane receptors such as Toll-like receptors (TLRs), the intracellular nucleotide-binding domain and leucine-rich-repeat-containing receptors (NLRs) [7,8,9]. Activation of these receptors often culminates in the potentiation of a critical transcription factor, the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) that leads to translocation of NF-κB into the nucleus for promoting the transcription of target genes. Transcription and translation enable the expression of a set of these target genes that are mostly pro-inflammatory cytokines such as interleukins (ILs), tumor necrosis factor-alpha (TNF-α) and others [10]. Inflammatory cytokines further coordinate with various cellular intermediates facilitating the recruitment of effector cells such as neutrophils and monocytes in the periphery and microglia and astrocytes in the central nervous system (CNS) [11]. These effector cells in turn undergo degranulation releasing reactive oxygen species (ROS), reactive nitric oxide species (RNS) and proteinases that promote the inflammatory process [11]. While infections are the primary factors underlying inflammation, injury or trauma and exposure to foreign matter can trigger inflammatory responses [12]. Emerging evidence shows that aging and environmental factors such as lifestyle, food habits and exercise can influence inflammation [13,14,15,16,17].
Inflammation is the underlying cause of several pathological conditions that impact more than 500 million patients across major pharmaceutical markets [2]. Despite significant strides in the development of therapeutic strategies against various inflammatory diseases, the major public health concern has been the link to a more persistent issue of chronic inflammation [2]. There is accruing evidence of the role of neuronal cholinergic mechanisms influencing neuroinflammatory cascades implicated in trauma and debilitating nervous system manifestations, such as clusters of neurodegenerative disorders, autoimmunity, and chronic nociception [4,18,19,20,21].
Historically, the cholinergic anti-inflammatory pathway emerged with the discovery of T-lymphocyte cytotoxicity via the muscarinic cholinergic system [22]. These claims were further corroborated in experiments demonstrating inhibition of T-cell differentiation and responses due to nicotine exposure [23,24]. Recent evidence points towards a nAChR mediated cholinergic regulation of inflammation in the CNS [2]. For example, the cholinergic anti-inflammatory pathway involving activation of nAChRs on the vagus nerve can inhibit cytokine release, thereby preventing their damaging effects, and has become an essential link between CNS and immunomodulation in response to immune challenges [25,26]. Recent studies have also shown that the cholinergic anti-inflammatory mechanisms extend beyond the aforementioned pathway involving discrete cascades in the CNS. α7 nAChR expression on primary dorsal root ganglion (DRG) neurons influences excitatory glutamatergic-signaling-mediating spinal analgesia [27,28,29,30]. Similarly, activation of α7 nAChR on astrocytes attenuates LPS-mediated upregulation of inflammatory cytokines through inhibition of the ubiquitous NF-κB inflammatory pathway [31]. Additionally, α7 involves phosphorylation of STAT3, a mediator of an anti-apoptotic cascade under inflammatory challenge conditions [32].
Pharmacological targeting of nAChRs has therefore gained traction in recent years to address its role in inflammation. Despite successful drug design, the molecules designed to target primary active (orthosteric) sites often have limitations, as these sites are highly conserved [33]. Thus, drugs designed for this site often end up having overlapping specificity. On the contrary, drugs targeted to the secondary (allosteric) binding sites carry greater pharmacological potential [33]. This review emphasizes the potential of cholinergic allosterism as an emerging approach to drug design for the treatment of chronic neuroinflammation. We predominantly focus on traumatic brain injury (TBI) and major neurodegenerative disorders, Alzheimer’s (AD), Parkinson’s (PD) and Multiple sclerosis (MS) in which inflammation forms a significant pathological basis [34]. We highlight the role of nAChRs in influencing the neuroinflammatory mechanisms underlying these pathophysiologies and how allosteric targeting of some of the nAChRs subtypes can formulate a potential therapeutic strategy for successfully treating these disorders.

2. Nicotinic Acetylcholine Receptors and Allosterism

The nAChRs are members of the Cys-loop superfamily of receptors that include GABAA, glycine and serotonin receptors [35,36,37]. nAChRs are large pentameric membrane-bound proteins of a molecular mass of ~290 KD. Each receptor is formed by combinations of five identical (i.e., homomeric) or different (i.e., heteromeric) subunits. The subunits that contribute to nAChRs are α (α2–α9) and β (β2–β4) [35]. The diversity of subunit configuration and assembly results in a myriad of receptor subtypes with varied sensitivity to ligands. Adding to the complexity and variability of nAChRs, each heteromeric receptor may potentially exhibit different stoichiometry based on the ratio of α:β subunits present and varied sensitivities to endogenous ligand (acetylcholine, ACh) [38]. One of the widely studied nAChRs and their stoichiometries are the α4β2 receptors, which comprise of high sensitive (HS)(α4)2(β2)3 and low sensitive (LS)(α4)3(β2)2 receptor organization [39,40]. Such stoichiometric variation coupled with context-dependent intrinsic biophysical properties and participation of other α or β subunits imparts unique heterogeneity in the physiological functionality of the nAChRs [38]. The predominant nAChR subtypes expressed in the CNS comprise of α4 and β2 subunits [36,41]. Additionally, α6β2 and α7 subtypes are abundantly found in the CNS, while α6β4 exhibit limited distribution [42]. Post-synaptically, they facilitate fast cholinergic neurotransmission, and pre-synaptically they modulate the release of other neurotransmitters and, therefore, play diverse roles ranging from cognition to modulation of neurotransmitters, and neuroprotection [43].
nAChRs are fast desensitizing ligand-gated ion channels that are activated by ligands binding at the orthosteric site [44,45,46]. Most therapeutic ligands are targeted towards the orthosteric site where they alter synaptic neurotransmission and receptor expression profiles. Thus, prolonged activation of nAChRs at the orthosteric site often results in tolerance or insensitivity to the drug [47], suggesting that suboptimal or non-orthosteric activation may be beneficial. Despite the layers of heterogeneity in nAChRs exhibited by its subunits, stoichiometries and biophysical properties, the ACh binding site, where all agonist, partial agonist or antagonist bind, is highly conserved [48]. This posits a limitation in the development of a clinically usable new ligand with adequate specificity across diseases in which nAChRs are involved, including inflammation. Hence, besides tolerance/insensitivity, ligands developed for orthosteric sites lack specificity and may lead to potential side effects [49]. However, allosteric ligands present a great potential, as they act through binding to a non-orthosteric site and activate nAChRs only in the presence of endogenous ligands such as ACh [49].
By definition, allosteric ligands bind to non-orthosteric sites and may possess intrinsic activity or may be ineffective by themselves, i.e., possess no intrinsic activity. The allosteric ligands that possess no intrinsic activity are also referred to as allosteric modulators (AMs) [33,49,50]. The number of allosteric sites present, and their structural diversity exclusively depends on the subunit type and the subunits that assemble structurally to form a functional receptor [51,52,53,54,55,56]. This allows diversity on the allosteric site, as well as potential tunability of nAChRs to produce greater specificity. Allosteric modulators/ligands can either enhance or reduce the nAChR responses induced by ACh [47]. Ligands that enhance the ACh responses are positive allosteric modulators (PAMs), whereas those that reduce ACh responses are known as negative allosteric modulators (NAMs) [47]. Allosteric modulators may impart their effects on ACh-induced responses by inducing a change in ACh potency or by altering efficacy and/or receptor opening probability without affecting baseline neurotransmission [47,57].
Different types of PAMs targeting α7 nAChRs are studied and classified as type I and II, based on the mechanism by which they modulate the receptors [58]. The type I PAM increases Ach-induced current amplitudes, potentially through increasing ACh potency and efficacy, with minimal effect on receptor kinetics. Type II PAMs, however, work by altering receptor kinetics, specifically by slowing the desensitization and deactivation, leading to prolonged activation of receptors [59]. Another class of PAMs exists that exhibit intrinsic activity, i.e., they can directly activate the receptors. PAMs that potentiate orthosteric agonist-induced currents and possess intrinsic activity are called agonist-PAMs or Ago-PAMs [60]. For detailed accounts of allosteric modulation, underlying mechanism and receptor states please refer to these reviews [61,62,63].

3. Neuroinflammation and Potential of Cholinergic Allosterism

In the brain, chronic inflammation is a persistent cellular anomaly that is associated with several neurological disorders [64]. Despite having divergence in inflammatory inducers specific to each disease, there is convergence in mechanisms underlying amplification of inflammatory processes [64]. Under optimal physiological conditions, microglia, the resident immune cells of the brain, exhibit a deactivated phenotype [65]. Alteration to a more activated inflammatory phenotype occurs in response to an immune stimulus [64]. A sustained stimulus disrupts the homeostatic balance in the inflammatory mechanisms inflicting a chain of events that often results in neurotoxic challenges involving ROS, RNS and pro-inflammatory cytokines manifesting into cytokine-mediated diseases [66,67]. Additionally, the neurotoxic events often cause reactive astrocytosis, increased vascular permeability, extravasation of proteins, blood–brain barrier alterations and axonal demyelination that aid in amplifying the underlying disease states [64]. Neuroinflammation forms an integral aspect of some of the most devastating neurological disorders that will be discussed in the following sections.

3.1. Alzheimer’s Disease (AD)

AD is an age-related devastating neurodegenerative condition that progressively impairs cognition, affective functionality and social well-being [68]. At the pathological level, AD is characterized by β-amyloid plaques and neurofibrillary tangles and symptomatically often involves dementia, speech impairments and disorientation, eventually leading to loss of self-care and death [68,69,70]. Epidemiological observations and postmortem analysis have corroborated inflammatory dysregulation as a major contributing factor to the disease state [71,72]. Decades of research have implicated various inflammatory mediators in the onset and progression of a persistent neuro-inflammatory cycle that initiates and exacerbates the AD pathology [73,74,75,76,77,78,79,80,81,82]. Age-related oxidative imbalance that might be further influenced through environmental triggers is often the starting point that leads to ROS and RNS production through the concerted action of several inflammatory cytokines such as IL-1, IL-6, TNF-α and oxidative enzymes such as NADH oxidase and nitric oxide synthase [69,73,74,75,76,77,78,79,80,81,82,83,84,85]. The disproportionate ratio of the oxidative species can further lead to exaggerated cytokine responses. Such incremental cytokine production over a period of time can synergistically produce distress signals to the astrocytes and microglia, which express more cytokines producing an inflammatory hotspot [86]. These anomalous mechanisms coupled with a genetic predisposition for AD disrupt the overall cellular homeostatic balance towards a more pathological phenotype [87].
Cholinergic hypofunction typical of AD is an established phenomenon and loss of cholinergic pathways is an important contributor to dementia pertaining to attention, spatial and episodic domains [88,89]. Using acetylcholinesterase (AChE) inhibitors has, therefore, proven to be one of the most viable therapeutic options for symptomatic improvement [90]. There is evidence on the role of nAChRs in mediating inflammatory mechanisms underlying AD [91]. The interactions of the cholinergic system and a ubiquitous neurotrophin, nerve growth factor (NGF) in the manifestation of AD is a well-known phenomenon [92,93,94,95,96]. The cholinergic neurons of the basal nucleus of Meynert are deprived of trophic support due to dysregulated retrograde transport of NGF. Such disruption of NGF transport mechanisms in conjunction with cholinergic transmission exacerbates β-amyloid mediated toxicity in cholinergic neurons [97,98,99,100]. Since NGF has an established role in influencing inflammatory mechanisms [101], the intertwining role of NGF and cholinergic transmission at the level of receptor pharmacology warrants investigation. Further, postmortem analysis has demonstrated up to a 50% reduction in α4β2 nAChRs in the brain of AD patients [102], early in pathogenesis [103]. Additionally, both α4 and α7 subunits exhibit reduced expression in autopsy samples of the human cerebral cortex of AD patients [104,105,106]. With such extensive involvement of the cholinergic system in AD pathophysiology, one of the pressing questions is regarding the pharmacological modulation of nAChRs in influencing the disease state. In-vitro administration of nicotinic agonists in PC12 cells and rat cortical neurons inhibits β-amyloid associated toxicity while chronic nicotine administration in a transgenic AD mouse model attenuates β-amyloidosis and neurite atrophy by decreasing astrogliosis, a phenomenon typical of neuroinflammation [107,108,109,110]. The neuroprotective mechanism against β-amyloid toxicity is mediated through the α7-Janus kinase 2 (JAK2) pathway that involves the association of JAK2 with α7 nAChR subsequently activating phosphoinositide 3-kinase (PI3K) and protein kinase B (Akt) phosphorylation [107,111,112]. These findings have further been supported through findings in the AD mouse model implicating an α7-regulated pro-survival cascade involving mitogen activated protein kinase (MAPK), Bcl2 and NF-κB. Blocking α7 receptors by specific antibodies causes neuroinflammation leading to Alzheimer’s-like symptoms in rodents [91]. Additionally, the interactions of α7 nAChRs with β-amyloid have been well documented [113,114,115] implicating nAChRs and their pharmacological targeting as a critical avenue for therapeutic value in AD.

