Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Nov 1.
Published in final edited form as: CNS Drugs. 2018 Nov;32(11):981–996. doi: 10.1007/s40263-018-0572-y

The Cholinergic System as a Treatment Target for Opioid Use Disorder

Kevin P Jensen 1, Elise E DeVito 1, Sarah Yip 1, Kathleen M Carroll 1, Mehmet Sofuoglu 1
PMCID: PMC6314885  NIHMSID: NIHMS1002961  PMID: 30259415

Abstract

Opioid overdoses recently became the leading cause of accidental death in the United States, marking an increase in the severity of the opioid use disorder (OUD) epidemic that is impacting global health. Current treatment protocols for OUD are limited to opioid medications, including methadone, buprenorphine and naltrexone. While these medications are effective in many cases, new treatments are required to more effectively address the rising societal and interpersonal costs associated with OUD. Here we review the opioid and cholinergic systems, and we examine the potential of acetylcholine (ACh) as a treatment target for OUD. The cholinergic system includes enzymes that synthesize and degrade ACh and receptors that mediate ACh’s effects. ACh is involved in many central nervous system functions that are critical to the development and maintenance of OUD, such as reward and cognition. Medications that target the cholinergic system have been approved for the treatment of Alzheimer’s disease, tobacco use disorder and nausea. Clinical and preclinical studies suggest that medications such as cholinesterase inhibitors and scopolamine, which target components of the cholinergic system, show promise for the treatment of OUD and further investigations are warranted.

1. Introduction

The United States is currently facing an opioid use disorder (OUD) epidemic, which started with large increases in opioid prescriptions in 1990 and expanded by the widespread availability of heroin and synthetic opioids [1, 2]. In addition to affecting the United States, OUD is problematic in several other countries and significantly contributes to global disease burden [3]. The current epidemic has resulted in an estimated 2.1 million individuals in the United States with OUD in 2016 [4, 5]. In 2016, over 42,000 people died from opioid overdose, making it the leading cause of accidental death in the United States [6]. Medication assisted treatment (MAT), including methadone, buprenorphine and naltrexone, is effective in reducing opioid use, rate of OUD-associated infections, and psychosocial consequences of OUD [79]. However, high rates of attrition limit the effectiveness of MAT, underscoring the need to develop novel primary or adjunct treatments for OUD [7, 8].

As will be discussed in this review, among potential treatment targets for OUD, the brain cholinergic system shows a particular promise. Acetylcholine (ACh) participates in a wide range of central nervous system (CNS) functions that are thought to be critical in development and maintenance of OUD including reward, motivation, attention, mood, nociception, stress response and neuroimmune functions [1015]. Accumulating evidence from many studies support a close functional coupling between ACh and endogenous opioids. Further, preclinical and clinical studies suggest that medications targeting the cholinergic system may have utility for OUD treatment. This paper synthesizes studies that have examined the potential role of the cholinergic system as a treatment target for OUD. We first summarize clinical aspects of OUD, followed by current treatment approaches and clinical challenges. Next, we overview neurobiology, pharmacology and genetics of endogenous opioid and cholinergic system for CNS functions that are relevant for OUD. We then review preclinical and clinical studies that have examined the use of cholinergic medications for outcomes relevant for OUD. We conclude with a discussion of research gaps and future directions.

2. Overview of OUD

OUD is a chronic relapsing disorder characterized by compulsive and uncontrollable opioid use, most commonly heroin or prescription opioids. OUD increases the mortality rate of affected individuals 6 to 20 times over the general population, primarily due to overdose deaths [16]. Opioid overdose deaths are mainly due to respiratory depression; risk of overdose is accentuated by concurrent benzodiazepine use [17]. Typically, first exposure to opioids is through prescription opioids which is followed by non-prescription opioid and eventually heroin use [18]. Following initial exposure, individual vulnerability factors to develop OUD include depression, posttraumatic stress disorder (PTSD), presence of an additional substance use disorder and adolescence [19]. In addition, multiple genetic variations have been associated with the risk of developing OUD [2022]. OUD is highly comorbid with many psychiatric and medical problems including depression, anxiety disorders, PTSD, chronic pain, and infections including the human immunodeficiency virus and Hepatitis C Virus [2325].