3.2. Parkinson’s Disease (PD)

PD is a neurodegenerative disorder that progressively worsens with age [116]. Aggregation of α-synuclein in cells of the substantia nigra in the brain and loss of dopaminergic neurons are the major pathological hallmarks [117,118,119]. PD is associated with impaired movement and reduced cognitive function [120,121]. The etiology of PD can be attributed to a combination of environmental and genetic factors [122,123,124,125]. Nicotinic neuroprotection in PD is evidenced through several experimental studies [126,127,128,129]. Nicotine protects against nigrostriatal lesions caused by 6-hydroxydopamine (6-OHDA) and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) insults that recapitulate PD-like pathology [127,130,131,132,133,134]. 6-OHDA and MPTP produce neuronal atrophy through oxidative stress and exacerbated microglial activation mechanisms, which are essential for neuroinflammation [135]. Therefore, the role of nAChRs becomes critical in PD. The striatal nAChR subtypes (including α4β2) are also reduced due to nigrostriatal damage [136,137]. Further, post-mortem brain analysis of PD patients has revealed neuroinflammation as a contributing factor in α-synuclein mediated neurotoxicity and PD patients display higher striatal levels of TGF-β, IL-1β, IL-6, IFN-γ and IL-1 [138,139,140,141,142]. Pharmacological targeting of both α4β2 and α7 receptors [132,133,143] has shown neuroprotective effects in 6-OHDA-lesioned rats by partially modulating Parkinson’s pathology, while α4 knockout does not show nicotine-mediated neuroprotection of dopaminergic neurons [144] emphasizing the importance of nAChRs in modulating pathological endpoints. Moreover, nicotine administered both before and after 6-OHDA insult was shown to be more effective against partial, but not complete dopaminergic lesions in the substantia nigra [130].
In addition to the conventional nAChR subtypes, there is emerging evidence of the importance of α6 and α5 subunits in regulating dopaminergic transmission [145,146]. Recent research implicates cellular calcium imbalance [147,148] as a probable mechanism underlying nAChR regulation. nAChR-mediated calcium alterations lead to signal transduction involving PI3K and or JAK2/STAT3 signaling that eventually alters inflammatory endpoints and pro-survival cascades [148].

3.3. Multiple Sclerosis (MS)

MS is an inflammatory autoimmune disorder that is characterized by chronic inflammatory demyelination and axonal degeneration in the CNS [149]. This process occurs both in white matter and grey matter [150], which results in disruption of axonal transmission causing motor and sensory impairments [149]. Though the precise molecular events that lead to MS are unknown, inflammatory anomalies, a key phenomenon in the disease pathology, result in the breakdown of the blood–brain barrier vascular endothelium affecting myelin and oligodendrocytes [151,152]. Further, activation of lymphocytes, macrophages, dendritic cells, and microglia are observed at the onset of the disease [153,154]. A cholinergic role in neuroinflammation and MS is evident by the presence of acetylcholinesterase (AChE), an enzyme regulating ACh turnover, and its variants in neurons, blood cells, white matter, glia and lymphocytes [155,156,157]. Nicotine has been shown to inhibit experimental MS in rodents [158,159], suggesting immune function could be manipulated by targeting cholinergic pathways of immune cells by specific ligands, potentially AMs [160,161]. The most promising target thus far has been α7 nAChR. The immune cells expressing α7 also express a protein called RIC-3 (resistance to inhibitors of cholinesterase 3) that aids in the surface expression of α7 nAChR, influencing the disease state and are being considered as cholinergic targets of MS [162]. Additionally, neuroinflammation induced by lipopolysaccharide (LPS) insult in rats was inhibited through activation of α7 nAChR [163]. Inhibition of AChE is shown to curb inflammation by reducing lymphocyte proliferation and secretion of proinflammatory cytokines such as TNF-α, IL-B and IL-6 in mice, indicating that targeting AChE may provide a positive outcome in the treatment of MS [149,164,165]. In the experimental autoimmune encephalomyelitis (EAE) mouse model of human MS, choline acetyltransferase (ChAT) expression, an indicator for ACh presence/synthesis, in natural Killer (NK) cells is observed. ChAT positive cells promote migration of NK cells into the CNS, ameliorating the disease severity [166,167]. It was found that mature NK cells expressing ChAT were able to repress EAE induction and had a greater capacity to delay disease onset and decrease symptom severity compared to NK cells devoid of ChAT [168,169]. Using an agonist of α7 nAChRs in EAE showed that the activation of α7 is necessary for the suppression of EAE clinical severity [170]. These findings support other accounts that higher serum levels of ACh are observed in MS patients receiving treatments [171,172]. Hence there is substantial evidence suggesting that increasing cholinergic signaling in MS patients may ameliorate their symptoms.

3.4. Traumatic Brain Injury (TBI)

TBI is due to injury from an external force and is a major human health concern that impacts people of all ages [173]. TBI poses a major socioeconomic burden with devastating long-term consequences [174]. External trauma results in leakage of the blood–brain barrier (BBB) that initiates the infiltration of inflammatory cells [175,176,177]. Preclinical studies have established that TBI dysregulates cholinergic mechanisms that are characterized by reductions in choline uptake, choline acetyltransferase activity and vesicular acetylcholine transporter activity [178,179,180,181]. At the receptor level, α7 receptor expression is attenuated by 50% [181]. These results have further been substantiated in human TBI studies [182,183,184]. More recent evidence is emerging on the role of nAChRs in modulating the inflammatory profile in TBI [185,186]. nAChR α7 null mice exhibit potentiated levels of TNF-α and IL-β levels concomitant with a leaky BBB [156]. Whereas systemic administration of the nAChR α7 agonist PNU-282987 or the positive allosteric modulator PNU-120596 significantly attenuates TBI-triggered BBB compromise [156]. Further, quantitative autoradiography in TBI rats shows reduced α7, α4 and α3 binding in various brain regions and activating α7 results in a dampened inflammatory cytokine response [187,188].

4. Potential Allosteric Modulators as Therapeutics

The development of AMs-based therapeutics targeting nAChRs remains challenging because of the heterogeneous and unique role of nAChRs in promoting neuroinflammation. However, several selective and non-selective AMs are available that can be repurposed as lead molecules to design promising candidates. We believe the greater strategy would be to employ deconstruction-reconstruction (D-R) approaches to the existing AMs, generate analogs and screen them for positive or negative allosteric modulation. Briefly, the D-R approach involves fragmentation of known ligands, where each fragment can serve as a key pharmacophore. Upon analysis and optimization, a suitable fragment that enables orthosteric ligand to stabilize active (open) receptor conformation is selected for reconstruction. Reconstruction is relatively challenging as it not only calls for merging, linking or growing fragments to develop drugs but also to adhere to classical guidelines for maintaining physicochemical properties such as LogP, topological polar surface area, molecular weight, etc. [189,190,191].
Desformylfulstrabromide (dFBr) is a PAM of neuronal α4β2 receptors, first extracted from the bryozoan Flustra foliacea [192]. dFBr exhibits a bell-shaped dose-response curve where it enhances ACh induced currents in α4β2 receptors at lower concentrations and inhibits them at higher concentrations [193]. At potentiating concentrations, dFBr is thought to rescue the receptor from the desensitizing state leading to greater potentiation [193]. dFBr does not show any potentiation of nAChRs containing α3 or α7 receptors [52,194]. In rats and mice, dFBr was shown to reduce intravenous nicotine self-administration without supporting self-administration behavior [195] and compulsive-like behavior [196,197], respectively. In mouse models of neuropathic pain, dFBr potentiates antiallodynic responses of nicotine [198], suggesting that dFBr can be used in combination with an agonist or partial agonist to enhance or maintain cholinergic tone. In the same study, dFBr failed to affect allodynia when solely injected on its own. In vivo and in vitro activation of α4β2 nAChRs on mouse inflammatory macrophages by agonists are shown to alleviate inflammation-mediated neuropathic pain [199]. Also, dFBr has been found to relieve β-amyloid peptide (Aβ1–42) mediated loss of α4β2 function in oocytes [200]. This further solidifies the role of α4β2 and the potential of dFBr as a lead therapeutic molecule.
NS9283 is a selective PAM of α4β2 receptors, developed at Neurosearch Inc. Specifically, it enhances ACh-induced currents of LS stoichiometry [55,56]. Analogs of NS9283 have been shown to be selective for other variants of the receptor, including α4α5β2 [201]. Given the potential involvement of α5-containing receptors in neuroinflammation, these analogs of NS9283 may be valuable leads in developing selective and therapeutically useful anti-inflammatory AMs.
NS206 is a PAM selective for the α4 subunit [56] and it potentiates both the LS and HS stoichiometry. NS206 has similar potencies for α4β2 and α4β4 receptors but does not potentiate α3β4 or α7 receptors. The ability to potentiate HS α4β2 receptors is of particular advantage as they are thought to be involved in neurological disorders including neuroinflammation [56,202]. Unlike NS9283, NS206 enhances ACh efficacy rather than potency [203]. Both NS206 and NS9283 are selective for the α4 subunit, however, their binding sites have been mapped to different domains of the same subunit. When co-applied, their effects are additive [203].
Galantamine is a plant-based alkaloid from the amaryllis family [204]. It was first identified as an anti-acetylcholinesterase and later found to be a type I PAM for nAChRs [205,206]. Galantamine has been used clinically for Alzheimer’s disease [207] and has also been explored as a potential therapeutic option for Autism [208]. Evidence suggests that galantamine is a PAM for human α4β2 and α7 receptors. Galantamine was shown to enhance ACh responses by 22% in α7 receptors at lower concentrations and reduce them at higher concentrations, producing a bell-shaped dose-response curve [209], a typical feature of most PAMs. As galantamine exhibits both PAM and NAM activity at different concentrations, it is possible to construct analogs possessing either PAM or NAM activity without acetylcholinesterase activity [210]. This notion is further substantiated by a recent finding that galantamine’s [211] and a selective agonist’s [212] anti-inflammatory effects are mediated by α7 receptors. Similarly, other acetylcholinesterase inhibitors such as physostigmine [213] can also be explored for cholinergic anti-inflammatory activity. A recent conflicting study suggested that galantamine is not a PAM of either α4β2 or α7 receptors expressed in Xenopus oocytes and HEK 293 cells [209]. Nevertheless, galantamine does provide a substrate for designing a specific PAM or NAM that could enable the optimal functioning of the nAChRs for recovering aberrant inflammatory activation. Furthermore, the PNU series of PAMs (e.g., PNU-120596) and Ago-PAM (GAT-107) for α7 receptors are shown to reduce nociceptive behavior, neuropathic pain and thermal hyperalgesia [214,215,216,217] and could be potential leads for developing AMs in treating neuroinflammation.
Levamisole is an effective anthelmintic that binds to nematode muscle nAChRs [218]. Due to side effects like severe dermatological lesions, its use in humans is curbed, however, they are still used in veterinary medicine [218,219,220]. In humans, levamisole has also been used as an adjuvant in colon cancer therapy [221] and is a common intentional contaminant of cocaine [222,223]. Levamisole was identified as a PAM of α3 containing nAChRs. It potentiates ACh-induced responses at α3β2 receptors at lower concentrations and inhibits them at higher concentrations [224]. It also acts as a partial agonist of α3β4 receptors at very high concentration. Levamisole poses several serious side effects including elicitation of inflammatory diseases [225], however, they are not explicitly attributed to its PAM activity on nAChRs. It is possible to design receptor subtype-specific analogs of levamisole while retaining its PAM activity and minimizing observed side effects. Further thorough research is warranted before levamisole can be developed as an anti-inflammatory therapeutic agent.
Another promising compound, HEPES, is a commonly used buffering agent, that is shown to selectively potentiate HS α4β2 while slightly inhibiting LS α4β2 receptors [39]. Likewise, several piperidines (e.g., CMPI) and their analogs have been identified as potent, as well as selective α4β2 nAChR PAMs [53,54].

5. Conclusions

Appropriate drug candidates and cellular targets to combat dysregulation of fundamental inflammatory pathways underlying numerous inflammatory driven neurodegenerative conditions are currently lacking. With chronic inflammation being implicated in several devastating diseases, such as AD, PD, MS and TBI, a thorough screening of some of the cellular mediators that lie at the crossroads of inflammation needs to occur. Mounting evidence shows that cholinergic mechanisms significantly overlap with inflammatory cascades and carry the potential to modulate the molecular mechanisms and functional outcomes of inflammation. Current research on the neuro- immunomodulatory role of nAChRs has been minimal. Further, most AMs that are synthetically designed in the laboratories are often not pursued extensively with regards to their therapeutic potential. One of the primary reasons for this shortcoming is due to the lack of understanding of the structure-function relationship. For example, multiple allosteric binding sites have been proposed for dfBr that can presumably result in varied functional outcomes at the cellular level. This has however not been explored with respect to other AMs of nAChRs discussed here in the review. Thus, there remains a great potential in understanding how characterizing individual allosteric binding of these drug molecules can provide a resolution on the functional outputs augmenting their therapeutic potential. As this review highlights, nAChRs carry the prospect of being candidate targets to restore inflammatory homeostasis through allosteric regulation (Figure 1). This could potentially fill a gap in the therapeutic targeting of one of the key mediators of inflammatory conditions, thereby addressing a pressing public health issue of the current times.

Author Contributions

S.M., S.N.K. and M.M. conceptualized and wrote the manuscript with inputs from A.B.-I. and M.S. All authors have read and agreed to the published version of the manuscript.

Funding

The funding for publication cost of this article is provided by the Department of Biomedical and Pharmaceutical Sciences, Idaho State University College of Pharmacy.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