3. Pharmacological treatment of OUD

The primary pharmacological approach for OUD is referred to as MAT, comprising methadone, buprenorphine and naltrexone. MAT reduces or eliminates opioid use, prevents overdose deaths, and reduces risk of contracting infections [26]. The principal limitation of MAT is high drop-out rates and subsequent relapse to opioid use. For buprenorphine and methadone maintenance treatments, retention rates at 1 year are typically less than 50% [7, 8]. Retention rates for injectable sustained-release naltrexone are even lower [2730]. In addition, long-term treatment with opioid medication is associated with adverse effects including cognitive deficits, endocrine disturbances, decreased libido and increased pain sensitivity or hyperalgesia [3134]. Thus, there is a great need to identify novel non-opioid medications for OUD treatment, including those that could be used alone or in combination with MAT.

4. Overview of the Opioid System

Endogenous opioids and their receptors are widely distributed in the CNS and peripheral tissues, reflecting their participation in multiple functions including reward, pain, emotion, cognition, and immune response. The endogenous opioid system includes four opioid receptors, mu (MOR), delta (DOR), kappa (KOR), and NOP, and 4 families of opioid peptides, β-endorphin, enkephalins, dynorphins and prepronociception [35]. Opioid receptors belong to the G-protein-coupled receptor superfamily and their activation leads to inhibition of cyclic adenosine monophosphate production and voltage-gated calcium channels, and activation of inwardly rectifying K+ channels and mitogen-activated protein kinase activity [36, 37]. These effects result in presynaptic inhibition of neurotransmitter release and inhibition of neuronal excitability. In the brain reward circuitry, MOR increases dopamine (DA) release by disinhibition; that is, inhibiting GABAergic interneurons which inhibit DA neurons [38].

4.1. Endogenous opioid peptides:

The endogenous opioid peptides are derived from 4 large precursor proteins (Table 1): 1) proopiomelanocortin (POMC), the precursor for β-endorphin; 2) preproenkephalin (PENK), the precursor for leucine (Leu)- and methionine (Met)-enkephalins; 3) preprodynorphin (PDYN), the precursor of dynorphins A and B, and neoendorphins [3941] and 4) prepronociception (PNOC), the precursor of nociceptin or orphanin FQ (N/OFQ) [42]. The POMC synthesizing neurons are located in the arcuate nucleus of the hypothalamus and the nucleus tractus solitarius in the dorsal medulla. The arcuate nucleus POMC neurons project to cortical and limbic regions, including the amygdala, hypothalamus, nucleus accumbens, periaqueductal gray and ventral tegmental area. In contrast, the POMC neurons located in the nucleus tractus solitarius project mainly to the spinal cord and brainstem. PENK and PDYN are synthesized locally in neurons in multiple regions of the CNS including the cortex, hippocampus, basal ganglia, thalamus, hypothalamus, periaqueductal gray area, ventral tegmental area, rostral ventromedial medulla, and the dorsal horn of the spinal cord. PNOC is expressed in multiple areas in the CNS including amygdala, thalamus, subthalamic nuclei, hypothalamus and basal ganglia [42]. β-endorphin, N/OFQ, and enkephalins are released in response to a multitude of painful or stressful stimuli [43]. Analgesia induced by physical and mental stressors is cross-tolerant with morphine and blocked by naloxone [44]. Overall, endogenous opioids have anti-stress effects mediated by MOR, DOR and KOR activation.

Table 1:

Endogenous opioid peptides and their high affinity receptors

PRECURSOR PEPTIDES RECEPTOR ACIVITY
POMC β-endorphin delta, mu
PENK leucine-enkephalin delta, mu
methionine -enkephalin
PDYN dynorphins A and B, and neoendorphins delta, kappa, mu
PNOC nociceptin NOP

POMC, proopiomelanocortin; PENK, preproenkephalin; PDYN, preprodynorphin; PNOC, prepronociception; NOP, nociception opioid peptide receptor.

4.2. Opioid receptors:

Opioid receptors are widely distributed in the central and peripheral neurons, neuroendocrine and immune cells [45]. In the CNS, opioid receptors are found in the ventral tegmental area, nucleus accumbens, hypothalamus, prefrontal cortex and amygdala. This distribution pattern is consistent with their participation in analgesia, reward, cognitive and emotional functions. The MOR, DOR, and KOR have differential sensitivity to endogenous opioid peptides. While β-endorphin has a high affinity to MOR and DOR, enkephalins and dynorphin have high affinity for DOR and KOR, respectively.