References

  1. Lennertz, R.C.; Kossyreva, E.A.; Smith, A.K.; Stucky, C.L. TRPA1 mediates mechanical sensitization in nociceptors during inflammation. PLoS ONE 2012, 7, e43597. [Google Scholar] [CrossRef] [PubMed]
  2. Bencherif, M. Neuronal nicotinic receptors as novel targets for inflammation and neuroprotection: Mechanistic considerations and clinical relevance. Acta Pharmacol. Sin. 2009, 30, 702–714. [Google Scholar] [CrossRef] [PubMed]
  3. Bencherif, M.; Lovette, M.E.; Fowler, K.W.; Arrington, S.; Reeves, L.; Caldwell, W.S.; Lippiello, P.M. RJR-2403: A nicotinic agonist with CNS selectivity I. In vitro characterization. J. Pharmacol. Exp. Ther. 1996, 279, 1413–1421. [Google Scholar] [PubMed]
  4. Weiner, H.L.; Selkoe, D.J. Inflammation and therapeutic vaccination in CNS diseases. Nature 2002, 420, 879–884. [Google Scholar] [CrossRef] [PubMed]
  5. Vigano, P.; Rabellotti, E.; Pagliardini, L.; Somigliana, E.; Candiani, M.; Vercellini, P. Progesterone Resistance, Aromatase, and Inflammation: The Important Relationships Between Hormones and Inflammation. Curr. Obstet. Gynecol. Rep. 2012, 1, 146–152. [Google Scholar] [CrossRef] [Green Version]
  6. Ashley, N.T.; Weil, Z.M.; Nelson, R.J. Inflammation: Mechanisms, Costs, and Natural Variation. Annu. Rev. Ecol. Evol. Syst. 2012, 43, 385–406. [Google Scholar] [CrossRef]
  7. Roach, J.C.; Glusman, G.; Rowen, L.; Kaur, A.; Purcell, M.K.; Smith, K.D.; Hood, L.E.; Aderem, A. The evolution of vertebrate Toll-like receptors. Proc. Natl. Acad. Sci. USA 2005, 102, 9577–9582. [Google Scholar] [CrossRef] [Green Version]
  8. Gao, W.; Xiong, Y.; Li, Q.; Yang, H. Inhibition of Toll-Like Receptor Signaling as a Promising Therapy for Inflammatory Diseases: A Journey from Molecular to Nano Therapeutics. Front. Physiol. 2017, 8. [Google Scholar] [CrossRef]
  9. Bell, J.K.; Mullen, G.E.D.; Leifer, C.A.; Mazzoni, A.; Davies, D.R.; Segal, D.M. Leucine-rich repeats and pathogen recognition in Toll-like receptors. Trends Immunol. 2003, 24, 528–533. [Google Scholar] [CrossRef]
  10. Ghosh, S.; May, M.J.; Kopp, E.B. NF-kappa B and Rel proteins: Evolutionarily conserved mediators of immune responses. Annu. Rev. Immunol. 1998, 16, 225–260. [Google Scholar] [CrossRef]
  11. Nathan, C. Points of control in inflammation. Nature 2002, 420, 846–852. [Google Scholar] [CrossRef] [PubMed]
  12. Medzhitov, R. Origin and physiological roles of inflammation. Nature 2008, 454, 428–435. [Google Scholar] [CrossRef] [PubMed]
  13. De Vries, M.A.; Klop, B.; Janssen, H.W.; Njo, T.L.; Westerman, E.M.; Castro Cabezas, M. Postprandial inflammation: Targeting glucose and lipids. Adv. Exp. Med. Biol. 2014, 824, 161–170. [Google Scholar] [CrossRef] [PubMed]
  14. Herieka, M.; Erridge, C. High-fat meal induced postprandial inflammation. Mol. Nutr. Food Res. 2014, 58, 136–146. [Google Scholar] [CrossRef] [PubMed]
  15. Choi, J.; Joseph, L.; Pilote, L. Obesity and C-reactive protein in various populations: A systematic review and meta-analysis. Obes. Rev. 2013, 14, 232–244. [Google Scholar] [CrossRef] [PubMed]
  16. You, T.; Arsenis, N.C.; Disanzo, B.L.; Lamonte, M.J. Effects of exercise training on chronic inflammation in obesity: Current evidence and potential mechanisms. Sports Med. 2013, 43, 243–256. [Google Scholar] [CrossRef]
  17. Gjevestad, G.O.; Holven, K.B.; Ulven, S.M. Effects of Exercise on Gene Expression of Inflammatory Markers in Human Peripheral Blood Cells: A Systematic Review. Curr. Cardiovasc. Risk Rep. 2015, 9, 34. [Google Scholar] [CrossRef] [Green Version]
  18. Davidson, A.; Diamond, B. Autoimmune diseases. N. Engl. J. Med. 2001, 345, 340–350. [Google Scholar] [CrossRef]
  19. Franklin, R.J.; Ffrench-Constant, C.; Edgar, J.M.; Smith, K.J. Neuroprotection and repair in multiple sclerosis. Nat. Rev. Neurol. 2012, 8, 624–634. [Google Scholar] [CrossRef]
  20. Laveti, D.; Kumar, M.; Hemalatha, R.; Sistla, R.; Naidu, V.G.; Talla, V.; Verma, V.; Kaur, N.; Nagpal, R. Anti-inflammatory treatments for chronic diseases: A review. Inflamm. Allergy Drug Targets 2013, 12, 349–361. [Google Scholar] [CrossRef]
  21. Liu, Q.; Whiteaker, P.; Morley, B.J.; Shi, F.-D.; Lukas, R.J. Distinctive Roles for α7*- and α9*-Nicotinic Acetylcholine Receptors in Inflammatory and Autoimmune Responses in the Murine Experimental Autoimmune Encephalomyelitis Model of Multiple Sclerosis. Front. Cell. Neurosci. 2017, 11. [Google Scholar] [CrossRef] [PubMed]
  22. Strom, T.B.; Deisseroth, A.; Morganroth, J.; Carpenter, C.B.; Merrill, J.P. Alteration of the cytotoxic action of sensitized lymphocytes by cholinergic agents and activators of adenylate cyclase. Proc. Natl. Acad. Sci. USA 1972, 69, 2995–2999. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Middlebrook, A.J.; Martina, C.; Chang, Y.; Lukas, R.J.; DeLuca, D. Effects of nicotine exposure on T cell development in fetal thymus organ culture: Arrest of T cell maturation. J. Immunol. 2002, 169, 2915–2924. [Google Scholar] [CrossRef] [Green Version]
  24. Nouri-Shirazi, M.; Guinet, E. Evidence for the immunosuppressive role of nicotine on human dendritic cell functions. Immunology 2003, 109, 365–373. [Google Scholar] [CrossRef] [PubMed]
  25. Pavlov, V.A.; Wang, H.; Czura, C.J.; Friedman, S.G.; Tracey, K.J. The cholinergic anti-inflammatory pathway: A missing link in neuroimmunomodulation. Mol. Med. 2003, 9, 125–134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Tracey, K.J. Physiology and immunology of the cholinergic antiinflammatory pathway. J. Clin. Investig. 2007, 117, 289–296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Shoop, R.D.; Martone, M.E.; Yamada, N.; Ellisman, M.H.; Berg, D.K. Neuronal acetylcholine receptors with alpha7 subunits are concentrated on somatic spines for synaptic signaling in embryonic chick ciliary ganglia. J. Neurosci. 1999, 19, 692–704. [Google Scholar] [CrossRef] [Green Version]
  28. Voitenko, L.P.; Voitenko, S.V.; Skok, M.V.; Purnyn, H.E.; Skok, V.I. Nicotinic acetylcholine receptor subtypes in rat superior cervical ganglion neurons as studied by sequential application of two alpha-subunit-specific antibodies. Neurosci. Lett. 2001, 303, 37–40. [Google Scholar] [CrossRef]
  29. Rau, K.K.; Johnson, R.D.; Cooper, B.Y. Nicotinic AChR in subclassified capsaicin-sensitive and -insensitive nociceptors of the rat DRG. J. Neurophysiol. 2005, 93, 1358–1371. [Google Scholar] [CrossRef]
  30. Genzen, J.R.; Van Cleve, W.; McGehee, D.S. Dorsal root ganglion neurons express multiple nicotinic acetylcholine receptor subtypes. J. Neurophysiol. 2001, 86, 1773–1782. [Google Scholar] [CrossRef]
  31. Patel, H.; McIntire, J.; Ryan, S.; Dunah, A.; Loring, R. Anti-inflammatory effects of astroglial α7 nicotinic acetylcholine receptors are mediated by inhibition of the NF-κB pathway and activation of the Nrf2 pathway. J. Neuroinflammation 2017, 14, 192. [Google Scholar] [CrossRef] [PubMed]
  32. Marrero, M.B.; Bencherif, M. Convergence of alpha 7 nicotinic acetylcholine receptor-activated pathways for anti-apoptosis and anti-inflammation: Central role for JAK2 activation of STAT3 and NF-kappaB. Brain Res. 2009, 1256, 1–7. [Google Scholar] [CrossRef] [PubMed]
  33. Abdel-Magid, A.F. Allosteric modulators: An emerging concept in drug discovery. ACS Med. Chem. Lett. 2015, 6, 104–107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Amor, S.; Puentes, F.; Baker, D.; van der Valk, P. Inflammation in neurodegenerative diseases. Immunology 2010, 129, 154–169. [Google Scholar] [CrossRef] [PubMed]
  35. Gotti, C.; Clementi, F. Neuronal nicotinic receptors: From structure to pathology. Prog. Neurobiol. 2004, 74, 363–396. [Google Scholar] [CrossRef] [PubMed]
  36. Hogg, R.C.; Raggenbass, M.; Bertrand, D. Nicotinic acetylcholine receptors: From structure to brain function. Rev. Physiol. Biochem. Pharmacol. 2003, 147, 1–46. [Google Scholar] [CrossRef] [PubMed]
  37. Corringer, P.J.; Le Novère, N.; Changeux, J.P. Nicotinic receptors at the amino acid level. Annu. Rev. Pharmacol. Toxicol. 2000, 40, 431–458. [Google Scholar] [CrossRef]
  38. Albuquerque, E.X.; Pereira, E.F.R.; Alkondon, M.; Rogers, S.W. Mammalian nicotinic acetylcholine receptors: From structure to function. Physiol. Rev. 2009, 89, 73–120. [Google Scholar] [CrossRef] [Green Version]
  39. Weltzin, M.M.; Huang, Y.; Schulte, M.K. Allosteric modulation of alpha4beta2 nicotinic acetylcholine receptors by HEPES. Eur. J. Pharmacol. 2014, 732, 159–168. [Google Scholar] [CrossRef] [Green Version]
  40. Zwart, R.; Vijverberg, H.P. Four pharmacologically distinct subtypes of alpha4beta2 nicotinic acetylcholine receptor expressed in Xenopus laevis oocytes. Mol. Pharmacol. 1998, 54, 1124–1131. [Google Scholar] [CrossRef] [Green Version]
  41. McGranahan, T.M.; Patzlaff, N.E.; Grady, S.R.; Heinemann, S.F.; Booker, T.K. α4β2 Nicotinic Acetylcholine Receptors on Dopaminergic Neurons Mediate Nicotine Reward and Anxiety Relief. J. Neurosci. 2011, 31, 10891–10902. [Google Scholar] [CrossRef] [Green Version]
  42. Hone, A.J.; McIntosh, J.M. Nicotinic acetylcholine receptors in neuropathic and inflammatory pain. FEBS Lett. 2018, 592, 1045–1062. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Posadas, I.; López-Hernández, B.; Ceña, V. Nicotinic receptors in neurodegeneration. Curr. Neuropharmacol. 2013, 11, 298–314. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Changeux, J.-P. The nicotinic acetylcholine receptor: A typical ‘allosteric machine’. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2018, 373, 20170174. [Google Scholar] [CrossRef] [PubMed]
  45. Lentz, T.L. Rabies virus binding to an acetylcholine receptor alpha-subunit peptide. J. Mol. Recognit. 1990, 3, 82–88. [Google Scholar] [CrossRef] [PubMed]
  46. Hueffer, K.; Khatri, S.; Rideout, S.; Harris, M.B.; Papke, R.L.; Stokes, C.; Schulte, M.K. Rabies virus modifies host behaviour through a snake-toxin like region of its glycoprotein that inhibits neurotransmitter receptors in the CNS. Sci. Rep. 2017, 7, 12818. [Google Scholar] [CrossRef] [PubMed]
  47. Williams, D.K.; Wang, J.; Papke, R.L. Positive allosteric modulators as an approach to nicotinic acetylcholine receptor-targeted therapeutics: Advantages and limitations. Biochem. Pharmacol. 2011, 82, 915–930. [Google Scholar] [CrossRef] [Green Version]
  48. Zoli, M.; Pucci, S.; Vilella, A.; Gotti, C. Neuronal and Extraneuronal Nicotinic Acetylcholine Receptors. Curr. Neuropharmacol. 2018, 16, 338–349. [Google Scholar] [CrossRef] [PubMed]
  49. Mohamed, T.S.; Jayakar, S.S.; Hamouda, A.K. Orthosteric and Allosteric Ligands of Nicotinic Acetylcholine Receptors for Smoking Cessation. Front. Mol. Neurosci. 2015, 8, 71. [Google Scholar] [CrossRef] [Green Version]
  50. Luttmann, E.; Ludwig, J.; Höffle-Maas, A.; Samochocki, M.; Maelicke, A.; Fels, G. Structural model for the binding sites of allosterically potentiating ligands on nicotinic acetylcholine receptors. ChemMedChem 2009, 4, 1874–1882. [Google Scholar] [CrossRef]
  51. Taly, A.; Corringer, P.-J.; Guedin, D.; Lestage, P.; Changeux, J.-P. Nicotinic receptors: Allosteric transitions and therapeutic targets in the nervous system. Nat. Rev. Drug Discov. 2009, 8, 733–750. [Google Scholar] [CrossRef] [PubMed]
  52. Sala, F.; Mulet, J.; Reddy, K.P.; Bernal, J.A.; Wikman, P.; Valor, L.M.; Peters, L.; König, G.M.; Criado, M.; Sala, S. Potentiation of human alpha4beta2 neuronal nicotinic receptors by a Flustra foliacea metabolite. Neurosci. Lett. 2005, 373, 144–149. [Google Scholar] [CrossRef] [PubMed]
  53. Springer, S.K.; Woodin, K.S.; Berry, V.; Boezio, A.A.; Cao, L.; Clarkin, K.; Harmange, J.C.; Hierl, M.; Knop, J.; Malmberg, A.B.; et al. Synthesis and activity of substituted carbamates as potentiators of the alpha4beta2 nicotinic acetylcholine receptor. Bioorg. Med. Chem. Lett. 2008, 18, 5643–5647. [Google Scholar] [CrossRef] [PubMed]
  54. Albrecht, B.K.; Berry, V.; Boezio, A.A.; Cao, L.; Clarkin, K.; Guo, W.; Harmange, J.C.; Hierl, M.; Huang, L.; Janosky, B.; et al. Discovery and optimization of substituted piperidines as potent, selective, CNS-penetrant alpha4beta2 nicotinic acetylcholine receptor potentiators. Bioorg. Med. Chem. Lett. 2008, 18, 5209–5212. [Google Scholar] [CrossRef] [PubMed]
  55. Timmermann, D.B.; Sandager-Nielsen, K.; Dyhring, T.; Smith, M.; Jacobsen, A.M.; Nielsen, E.; Grunnet, M.; Christensen, J.K.; Peters, D.; Kohlhaas, K.; et al. Augmentation of cognitive function by NS9283, a stoichiometry-dependent positive allosteric modulator of α2- and α4-containing nicotinic acetylcholine receptors. Br. J. Pharmacol. 2012, 167, 164–182. [Google Scholar] [CrossRef] [PubMed]
  56. Olsen, J.A.; Ahring, P.K.; Kastrup, J.S.; Gajhede, M.; Balle, T. Structural and functional studies of the modulator NS9283 reveal agonist-like mechanism of action at α4β2 nicotinic acetylcholine receptors. J. Biol. Chem. 2014, 289, 24911–24921. [Google Scholar] [CrossRef] [Green Version]
  57. Uteshev, V.V. The therapeutic promise of positive allosteric modulation of nicotinic receptors. Eur. J. Pharmacol. 2014, 727, 181–185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Grønlien, J.H.; Håkerud, M.; Ween, H.; Thorin-Hagene, K.; Briggs, C.A.; Gopalakrishnan, M.; Malysz, J. Distinct profiles of alpha7 nAChR positive allosteric modulation revealed by structurally diverse chemotypes. Mol. Pharmacol. 2007, 72, 715–724. [Google Scholar] [CrossRef] [Green Version]
  59. Thomsen, M.S.; Mikkelsen, J.D. Type I and II positive allosteric modulators differentially modulate agonist-induced up-regulation of α7 nicotinic acetylcholine receptors. J. Neurochem. 2012, 123, 73–83. [Google Scholar] [CrossRef]
  60. Foster, D.J.; Conn, P.J. Allosteric Modulation of GPCRs: New Insights and Potential Utility for Treatment of Schizophrenia and Other CNS Disorders. Neuron 2017, 94, 431–446. [Google Scholar] [CrossRef] [Green Version]
  61. Chatzidaki, A.; Millar, N.S. Allosteric modulation of nicotinic acetylcholine receptors. Biochem. Pharmacol. 2015, 97, 408–417. [Google Scholar] [CrossRef] [PubMed]
  62. Chang, Y.; Huang, Y.; Whiteaker, P. Mechanism of Allosteric Modulation of the Cys-loop Receptors. Pharmaceuticals 2010, 3, 2592–2609. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Changeux, J.P.; Christopoulos, A. Allosteric Modulation as a Unifying Mechanism for Receptor Function and Regulation. Cell 2016, 166, 1084–1102. [Google Scholar] [CrossRef] [PubMed]
  64. Glass, C.K.; Saijo, K.; Winner, B.; Marchetto, M.C.; Gage, F.H. Mechanisms underlying inflammation in neurodegeneration. Cell 2010, 140, 918–934. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Streit, W.J. Microglia as neuroprotective, immunocompetent cells of the CNS. Glia 2002, 40, 133–139. [Google Scholar] [CrossRef] [PubMed]
  66. Rahal, A.; Kumar, A.; Singh, V.; Yadav, B.; Tiwari, R.; Chakraborty, S.; Dhama, K. Oxidative stress, prooxidants, and antioxidants: The interplay. Biomed. Res. Int. 2014, 2014, 761264. [Google Scholar] [CrossRef] [Green Version]
  67. Aguilera, G.; Colín-González, A.L.; Rangel-López, E.; Chavarría, A.; Santamaría, A. Redox Signaling, Neuroinflammation, and Neurodegeneration. Antioxid. Redox Signal. 2017, 28, 1626–1651. [Google Scholar] [CrossRef] [PubMed]
  68. Bondi, M.W.; Edmonds, E.C.; Salmon, D.P. Alzheimer’s Disease: Past, Present, and Future. J. Int. Neuropsychol. Soc. 2017, 23, 818–831. [Google Scholar] [CrossRef] [Green Version]
  69. Hensley, K.; Maidt, M.L.; Yu, Z.; Sang, H.; Markesbery, W.R.; Floyd, R.A. Electrochemical analysis of protein nitrotyrosine and dityrosine in the Alzheimer brain indicates region-specific accumulation. J. Neurosci. 1998, 18, 8126–8132. [Google Scholar] [CrossRef]
  70. Weller, J.; Budson, A. Current understanding of Alzheimer’s disease diagnosis and treatment. F1000Research 2018, 7. [Google Scholar] [CrossRef] [Green Version]
  71. Gomez-Nicola, D.; Boche, D. Post-mortem analysis of neuroinflammatory changes in human Alzheimer’s disease. Alzheimers Res. Ther. 2015, 7, 42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Hopperton, K.E.; Mohammad, D.; Trépanier, M.O.; Giuliano, V.; Bazinet, R.P. Markers of microglia in post-mortem brain samples from patients with Alzheimer’s disease: A systematic review. Mol. Psychiatry 2018, 23, 177–198. [Google Scholar] [CrossRef] [PubMed]
  73. Mrak, R.E.; Griffin, W.S. Potential inflammatory biomarkers in Alzheimer’s disease. J. Alzheimers Dis. 2005, 8, 369–375. [Google Scholar] [CrossRef] [PubMed]
  74. Mrak, R.E.; Sheng, J.G.; Griffin, W.S. Glial cytokines in Alzheimer’s disease: Review and pathogenic implications. Hum. Pathol. 1995, 26, 816–823. [Google Scholar] [CrossRef]
  75. Griffin, W.S.; Stanley, L.C.; Ling, C.; White, L.; MacLeod, V.; Perrot, L.J.; White, C.L., 3rd; Araoz, C. Brain interleukin 1 and S-100 immunoreactivity are elevated in Down syndrome and Alzheimer disease. Proc. Natl. Acad. Sci. USA 1989, 86, 7611–7615. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Rogers, J.; Luber-Narod, J.; Styren, S.D.; Civin, W.H. Expression of immune system-associated antigens by cells of the human central nervous system: Relationship to the pathology of Alzheimer’s disease. Neurobiol. Aging 1988, 9, 339–349. [Google Scholar] [CrossRef]
  77. McGeer, P.L.; Schulzer, M.; McGeer, E.G. Arthritis and anti-inflammatory agents as possible protective factors for Alzheimer’s disease: A review of 17 epidemiologic studies. Neurology 1996, 47, 425–432. [Google Scholar] [CrossRef]
  78. Luterman, J.D.; Haroutunian, V.; Yemul, S.; Ho, L.; Purohit, D.; Aisen, P.S.; Mohs, R.; Pasinetti, G.M. Cytokine gene expression as a function of the clinical progression of Alzheimer disease dementia. Arch. Neurol. 2000, 57, 1153–1160. [Google Scholar] [CrossRef] [Green Version]
  79. Van der Wal, E.A.; Gómez-Pinilla, F.; Cotman, C.W. Transforming growth factor-beta 1 is in plaques in Alzheimer and Down pathologies. Neuroreport 1993, 4, 69–72. [Google Scholar] [CrossRef]
  80. Rogers, J.; Cooper, N.R.; Webster, S.; Schultz, J.; McGeer, P.L.; Styren, S.D.; Civin, W.H.; Brachova, L.; Bradt, B.; Ward, P.; et al. Complement activation by beta-amyloid in Alzheimer disease. Proc. Natl. Acad. Sci. USA 1992, 89, 10016–10020. [Google Scholar] [CrossRef] [Green Version]
  81. Wood, J.A.; Wood, P.L.; Ryan, R.; Graff-Radford, N.R.; Pilapil, C.; Robitaille, Y.; Quirion, R. Cytokine indices in Alzheimer’s temporal cortex: No changes in mature IL-1β or IL-1RA but increases in the associated acute phase proteins IL-6, α2-macroglobulin and C-reactive protein. Brain Res. 1993, 629, 245–252. [Google Scholar] [CrossRef]
  82. Du Yan, S.; Fu, J.; Soto, C.; Chen, X.; Zhu, H.; Al-Mohanna, F.; Collison, K.; Zhu, A.; Stern, E.; Saido, T.; et al. An intracellular protein that binds amyloid-β peptide and mediates neurotoxicity in Alzheimer’s disease. Nature 1997, 389, 689–695. [Google Scholar] [CrossRef] [PubMed]
  83. Reich, E.E.; Markesbery, W.R.; Roberts, L.J., 2nd; Swift, L.L.; Morrow, J.D.; Montine, T.J. Brain regional quantification of F-ring and D-/E-ring isoprostanes and neuroprostanes in Alzheimer’s disease. Am. J. Pathol. 2001, 158, 293–297. [Google Scholar] [CrossRef]
  84. Aksenov, M.Y.; Aksenova, M.V.; Butterfield, D.A.; Geddes, J.W.; Markesbery, W.R. Protein oxidation in the brain in Alzheimer’s disease. Neuroscience 2001, 103, 373–383. [Google Scholar] [CrossRef]
  85. Williamson, K.S.; Gabbita, S.P.; Mou, S.; West, M.; Pye, Q.N.; Markesbery, W.R.; Cooney, R.V.; Grammas, P.; Reimann-Philipp, U.; Floyd, R.A.; et al. The nitration product 5-nitro-gamma-tocopherol is increased in the Alzheimer brain. Nitric Oxide 2002, 6, 221–227. [Google Scholar] [CrossRef] [Green Version]
  86. Fakhoury, M. Microglia and Astrocytes in Alzheimer’s Disease: Implications for Therapy. Curr. neuropharmacol. 2018, 16, 508–518. [Google Scholar] [CrossRef]
  87. Su, F.; Bai, F.; Zhang, Z. Inflammatory Cytokines and Alzheimer’s Disease: A Review from the Perspective of Genetic Polymorphisms. Neurosci. Bull. 2016, 32, 469–480. [Google Scholar] [CrossRef] [Green Version]
  88. Auld, D.S.; Kornecook, T.J.; Bastianetto, S.; Quirion, R. Alzheimer’s disease and the basal forebrain cholinergic system: Relations to beta-amyloid peptides, cognition, and treatment strategies. Prog. Neurobiol. 2002, 68, 209–245. [Google Scholar] [CrossRef]
  89. Mufson, E.J.; Ginsberg, S.D.; Ikonomovic, M.D.; DeKosky, S.T. Human cholinergic basal forebrain: Chemoanatomy and neurologic dysfunction. J. Chem. Neuroanat. 2003, 26, 233–242. [Google Scholar] [CrossRef]
  90. Mehta, M.; Adem, A.; Sabbagh, M. New acetylcholinesterase inhibitors for Alzheimer’s disease. Int. J. Alzheimers Dis. 2012, 2012, 728983. [Google Scholar] [CrossRef]
  91. Skok, M.; Lykhmus, O. The Role of α7 Nicotinic Acetylcholine Receptors and α7-Specific Antibodies in Neuroinflammation Related to Alzheimer Disease. Curr. Pharm. Des. 2016, 22, 2035–2049. [Google Scholar] [CrossRef] [PubMed]
  92. Mufson, E.J.; Counts, S.E.; Perez, S.E.; Ginsberg, S.D. Cholinergic system during the progression of Alzheimer’s disease: Therapeutic implications. Expert Rev. Neurother. 2008, 8, 1703–1718. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Schliebs, R.; Arendt, T. The cholinergic system in aging and neuronal degeneration. Behav. Brain Res. 2011, 221, 555–563. [Google Scholar] [CrossRef] [PubMed]
  94. Cattaneo, A.; Calissano, P. Nerve growth factor and Alzheimer’s disease: New facts for an old hypothesis. Mol. Neurobiol. 2012, 46, 588–604. [Google Scholar] [CrossRef] [PubMed]
  95. Triaca, V.; Calissano, P. Impairment of the nerve growth factor pathway driving amyloid accumulation in cholinergic neurons: The incipit of the Alzheimer’s disease story? Neural Regen. Res. 2016, 11, 1553–1556. [Google Scholar] [CrossRef]
  96. Turnbull, M.T.; Boskovic, Z.; Coulson, E.J. Acute Down-regulation of BDNF Signaling Does Not Replicate Exacerbated Amyloid-β Levels and Cognitive Impairment Induced by Cholinergic Basal Forebrain Lesion. Front. Mol. Neurosci. 2018, 11, 51. [Google Scholar] [CrossRef] [Green Version]
  97. Cuello, A.C.; Bruno, M.A.; Bell, K.F. NGF-cholinergic dependency in brain aging, MCI and Alzheimer’s disease. Curr. Alzheimer Res. 2007, 4, 351–358. [Google Scholar] [CrossRef]
  98. Iulita, M.F.; Cuello, A.C. Nerve growth factor metabolic dysfunction in Alzheimer’s disease and Down syndrome. Trends Pharmacol. Sci. 2014, 35, 338–348. [Google Scholar] [CrossRef]
  99. Iulita, M.F.; Bistué Millón, M.B.; Pentz, R.; Aguilar, L.F.; Do Carmo, S.; Allard, S.; Michalski, B.; Wilson, E.N.; Ducatenzeiler, A.; Bruno, M.A.; et al. Differential deregulation of NGF and BDNF neurotrophins in a transgenic rat model of Alzheimer’s disease. Neurobiol. Dis. 2017, 108, 307–323. [Google Scholar] [CrossRef]
  100. 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]