4.2.1. MOR:

MOR is the main target for the rewarding, analgesic and addictive effects of opioids like morphine, heroin, fentanyl and oxycodone. For example, unlike wild-type mice, MOR gene knockout mice do not show preference for morphine in a conditioned place preference task, do not self-administer morphine or heroin, and do not show signs of opioid withdrawal following chronic opioid exposure [46, 47]. MOR also has a key role in mediating rewarding effects from non-opioid drugs of abuse including alcohol, cocaine, nicotine and tetrahydrocannabinol. MOR knockout mice are less sensitive than wild-type mice to rewarding effects of these drugs of abuse [4850]. These effects are mediated mainly by MORs that are located on GABAergic inhibitory cells in the ventral tegmental area, which provides tonic inhibition to ventral tegmental area DA neurons. The inhibition of GABAergic cells by MOR (i.e., disinhibition), results in increased DA release in the nucleus accumbens [51]. In humans, naltrexone, an antagonist at MOR and to a lesser extent KOR and DOR, attenuates reward from food [52], physical activity [53], music [54] and drugs of abuse including cocaine [55], alcohol and nicotine [56].

4.2.2. DOR:

DOR does not seem to be essential for the rewarding effects of opioids, as morphine self-administration in DOR knockout mice is similar to wild-type mice [57]. However, DOR knockout mice show enhanced depressive-like behavior and DOR agonists show antidepressant effects in animal models of depression suggesting that DOR may be related to mood-regulating effects of opioids [58].

4.2.3. KOR:

Dynorphin A is the main endogenous KOR agonist. Dynorphin, through KOR activation, counteracts the rewarding effects of MOR in the ventral tegmental area and reduces DA release in the nucleus accumbens [59]. As a result, activation of KOR induces dysphoric effects and blocks rewarding effects from drugs of abuse including nicotine, alcohol, cocaine and tetrahydrocannabinol [6062].

4.2.4. NOP:

NOP (also known as ORL1) are not blocked by opioid antagonists, naloxone or naltrexone. The main endogenous ligand for NOP is a peptide named N/OFQ [35]. Similar to other opioid receptors, when activated, NOP inhibit adenylate cyclase, increase potassium conductance and reduce calcium conductance [63]. These inhibitory effects result in reduced neuronal activity and neurotransmitter release [64]. N/OFQ has complex effects on pain: it blocks morphine-induced anti-nociception and conditioned place preference when administered supraspinally [65], but has analgesic effects at the spinal cord in mice [66].

4.3. Molecular mechanisms of opioid effects:

Exposure to opioids leads to upregulation and desensitization of the opioid receptors [67]. Desensitization is a likely mechanism for development of opioid tolerance and includes several processes including receptor phosphorylation, uncoupling of receptors to G proteins, and internalization of receptors [67]. Phosphorylation of opioid receptors by G-protein-coupled receptor kinases increases their affinity for intracellular β-arrestin molecules. β-arrestin-opioid receptor complex formation results in uncoupling of G-proteins and facilitates opioid receptor internalization[68]. The internalization process disrupts the heterodimer between MOR and DOR, resulting in reduced function of opioid receptors. Opioid agonists differ in their ability to induce internalization resulting in different level of tolerance in response to chronic exposure [69].

4.4. Adaptations to chronic opioid exposure:

Chronic exposure to opioids leads to tolerance and withdrawal as the main components of physical dependence to opioids[68]. Multiple mechanisms for the development of opioid tolerance have been discussed, including uncoupling of MOR to co-effectors like G-proteins [70] and changes in secondary signaling cascades (e.g. upregulate of cyclic adenosine monophosphate-dependent signaling)[71]. Tolerance development leads to dose increases by the individuals in order to receive the desired effects from opioids (e.g., pain relief or euphoria). In the presence of physical dependence, abstinence (or significant reduction in opioid intake) leads to opioid withdrawal syndrome, which includes both physical and affective components [72]. Physical signs and symptoms include abdominal pain, diarrhea, vomiting, nasal discharge, enlarged pupils, pain and chills. Affective symptoms include dysphoria, anhedonia, anxiety/irritability, and craving. Depending on the elimination half-life of the particular opioid drug, physical components of opioid withdrawal syndrome subside within 1 to 2 weeks [73].