  101. Minnone, G.; De Benedetti, F.; Bracci-Laudiero, L. NGF and Its Receptors in the Regulation of Inflammatory Response. Int. J. Mol. Sci. 2017, 18, 1028. [Google Scholar] [CrossRef] [PubMed]
  102. Warpman, U.; Nordberg, A. Epibatidine and ABT 418 reveal selective losses of α4β2 nicotinic receptors in Alzheimer brains. Neuroreport 1995, 6, 2419–2423. [Google Scholar] [CrossRef] [PubMed]
  103. Marutle, A.; Warpman, U.; Bogdanovic, N.; Lannfelt, L.; Nordberg, A. Neuronal nicotinic receptor deficits in Alzheimer patients with the Swedish amyloid precursor protein 670/671 mutation. J. Neurochem. 1999, 72, 1161–1169. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Guan, Z.-Z.; Zhang, X.; Ravid, R.; Nordberg, A. Decreased Protein Levels of Nicotinic Receptor Subunits in the Hippocampus and Temporal Cortex of Patients with Alzheimer’s Disease. J. Neurochem. 2000, 74, 237–243. [Google Scholar] [CrossRef]
  105. Burghaus, L.; Schütz, U.; Krempel, U.; de Vos, R.A.; Jansen Steur, E.N.; Wevers, A.; Lindstrom, J.; Schröder, H. Quantitative assessment of nicotinic acetylcholine receptor proteins in the cerebral cortex of Alzheimer patients. Brain Res. Mol. Brain Res. 2000, 76, 385–388. [Google Scholar] [CrossRef]
  106. Wevers, A.; Burghaus, L.; Moser, N.; Witter, B.; Steinlein, O.K.; Schütz, U.; Achnitz, B.; Krempel, U.; Nowacki, S.; Pilz, K.; et al. Expression of nicotinic acetylcholine receptors in Alzheimer’s disease: Postmortem investigations and experimental approaches. Behav. Brain Res. 2000, 113, 207–215. [Google Scholar] [CrossRef]
  107. Shaw, S.; Bencherif, M.; Marrero, M.B. Janus kinase 2, an early target of alpha 7 nicotinic acetylcholine receptor-mediated neuroprotection against Abeta-(1-42) amyloid. J. Biol. Chem. 2002, 277, 44920–44924. [Google Scholar] [CrossRef] [Green Version]
  108. Kihara, T.; Shimohama, S.; Sawada, H.; Kimura, J.; Kume, T.; Kochiyama, H.; Maeda, T.; Akaike, A. Nicotinic receptor stimulation protects neurons against β-amyloid toxicity. Ann. Neurol. 1997, 42, 159–163. [Google Scholar] [CrossRef]
  109. Medeiros, R.; Castello, N.A.; Cheng, D.; Kitazawa, M.; Baglietto-Vargas, D.; Green, K.N.; Esbenshade, T.A.; Bitner, R.S.; Decker, M.W.; LaFerla, F.M. α7 Nicotinic receptor agonist enhances cognition in aged 3xTg-AD mice with robust plaques and tangles. Am. J. Pathol. 2014, 184, 520–529. [Google Scholar] [CrossRef]
  110. Nordberg, A.; Hellström-Lindahl, E.; Lee, M.; Johnson, M.; Mousavi, M.; Hall, R.; Perry, E.; Bednar, I.; Court, J. Chronic nicotine treatment reduces β-amyloidosis in the brain of a mouse model of Alzheimer’s disease (APPsw). J. Neurochem. 2002, 81, 655–658. [Google Scholar] [CrossRef]
  111. Buckingham, S.D.; Jones, A.K.; Brown, L.A.; Sattelle, D.B. Nicotinic acetylcholine receptor signalling: Roles in Alzheimer’s disease and amyloid neuroprotection. Pharmacol. Rev. 2009, 61, 39–61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. De Jonge, W.J.; van der Zanden, E.P.; The, F.O.; Bijlsma, M.F.; van Westerloo, D.J.; Bennink, R.J.; Berthoud, H.R.; Uematsu, S.; Akira, S.; van den Wijngaard, R.M.; et al. Stimulation of the vagus nerve attenuates macrophage activation by activating the Jak2-STAT3 signaling pathway. Nat. Immunol. 2005, 6, 844–851. [Google Scholar] [CrossRef] [PubMed]
  113. Lilja, A.M.; Porras, O.; Storelli, E.; Nordberg, A.; Marutle, A. Functional interactions of fibrillar and oligomeric amyloid-β with alpha7 nicotinic receptors in Alzheimer’s disease. J. Alzheimers Dis. 2011, 23, 335–347. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Oz, M.; Lorke, D.E.; Yang, K.H.; Petroianu, G. On the interaction of β-amyloid peptides and α7-nicotinic acetylcholine receptors in Alzheimer’s disease. Curr. Alzheimer Res. 2013, 10, 618–630. [Google Scholar] [CrossRef]
  115. Lasala, M.; Fabiani, C.; Corradi, J.; Antollini, S.; Bouzat, C. Molecular Modulation of Human α7 Nicotinic Receptor by Amyloid-β Peptides. Front. Cell. Neurosci. 2019, 13. [Google Scholar] [CrossRef]
  116. Beitz, J.M. Parkinson’s disease: A review. Front. Biosci. (Sch. Ed.) 2014, 6, 65–74. [Google Scholar] [CrossRef]
  117. Spillantini, M.G.; Schmidt, M.L.; Lee, V.M.Y.; Trojanowski, J.Q.; Jakes, R.; Goedert, M. α-Synuclein in Lewy bodies. Nature 1997, 388, 839–840. [Google Scholar] [CrossRef]
  118. Dickson, D.W. Parkinson’s disease and parkinsonism: Neuropathology. Cold Spring Harb. Perspect. Med. 2012, 2. [Google Scholar] [CrossRef] [Green Version]
  119. Michel, P.P.; Toulorge, D.; Guerreiro, S.; Hirsch, E.C. Specific needs of dopamine neurons for stimulation in order to survive: Implication for Parkinson disease. FASEB J. 2013, 27, 3414–3423. [Google Scholar] [CrossRef] [Green Version]
  120. Jankovic, J. Parkinson’s disease: Clinical features and diagnosis. J. Neurol. Neurosurg. Psychiatry 2008, 79, 368–376. [Google Scholar] [CrossRef] [Green Version]
  121. Dubois, B.; Pillon, B. Cognitive deficits in Parkinson’s disease. J. Neurol. 1996, 244, 2–8. [Google Scholar] [CrossRef] [PubMed]
  122. Gatto, N.M.; Rhodes, S.L.; Manthripragada, A.D.; Bronstein, J.; Cockburn, M.; Farrer, M.; Ritz, B. α-Synuclein Gene May Interact with Environmental Factors in Increasing Risk of Parkinson’s Disease. Neuroepidemiology 2010, 35, 191–195. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Warner, T.T.; Schapira, A.H. Genetic and environmental factors in the cause of Parkinson’s disease. Ann. Neurol. 2003, 53, 16–23. [Google Scholar] [CrossRef] [PubMed]
  124. Goldman, S.M.; Quinlan, P.J.; Ross, G.W.; Marras, C.; Meng, C.; Bhudhikanok, G.S.; Comyns, K.; Korell, M.; Chade, A.R.; Kasten, M.; et al. Solvent exposures and parkinson disease risk in twins. Ann. Neurol. 2012, 71, 776–784. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Maulik, M.; Mitra, S.; Bult-Ito, A.; Taylor, B.E.; Vayndorf, E.M. Behavioral Phenotyping and Pathological Indicators of Parkinson’s Disease in C. elegans Models. Front. Genet. 2017, 8, 77. [Google Scholar] [CrossRef]
  126. Quik, M.; Bordia, T.; Huang, L.; Perez, X. Targeting nicotinic receptors for Parkinson’s disease therapy. CNS Neurol. Disord. Drug Targets 2011, 10, 651–658. [Google Scholar] [CrossRef] [Green Version]
  127. Quik, M.; O’Neill, M.; Perez, X. Nicotine neuroprotection against nigrostriatal damage: Importance of the animal model. Trends Pharmacol. Sci. 2007, 28, 229–235. [Google Scholar] [CrossRef]
  128. Quik, M.; O’Leary, K.; Tanner, C.M. Nicotine and Parkinson’s disease: Implications for therapy. Mov. Disord. 2008, 23, 1641–1652. [Google Scholar] [CrossRef] [Green Version]
  129. Lu, J.Y.D.; Su, P.; Barber, J.E.M.; Nash, J.E.; Le, A.D.; Liu, F.; Wong, A.H.C. The neuroprotective effect of nicotine in Parkinson’s disease models is associated with inhibiting PARP-1 and caspase-3 cleavage. PeerJ 2017, 5, 3933. [Google Scholar] [CrossRef] [Green Version]
  130. Costa, G.; Abin-Carriquiry, J.A.; Dajas, F. Nicotine prevents striatal dopamine loss produced by 6-hydroxydopamine lesion in the substantia nigra. Brain Res. 2001, 888, 336–342. [Google Scholar] [CrossRef]
  131. Parain, K.; Hapdey, C.; Rousselet, E.; Marchand, V.; Dumery, B.; Hirsch, E.C. Cigarette smoke and nicotine protect dopaminergic neurons against the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine Parkinsonian toxin. Brain Res. 2003, 984, 224–232. [Google Scholar] [CrossRef]
  132. Bordia, T.; McGregor, M.; Papke, R.L.; Decker, M.W.; McIntosh, J.M.; Quik, M. The α7 nicotinic receptor agonist ABT-107 protects against nigrostriatal damage in rats with unilateral 6-hydroxydopamine lesions. Exp. Neurol. 2015, 263, 277–284. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Quik, M.; Campos, C.; Bordia, T.; Strachan, J.-P.; Zhang, J.; McIntosh, J.M.; Letchworth, S.; Jordan, K. α4β2 Nicotinic receptors play a role in the nAChR-mediated decline in L-dopa-induced dyskinesias in parkinsonian rats. Neuropharmacology 2013, 71, 191–203. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Quik, M.; Chen, L.; Parameswaran, N.; Xie, X.; Langston, J.W.; McCallum, S.E. Chronic Oral Nicotine Normalizes Dopaminergic Function and Synaptic Plasticity in 1-Methyl-4-Phenyl-1,2,3,6-Tetrahydropyridine-Lesioned Primates. J. Neurosci. 2006, 26, 4681. [Google Scholar] [CrossRef] [PubMed]
  135. Wang, Q.; Liu, Y.; Zhou, J. Neuroinflammation in Parkinson’s disease and its potential as therapeutic target. Transl. Neurodegener. 2015, 4, 19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Kulak, J.M.; McIntosh, J.M.; Quik, M. Loss of nicotinic receptors in monkey striatum after 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine treatment is due to a decline in alpha-conotoxin MII sites. Mol. Pharmacol. 2002, 61, 230–238. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Quik, M.; Sum, J.D.; Whiteaker, P.; McCallum, S.E.; Marks, M.J.; Musachio, J.; McIntosh, J.M.; Collins, A.C.; Grady, S.R. Differential declines in striatal nicotinic receptor subtype function after nigrostriatal damage in mice. Mol. Pharmacol. 2003, 63, 1169–1179. [Google Scholar] [CrossRef] [Green Version]
  138. Mogi, M.; Harada, M.; Kondo, T.; Riederer, P.; Inagaki, H.; Minami, M.; Nagatsu, T. Interleukin-1β, interleukin-6, epidermal growth factor and transforming growth factor-α are elevated in the brain from parkinsonian patients. Neurosci. Lett. 1994, 180, 147–150. [Google Scholar] [CrossRef]
  139. Mogi, M.; Harada, M.; Kondo, T.; Riederer, P.; Nagatsu, T. Brain β2-microglobulin levels are elevated in the striatum in Parkinson’s diseaselevels are elevated in the striatum in Parkinson’s disease. J. Neural Transm. Park. Dis. Dement. Sect. 1995, 9, 87–92. [Google Scholar] [CrossRef]
  140. Blum-Degena, D.; Müller, T.; Kuhn, W.; Gerlach, M.; Przuntek, H.; Riederer, P. Interleukin-1β and interleukin-6 are elevated in the cerebrospinal fluid of Alzheimer’s and de novo Parkinson’s disease patients. Neurosci. Lett. 1995, 202, 17–20. [Google Scholar] [CrossRef]
  141. Mount, M.P.; Lira, A.; Grimes, D.; Smith, P.D.; Faucher, S.; Slack, R.; Anisman, H.; Hayley, S.; Park, D.S. Involvement of interferon-gamma in microglial-mediated loss of dopaminergic neurons. J. Neurosci. 2007, 27, 3328–3337. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Chao, Y.; Wong, S.C.; Tan, E.K. Evidence of inflammatory system involvement in Parkinson’s disease. Biomed. Res. Int. 2014, 2014, 308654. [Google Scholar] [CrossRef] [PubMed]
  143. Visanji, N.P.; O’Neill, M.J.; Duty, S. Nicotine, but neither the α4β2 ligand RJR2403 nor an α7 nAChR subtype selective agonist, protects against a partial 6-hydroxydopamine lesion of the rat median forebrain bundle. Neuropharmacology 2006, 51, 506–516. [Google Scholar] [CrossRef] [PubMed]
  144. Ryan, R.E.; Ross, S.A.; Drago, J.; Loiacono, R.E. Dose-related neuroprotective effects of chronic nicotine in 6-hydroxydopamine treated rats, and loss of neuroprotection in alpha4 nicotinic receptor subunit knockout mice. Br. J. Pharmacol. 2001, 132, 1650–1656. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Exley, R.; McIntosh, J.M.; Marks, M.J.; Maskos, U.; Cragg, S.J. Striatal α5 nicotinic receptor subunit regulates dopamine transmission in dorsal striatum. J. Neurosci. 2012, 32, 2352–2356. [Google Scholar] [CrossRef] [PubMed]