4.5. Insights into the opioid system from human genetics studies:

Genetic variation in genes that encode opioid receptors and endogenous opioid peptides has received much attention. The genes that encodes MOR, DOR, KOR and NOP in humans are OPRM1, OPRD1, OPRK1 and OPRL1, respectively. Genetic studies can be useful for understanding how certain genes function in humans, for prioritizing molecules as therapeutic targets, and for stratifying patients for precision medicine approaches. Genomewide association studies (GWAS), which interrogate most common variants in the genome without bias, are the current “gold-standard” method for conducting genetic association studies [74]. GWAS and other genetic studies of OUD and OUD treatment have been more extensively reviewed elsewhere by Jensen [75], and more recently by Crist and Berrettini [76, 77]. Among these genetic studies are several that highlight an important role of opioid receptors for OUD (Table 2). As discussed in prior sections, opioid receptors are important for several processes such as reward, pain, emotion, cognition, and immune response. MOR, in particular, is essential for the rewarding effects of drugs of abuse, including opioids and genetic variation in OPRM1, the gene encoding MOR, has been studied intensely. A nonsynonymous single nucleotide polymorphism (SNP), rs1799971, that causes an asparagine to aspartic acid substitution (Asn40Asp) in MOR, has been widely studied due to its possible effects on MOR function, although some in vitro studies suggest that effects on protein function are modest [78, 79] and might involve alternative mechanisms, such as effects on OPRM1 mRNA expression [80]. Moreover, genetic association studies of rs1799971 to OUD have yielded conflicting results, indicating that research on other variations encoded within OPRM1, and elsewhere in the genome, is warranted. For example, a recent study of heroin dependence that focused on 103 OPRM1 cis-eQTLs (i.e. SNPs associated with OPRM1 mRNA expression in brain tissue) identified a robust association (p = 4.3 × 10−8) with rs3778150, a SNP in the first intron of OPRM1[81]. This association was based on a meta-analysis that included 16,729 subjects and it was noted that rs1799971 was not associated with heroin dependence in any cohort or in the meta-analysis [81]. In a GWAS of methadone dose requirements by Smith et al., the most statistically robust association (p = 2.8 × 10−8) identified was for a SNP 5’ the OPRM1 transcription start site in an African American sample (n= 383) with OUD [82]. The SNP was also associated to morphine dose in a separate sample of opioid-naïve African American pediatric subjects (n=241) [82]. Crist et al. tested the association of several OPRM1 haplotypes to OUD treatment response and identified a SNP, rs10485058, in the OPRM1 3’ untranslated region that was associated with response to methadone in two European ancestor samples of OUD [83]. In an important extension of this work, they found that the SNP modified microRNA regulation of gene expression in a cell culture system [83]. In a separate study of variation in OPRD1, the gene encoding DOR, Crist et al. tested the association of on OUD treatment response to an OPRD1 SNP, rs678849, which had been previously linked to opioid dependence risk. They observed an association to treatment response in an African American sample with OUD that differed in terms of the effect direction based on the medication group (methadone or buprenorphine) [84]. Many association signals have emerged for GWAS of OUD-related phenotypes (e.g. OUD symptom count, sensitivity to opioids) that have implicated genes with no clear link to opioid signaling, and functional studies to elucidate the biological mechanisms are required [2022, 85].

Table 2:

Potentially clinically relevant genetic polymorphisms in opioid receptor genes

Gene SNP Potential Functional Effect Association findings
OPRM1 rs1799971 Missense Mutation (N40D) and OPRM1 mRNA levels Mixed for OUD and OUD treatment
rs3778150 OPRM1 mRNA levels Heroin dependence
rs73568641 None observed Methadone dose
rs10485058 Disrupts microRNA regulation of OPRM1 OUD treatment response
OPRD1 rs678849 None observed OUD treatment response

OUD, opioid use disorder; SNP, single nucleotide polymorphism; OPRM1, mu opioid receptor gene; OPRD1, delta opioid receptor gene. N, asparagine; D, aspartic acid.

5. Overview of Acetylcholine

ACh, the first discovered neurotransmitter, contributes to multiple CNS functions including sensory and motor processing, sleep, nociception, mood, stress response, attention, arousal, memory, motivation and reward [1014]. A large body of evidence supports the role of ACh in initiation and maintenance of addictive processes as well. It has been suggested that the DA/ACh balance in the nucleus accumbens may affect reward and aversion spectrum such that an increased ratio facilitates reward and decreased ratio generates an aversive state [86]. For example, drugs that increase ACh levels reduce self-administration of drugs of abuse including stimulants and opioids [87, 88]. Conversely, drug withdrawal states for opioids, cocaine and nicotine are associated with reduced DA and increased ACh levels [8991].