  146. Exley, R.; Maubourguet, N.; David, V.; Eddine, R.; Evrard, A.; Pons, S.; Marti, F.; Threlfell, S.; Cazala, P.; McIntosh, J.M.; et al. Distinct contributions of nicotinic acetylcholine receptor subunit α4 and subunit α6 to the reinforcing effects of nicotine. Proc. Natl. Acad. Sci. USA 2011, 108, 7577–7582. [Google Scholar] [CrossRef] [Green Version]
  147. Shen, J.-x.; Yakel, J.L. Nicotinic acetylcholine receptor-mediated calcium signaling in the nervous system. Acta Pharmacol. Sin. 2009, 30, 673–680. [Google Scholar] [CrossRef] [Green Version]
  148. Kawamata, J.; Suzuki, S.; Shimohama, S. Enhancement of nicotinic receptors alleviates cytotoxicity in neurological disease models. Ther. Adv. Chronic Dis. 2011, 2, 197–208. [Google Scholar] [CrossRef] [PubMed]
  149. Li, Y.; Hai, S.; Zhou, Y.; Dong, B.R. Cholinesterase inhibitors for rarer dementias associated with neurological conditions. Cochrane Database Syst. Rev. 2015, 3, Cd009444. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Petiet, A.; Aigrot, M.-S.; Stankoff, B. Gray and White Matter Demyelination and Remyelination Detected with Multimodal Quantitative MRI Analysis at 11.7T in a Chronic Mouse Model of Multiple Sclerosis. Front. Neurosci. 2016, 10, 491. [Google Scholar] [CrossRef] [Green Version]
  151. Aktas, O.; Waiczies, S.; Zipp, F. Neurodegeneration in autoimmune demyelination: Recent mechanistic insights reveal novel therapeutic targets. J. Neuroimmunol. 2007, 184, 17–26. [Google Scholar] [CrossRef] [PubMed]
  152. Bazan, N.; Halabi, A.; Ertel, M.; Petasis, N. Neuroinflammation. In Basic Neurochemistry: Molecular, Cellular, and Medical Aspects, 8th ed.; Brady, S., Siegel, G., Albers, R.W., Price, D., Eds.; Elsevier: New York, NY, USA, 2012; pp. 610–620. [Google Scholar]
  153. Bjelobaba, I.; Savic, D.; Lavrnja, I. Multiple Sclerosis and Neuroinflammation: The Overview of Current and Prospective Therapies. Curr. Pharm. Des. 2017, 23, 693–730. [Google Scholar] [CrossRef] [PubMed]
  154. Hemmer, B.; Kerschensteiner, M.; Korn, T. Role of the innate and adaptive immune responses in the course of multiple sclerosis. Lancet. Neurol. 2015, 14, 406–419. [Google Scholar] [CrossRef]
  155. Darvesh, S.; Leblanc, A.M.; Macdonald, I.R.; Reid, G.A.; Bhan, V.; Macaulay, R.J.; Fisk, J.D. Butyrylcholinesterase activity in multiple sclerosis neuropathology. Chem. Biol. Interact. 2010, 187, 425–431. [Google Scholar] [CrossRef] [PubMed]
  156. Dash, P.K.; Zhao, J.; Kobori, N.; Redell, J.B.; Hylin, M.J.; Hood, K.N.; Moore, A.N. Activation of Alpha 7 Cholinergic Nicotinic Receptors Reduce Blood–Brain Barrier Permeability following Experimental Traumatic Brain Injury. J. Neurosci. 2016, 36, 2809–2818. [Google Scholar] [CrossRef] [Green Version]
  157. Anglister, L.; Etlin, A.; Finkel, E.; Durrant, A.R.; Lev-Tov, A. Cholinesterases in development and disease. Chem. Biol. Interact. 2008, 175, 92–100. [Google Scholar] [CrossRef]
  158. Gao, Z.; Nissen, J.C.; Ji, K.; Tsirka, S.E. The experimental autoimmune encephalomyelitis disease course is modulated by nicotine and other cigarette smoke components. PLoS ONE 2014, 9, e107979. [Google Scholar] [CrossRef] [Green Version]
  159. Nizri, E.; Irony-Tur-Sinai, M.; Lory, O.; Orr-Urtreger, A.; Lavi, E.; Brenner, T. Activation of the cholinergic anti-inflammatory system by nicotine attenuates neuroinflammation via suppression of Th1 and Th17 responses. J. Immunol. 2009, 183, 6681–6688. [Google Scholar] [CrossRef] [Green Version]
  160. Fujii, T.; Mashimo, M.; Moriwaki, Y.; Misawa, H.; Ono, S.; Horiguchi, K.; Kawashima, K. Physiological functions of the cholinergic system in immune cells. J. Pharmacol. Sci. 2017, 134, 1–21. [Google Scholar] [CrossRef]
  161. Nicolussi, E.M.; Huck, S.; Lassmann, H.; Bradl, M. The cholinergic anti-inflammatory system limits T cell infiltration into the neurodegenerative CNS, but cannot counteract complex CNS inflammation. Neurobiol. Dis. 2009, 35, 24–31. [Google Scholar] [CrossRef]
  162. Treinin, M.; Papke, R.L.; Nizri, E.; Ben-David, Y.; Mizrachi, T.; Brenner, T. Role of the α7 Nicotinic Acetylcholine Receptor and RIC-3 in the Cholinergic Anti-inflammatory Pathway. Cent. Nerv. Syst. Agents Med. Chem. 2017, 17, 90–99. [Google Scholar] [CrossRef] [PubMed]
  163. Tyagi, E.; Agrawal, R.; Nath, C.; Shukla, R. Inhibitory role of cholinergic system mediated via alpha7 nicotinic acetylcholine receptor in LPS-induced neuro-inflammation. Innate Immun. 2010, 16, 3–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Nizri, E.; Hamra-Amitay, Y.; Sicsic, C.; Lavon, I.; Brenner, T. Anti-inflammatory properties of cholinergic up-regulation: A new role for acetylcholinesterase inhibitors. Neuropharmacology 2006, 50, 540–547. [Google Scholar] [CrossRef]
  165. Zabrodskii, P.F. Effect of acetylcholine on mortality of mice from sepsis and proinflammatory cytokine production. Bull. Exp. Biol. Med. 2011, 150, 340–342. [Google Scholar] [CrossRef] [PubMed]
  166. Gross, C.C.; Schulte-Mecklenbeck, A.; Wiendl, H.; Marcenaro, E.; Kerlero de Rosbo, N.; Uccelli, A.; Laroni, A. Regulatory Functions of Natural Killer Cells in Multiple Sclerosis. Front. Immunol. 2016, 7, 606. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Frebel, H.; Nindl, V.; Schuepbach, R.A.; Braunschweiler, T.; Richter, K.; Vogel, J.; Wagner, C.A.; Loffing-Cueni, D.; Kurrer, M.; Ludewig, B.; et al. Programmed death 1 protects from fatal circulatory failure during systemic virus infection of mice. J. Exp. Med. 2012, 209, 2485–2499. [Google Scholar] [CrossRef] [PubMed]
  168. Jiang, W.; Li, D.; Han, R.; Zhang, C.; Jin, W.-N.; Wood, K.; Liu, Q.; Shi, F.-D.; Hao, J. Acetylcholine-producing NK cells attenuate CNS inflammation via modulation of infiltrating monocytes/macrophages. Proc. Natl. Acad. Sci. USA 2017, 114, 6202–6211. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  169. Jørgensen, N.P.; Alstrup, A.K.; Mortensen, F.V.; Knudsen, K.; Jakobsen, S.; Madsen, L.B.; Bender, D.; Breining, P.; Petersen, M.S.; Schleimann, M.H.; et al. Cholinergic PET imaging in infections and inflammation using (11)C-donepezil and (18)F-FEOBV. Eur. J. Nucl. Med. Mol. Imaging 2017, 44, 449–458. [Google Scholar] [CrossRef]
  170. Nizri, E.; Brenner, T. Modulation of inflammatory pathways by the immune cholinergic system. Amino Acids 2013, 45, 73–85. [Google Scholar] [CrossRef]
  171. Puzanov, I.; Diab, A.; Abdallah, K.; Bingham, C.O.; Brogdon, C.; Dadu, R.; Hamad, L.; Kim, S.; Lacouture, M.E.; LeBoeuf, N.R.; et al. Managing toxicities associated with immune checkpoint inhibitors: Consensus recommendations from the Society for Immunotherapy of Cancer (SITC) Toxicity Management Working Group. J. ImmunoTher. Cancer 2017, 5, 95. [Google Scholar] [CrossRef] [Green Version]
  172. Brudno, J.N.; Kochenderfer, J.N. Toxicities of chimeric antigen receptor T cells: Recognition and management. Blood 2016, 127, 3321–3330. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Galgano, M.; Toshkezi, G.; Qiu, X.; Russell, T.; Chin, L.; Zhao, L.R. Traumatic Brain Injury: Current Treatment Strategies and Future Endeavors. Cell Transplant. 2017, 26, 1118–1130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Kayani, N.A.; Homan, S.; Yun, S.; Zhu, B.P. Health and economic burden of traumatic brain injury: Missouri, 2001–2005. Public Health Rep. 2009, 124, 551–560. [Google Scholar] [CrossRef]
  175. Shlosberg, D.; Benifla, M.; Kaufer, D.; Friedman, A. Blood-brain barrier breakdown as a therapeutic target in traumatic brain injury. Nat. Rev. Neurol. 2010, 6, 393–403. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Chodobski, A.; Zink, B.J.; Szmydynger-Chodobska, J. Blood-brain barrier pathophysiology in traumatic brain injury. Transl. Stroke Res. 2011, 2, 492–516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Donat, C.K.; Scott, G.; Gentleman, S.M.; Sastre, M. Microglial Activation in Traumatic Brain Injury. Front. Aging Neurosci. 2017, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Dixon, C.E.; Bao, J.; Bergmann, J.S.; Johnson, K.M. Traumatic brain injury reduces hippocampal high-affinity [3H]choline uptake but not extracellular choline levels in rats. Neurosci. Lett. 1994, 180, 127–130. [Google Scholar] [CrossRef]
  179. Dixon, C.E.; Flinn, P.; Bao, J.; Venya, R.; Hayes, R.L. Nerve growth factor attenuates cholinergic deficits following traumatic brain injury in rats. Exp. Neurol. 1997, 146, 479–490. [Google Scholar] [CrossRef]
  180. Donat, C.K.; Schuhmann, M.U.; Voigt, C.; Nieber, K.; Deuther-Conrad, W.; Brust, P. Time-dependent alterations of cholinergic markers after experimental traumatic brain injury. Brain Res. 2008, 1246, 167–177. [Google Scholar] [CrossRef]
  181. Titus, D.J.; Johnstone, T.; Johnson, N.H.; London, S.H.; Chapalamadugu, M.; Hogenkamp, D.; Gee, K.W.; Atkins, C.M. Positive allosteric modulation of the α7 nicotinic acetylcholine receptor as a treatment for cognitive deficits after traumatic brain injury. PLoS ONE 2019, 14, e0223180. [Google Scholar] [CrossRef] [Green Version]
  182. Shin, S.S.; Dixon, C.E. Alterations in Cholinergic Pathways and Therapeutic Strategies Targeting Cholinergic System after Traumatic Brain Injury. J. Neurotrauma 2015, 32, 1429–1440. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Murdoch, I.; Nicoll, J.A.; Graham, D.I.; Dewar, D. Nucleus basalis of Meynert pathology in the human brain after fatal head injury. J. Neurotrauma 2002, 19, 279–284. [Google Scholar] [CrossRef] [PubMed]
  184. Östberg, A.; Virta, J.; Rinne, J.O.; Oikonen, V.; Luoto, P.; Någren, K.; Arponen, E.; Tenovuo, O. Cholinergic dysfunction after traumatic brain injury: Preliminary findings from a PET study. Neurology 2011, 76, 1046–1050. [Google Scholar] [CrossRef] [PubMed]
  185. Ramlackhansingh, A.F.; Brooks, D.J.; Greenwood, R.J.; Bose, S.K.; Turkheimer, F.E.; Kinnunen, K.M.; Gentleman, S.; Heckemann, R.A.; Gunanayagam, K.; Gelosa, G.; et al. Inflammation after trauma: Microglial activation and traumatic brain injury. Ann. Neurol. 2011, 70, 374–383. [Google Scholar] [CrossRef] [PubMed]
  186. Gatson, J.W.; Simpkins, J.W.; Uteshev, V.V. High therapeutic potential of positive allosteric modulation of α7 nAChRs in a rat model of traumatic brain injury: Proof-of-concept. Brain Res. Bull. 2015, 112, 35–41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Verbois, S.L.; Scheff, S.W.; Pauly, J.R. Time-dependent changes in rat brain cholinergic receptor expression after experimental brain injury. J. Neurotrauma 2002, 19, 1569–1585. [Google Scholar] [CrossRef]
  188. Han, Z.; Li, L.; Wang, L.; Degos, V.; Maze, M.; Su, H. Alpha-7 nicotinic acetylcholine receptor agonist treatment reduces neuroinflammation, oxidative stress, and brain injury in mice with ischemic stroke and bone fracture. J. Neurochem. 2014, 131, 498–508. [Google Scholar] [CrossRef] [Green Version]
  189. Chen, H.; Zhou, X.; Wang, A.; Zheng, Y.; Gao, Y.; Zhou, J. Evolutions in fragment-based drug design: The deconstruction-reconstruction approach. Drug Discov. Today 2015, 20, 105–113. [Google Scholar] [CrossRef] [Green Version]
  190. Lipinski, C.A.; Lombardo, F.; Dominy, B.W.; Feeney, P.J. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv. Drug Deliv. Rev. 1997, 23, 3–25. [Google Scholar] [CrossRef]
  191. Veber, D.F.; Johnson, S.R.; Cheng, H.Y.; Smith, B.R.; Ward, K.W.; Kopple, K.D. Molecular properties that influence the oral bioavailability of drug candidates. J. Med. Chem. 2002, 45, 2615–2623. [Google Scholar] [CrossRef]
  192. Peters, L.; König, G.M.; Terlau, H.; Wright, A.D. Four new bromotryptamine derivatives from the marine bryozoan Flustra foliacea. J. Nat. Prod. 2002, 65, 1633–1637. [Google Scholar] [CrossRef] [PubMed]
  193. Weltzin, M.M.; Schulte, M.K. Pharmacological characterization of the allosteric modulator desformylflustrabromine and its interaction with alpha4beta2 neuronal nicotinic acetylcholine receptor orthosteric ligands. J. Pharmacol. Exp. Ther. 2010, 334, 917–926. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Kim, J.-S.; Padnya, A.; Weltzin, M.; Edmonds, B.W.; Schulte, M.K.; Glennon, R.A. Synthesis of desformylflustrabromine and its evaluation as an alpha4beta2 and alpha7 nACh receptor modulator. Bioorg. Med. Chem. Lett. 2007, 17, 4855–4860. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Liu, X. Positive allosteric modulation of α4β2 nicotinic acetylcholine receptors as a new approach to smoking reduction: Evidence from a rat model of nicotine self-administration. Psychopharmacology 2013, 230, 203–213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Mitra, S.; Mucha, M.; Khatri, S.N.; Glenon, R.; Schulte, M.K.; Bult-Ito, A. Attenuation of Compulsive-Like Behavior Through Positive Allosteric Modulation of α4β2 Nicotinic Acetylcholine Receptors in Non-Induced Compulsive-Like Mice. Front. Behav. Neurosci. 2016, 10, 244. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Schulte, M.K.; Glennon, R.A.; Bult-Ito, A.; Khatri, S.; Mitra, S. Compositions and Methods for Treating Compulsive-Like Behavior in a Subject. Google Patent 16/084291, 21 February 2019. [Google Scholar]
  198. Bagdas, D.; Ergun, D.; Jackson, A.; Toma, W.; Schulte, M.K.; Damaj, M.I. Allosteric modulation of alpha4beta2* nicotinic acetylcholine receptors: Desformylflustrabromine potentiates antiallodynic response of nicotine in a mouse model of neuropathic pain. Eur. J. Pain 2018, 22, 84–93. [Google Scholar] [CrossRef]
  199. Saika, F.; Kiguchi, N.; Kobayashi, Y.; Kishioka, S. Peripheral alpha4beta2 nicotinic acetylcholine receptor signalling attenuates tactile allodynia and thermal hyperalgesia after nerve injury in mice. Acta Physiol. 2015, 213, 462–471. [Google Scholar] [CrossRef]
  200. Pandya, A.; Yakel, J.L. Allosteric modulator Desformylflustrabromine relieves the inhibition of α2β2 and α4β2 nicotinic acetylcholine receptors by β-amyloid(1-42) peptide. J. Mol. Neurosci. 2011, 45, 42–47. [Google Scholar] [CrossRef] [Green Version]
  201. Jin, X.; Bermudez, I.; Steinbach, J.H. The nicotinic α5 subunit can replace either an acetylcholine-binding or nonbinding subunit in the α4β2* neuronal nicotinic receptor. Mol. Pharmacol. 2014, 85, 11–17. [Google Scholar] [CrossRef] [Green Version]
  202. Anderson, D.J.; Malysz, J.; Grønlien, J.H.; El Kouhen, R.; Håkerud, M.; Wetterstrand, C.; Briggs, C.A.; Gopalakrishnan, M. Stimulation of dopamine release by nicotinic acetylcholine receptor ligands in rat brain slices correlates with the profile of high, but not low, sensitivity alpha4beta2 subunit combination. Biochem. Pharmacol. 2009, 78, 844–851. [Google Scholar] [CrossRef]
  203. Olsen, J.; Kastrup, J.; Peters, D.; Gajhede, M.; Balle, T.; Ahring, P. Two Distinct Allosteric Binding Sites at α4β2 Nicotinic Acetylcholine Receptors Revealed by NS206 and NS9283 Give Unique Insights to Binding-Activity Associated Linkage at Cys-Loop Receptors. J. Biol. Chem. 2013, 288. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Moraes-Cerdeira, R.M.; Burandt, C.L., Jr.; Bastos, J.K.; Nanayakkara, D.; Mikell, J.; Thurn, J.; McChesney, J.D. Evaluation of four Narcissus cultivars as potential sources for galanthamine production. Planta Med. 1997, 63, 472–474. [Google Scholar] [CrossRef] [PubMed]
  205. Samochocki, M.; Höffle, A.; Fehrenbacher, A.; Jostock, R.; Ludwig, J.; Christner, C.; Radina, M.; Zerlin, M.; Ullmer, C.; Pereira, E.F.; et al. Galantamine is an allosterically potentiating ligand of neuronal nicotinic but not of muscarinic acetylcholine receptors. J. Pharmacol. Exp. Ther. 2003, 305, 1024–1036. [Google Scholar] [CrossRef] [Green Version]
  206. Pereira, E.F.; Alkondon, M.; Reinhardt, S.; Maelicke, A.; Peng, X.; Lindstrom, J.; Whiting, P.; Albuquerque, E.X. Physostigmine and galanthamine: Probes for a novel binding site on the alpha 4 beta 2 subtype of neuronal nicotinic acetylcholine receptors stably expressed in fibroblast cells. J. Pharmacol. Exp. Ther. 1994, 270, 768–778. [Google Scholar] [PubMed]
  207. Maelicke, A.; Samochocki, M.; Jostock, R.; Fehrenbacher, A.; Ludwig, J.; Albuquerque, E.X.; Zerlin, M. Allosteric sensitization of nicotinic receptors by galantamine, a new treatment strategy for Alzheimer’s disease. Biol. Psychiatry 2001, 49, 279–288. [Google Scholar] [CrossRef]
  208. Niederhofer, H.; Staffen, W.; Mair, A. Galantamine may be effective in treating autistic disorder. BMJ 2002, 325, 1422. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  209. Kowal, N.M.; Ahring, P.K.; Liao, V.W.Y.; Indurti, D.C.; Harvey, B.S.; O’Connor, S.M.; Chebib, M.; Olafsdottir, E.S.; Balle, T. Galantamine is not a positive allosteric modulator of human α4β2 or α7 nicotinic acetylcholine receptors. Br. J. Pharmacol. 2018, 175, 2911–2925. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  210. Texidó, L.; Ros, E.; Martín-Satué, M.; López, S.; Aleu, J.; Marsal, J.; Solsona, C. Effect of galantamine on the human alpha7 neuronal nicotinic acetylcholine receptor, the Torpedo nicotinic acetylcholine receptor and spontaneous cholinergic synaptic activity. Br. J. Pharmacol. 2005, 145, 672–678. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  211. Wazea, S.A.; Wadie, W.; Bahgat, A.K.; El-Abhar, H.S. Galantamine anti-colitic effect: Role of alpha-7 nicotinic acetylcholine receptor in modulating Jak/STAT3, NF-κB/HMGB1/RAGE and p-AKT/Bcl-2 pathways. Sci. Rep. 2018, 8, 5110. [Google Scholar] [CrossRef] [Green Version]
  212. Godin, J.R.; Roy, P.; Quadri, M.; Bagdas, D.; Toma, W.; Narendrula-Kotha, R.; Kishta, O.A.; Damaj, M.I.; Horenstein, N.A.; Papke, R.L.; et al. A silent agonist of alpha7 nicotinic acetylcholine receptors modulates inflammation ex vivo and attenuates EAE. Brain Behav. Immun. 2020, 87, 286–300. [Google Scholar] [CrossRef]
  213. Triggle, D.J.; Mitchell, J.M.; Filler, R. The Pharmacology of Physostigmine. CNS Drug Rev. 1998, 4, 87–136. [Google Scholar] [CrossRef]
  214. Freitas, K.; Negus, S.S.; Carroll, F.I.; Damaj, M.I. In vivo pharmacological interactions between a type II positive allosteric modulator of α7 nicotinic ACh receptors and nicotinic agonists in a murine tonic pain model. Br. J. Pharmacol. 2013, 169, 567–579. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  215. Freitas, K.; Ghosh, S.; Ivy Carroll, F.; Lichtman, A.H.; Imad Damaj, M. Effects of α7 positive allosteric modulators in murine inflammatory and chronic neuropathic pain models. Neuropharmacology 2013, 65, 156–164. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  216. Freitas, K.; Carroll, F.I.; Damaj, M.I. The antinociceptive effects of nicotinic receptors α7-positive allosteric modulators in murine acute and tonic pain models. J. Pharmacol. Exp. Ther. 2013, 344, 264–275. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  217. Bagdas, D.; Wilkerson, J.L.; Kulkarni, A.; Toma, W.; AlSharari, S.; Gul, Z.; Lichtman, A.H.; Papke, R.L.; Thakur, G.A.; Damaj, M.I. The α7 nicotinic receptor dual allosteric agonist and positive allosteric modulator GAT107 reverses nociception in mouse models of inflammatory and neuropathic pain. Br. J. Pharmacol. 2016, 173, 2506–2520. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  218. Robertson, S.J.; Martin, R.J. Levamisole-activated single-channel currents from muscle of the nematode parasite Ascaris suum. Br. J. Pharmacol. 1993, 108, 170–178. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  219. Robertson, S.J.; Pennington, A.J.; Evans, A.M.; Martin, R.J. The action of pyrantel as an agonist and an open channel blocker at acetylcholine receptors in isolated Ascaris suum muscle vesicles. Eur. J. Pharmacol. 1994, 271, 273–282. [Google Scholar] [CrossRef]
  220. Martin, R.J. Modes of action of anthelmintic drugs. Vet. J. 1997, 154, 11–34. [Google Scholar] [CrossRef]
  221. Macdonald, J.S. Adjuvant therapy of colon cancer. CA Cancer J. Clin. 1999, 49, 202–219. [Google Scholar] [CrossRef] [Green Version]
  222. Gaertner, E.M.; Switlyk, S.A. Dermatologic complications from levamisole-contaminated cocaine: A case report and review of the literature. Cutis 2014, 93, 102–106. [Google Scholar]
  223. Ching, J.A.; Smith, D.J., Jr. Levamisole-induced necrosis of skin, soft tissue, and bone: Case report and review of literature. J. Burn Care Res. 2012, 33, 1–5. [Google Scholar] [CrossRef] [PubMed]
  224. Levandoski, M.M.; Piket, B.; Chang, J. The anthelmintic levamisole is an allosteric modulator of human neuronal nicotinic acetylcholine receptors. Eur. J. Pharmacol. 2003, 471, 9–20. [Google Scholar] [CrossRef]
  225. Emil, N.S.; Cisneros, D.R.; Penmetsa, S.; Duchesne, J.H.; Sibbitt, W.L., Jr.; Gibb, J.I.; Noronha, L.E.; Fangtham, M.; Fields, R.A.; Bankhurst, A.D. Atypical Chronic Inflammatory ANCA-Positive Deforming Arthritis After Cocaine-Levamisole Exposure. J. Clin. Rheumatol. 2020, 26, 24–32. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Proposed model of allosteric modulation in inflammation: Allosteric modulators can potentially restore cholinergic transmission in the cells by binding to neuronal acetylcholine receptors (nAChRs). This can attenuate the exacerbated expression of predominant inflammatory mediators such as reactive oxygen species (ROS), reactive nitric oxide species (RNS), astrocytes, microglia and cytokines such as TGF-β (Transforming growth factor-beta), interleukin 1 (IL-1), interleukin 6 (IL-6) and interferon-gamma (IF-γ).
Figure 1. Proposed model of allosteric modulation in inflammation: Allosteric modulators can potentially restore cholinergic transmission in the cells by binding to neuronal acetylcholine receptors (nAChRs). This can attenuate the exacerbated expression of predominant inflammatory mediators such as reactive oxygen species (ROS), reactive nitric oxide species (RNS), astrocytes, microglia and cytokines such as TGF-β (Transforming growth factor-beta), interleukin 1 (IL-1), interleukin 6 (IL-6) and interferon-gamma (IF-γ).
Ijms 21 04918 g001

Share and Cite

MDPI and ACS Style

Mitra, S.; Khatri, S.N.; Maulik, M.; Bult-Ito, A.; Schulte, M. Allosterism of Nicotinic Acetylcholine Receptors: Therapeutic Potential for Neuroinflammation Underlying Brain Trauma and Degenerative Disorders. Int. J. Mol. Sci. 2020, 21, 4918. https://doi.org/10.3390/ijms21144918

AMA Style

Mitra S, Khatri SN, Maulik M, Bult-Ito A, Schulte M. Allosterism of Nicotinic Acetylcholine Receptors: Therapeutic Potential for Neuroinflammation Underlying Brain Trauma and Degenerative Disorders. International Journal of Molecular Sciences. 2020; 21(14):4918. https://doi.org/10.3390/ijms21144918

Chicago/Turabian Style

Mitra, Swarup, Shailesh N. Khatri, Malabika Maulik, Abel Bult-Ito, and Marvin Schulte. 2020. "Allosterism of Nicotinic Acetylcholine Receptors: Therapeutic Potential for Neuroinflammation Underlying Brain Trauma and Degenerative Disorders" International Journal of Molecular Sciences 21, no. 14: 4918. https://doi.org/10.3390/ijms21144918

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