5.1. Endogenous Cholinergic System in the CNS:

In the cytoplasm of cholinergic neurons, ACh is synthesized from acetyl-coenzyme A and choline by choline acetyltransferase (ChAT) [92]. Following its release into the synaptic cleft, ACh signals through two classes of receptor: nicotinic (nAChR) or muscarinic (mAChR) type cholinergic receptors, which are both described in more detail below. ACh is rapidly inactivated by an enzyme, acetylcholinesterase (AChE), which is inhibited by a range of toxins and medications as well [93].

5.2. ACh Biosynthesis and distribution in the CNS:

Cholinergic neurons consist of two types: cholinergic interneurons and cholinergic projection neurons [94]. Cholinergic interneurons are located mainly in the striatum and modulate output from the basal ganglia. A subgroup of these neurons, the tonically-active striatal cholinergic interneurons, has important roles in stimulus salience and orienting functions [95].

The cholinergic projection neurons are located in the brainstem and the basal forebrain. The brainstem cholinergic neurons are located in pedunculopontine tegmental and the laterodorsal tegmental nuclei and project to the ventral tegmental area and thalamus [94]. These neurons modulate the sleep/wake cycle [96]. The basal forebrain cholinergic neurons are located in the nucleus basalis of Meynert, medial septal nucleus and vertical and horizontal limb nuclei of Broca [97]. These cholinergic neurons project to the hippocampus, amygdala, and cerebral cortex and modulate memory and attention functions [98]

5.3. Muscarinic receptors:

mAChRs are ACh receptors that are activated by muscarine. There are 5 types of muscarinic receptors that are classified into two groups: M1, M3 and M5 vs. M2 and M4 [99, 100]. Among these, M1, M2 and M4 are the main mAChR expressed in the CNS. M1, M3, and M5 mAChR are Gq-coupled and largely post-synaptic. They activate phospholipase C, intracellular calcium, inositol triphosphatase, and mitogen-activated protein kinase [100]. M1 mAChR, the predominant mAChR in the CNS, is implicated in learning and memory processes. Consistent with their functions, they are distributed in the cerebral cortex, hippocampus and striatum [101]. M3 mAChR are sparsely distributed in the CNS and their functions are not well-known [102] M5 receptors, expressed on the DA neurons in the ventral tegmental area and substantia nigra, facilitate DA release in the nucleus accumbens [103].

M2 and M4 mAChR are usually presynaptic and inhibit adenylyl cyclase and voltage-operated calcium channels and activate mitogen-activated protein kinases and G-protein-activated inwardly rectifying potassium channels M2 receptors are expressed in the brainstem, thalamus, cortex, hippocampus and striatum and inhibit ACh and DA release [101, 102, 104]. M4 mAChR are found in the midbrain, cortex, hippocampus, and striatum [101, 102]. Stimulation of M4 mAChR inhibits ventral tegmental area DA neurons, leading to reduced DA release in the nucleus accumbens [105].

The net effect of mAChR signaling is to reduce the number of synaptic inputs that neurons receive, resulting in increased responsivity for the remaining synaptic inputs [106]. This is achieved by increased membrane resistance and input sensitivity, through activation of M1, M3 and M5 receptors, and reduced neurotransmitter release, through activation of M2 and M4 mAChR. These effects are consistent with enhanced specificity of neuronal communication and memory encoding function of mAChRs [107, 108].

5.3.2. Nicotinic receptors:

nAChRs are ACh receptors that are activated by nicotine. nAChRs are ligand-gated ion channels arranged around a central pore, which is permeable to sodium, potassium, and calcium ions. nAChRs are comprised of pentameric combinations of α subunits (α2-α10) and β subunits (β2-β4) [109111]. They can be either homomeric nAChRs that consist of one type of α subunits (e.g., α6 or α7) or heteromeric nAChRs that consist of a combination of α and β subunits (e.g. α4β2, α3β4). α4β2 and α7 subtypes represent the majority of nAChRs in the brain [109111]. Most nAChRs in the brain are located presynaptically and increase the release of ACh, DA, serotonin, glutamate, GABA, and norepinephrine[112115]. Stimulation of α4β2 nAChR located on the DA cell bodies in the ventral tegmental area shifts these cells from tonic to phasic firing mode, which results in increased DA release in both the nucleus accumbens and the prefrontal cortex. β2-containing receptors are critical for the addictive as well as cognitive performance-enhancing properties of nicotine [116, 117]. Nicotine withdrawal has been shown to reduce brain reward function in rats, which may also be mediated by α4, β2, α7 subunits [118].

5.4. Insights into the cholinergic system from human genetics studies:

GWAS have highlighted some important functions for certain genes within the cholinergic system. There are 5 genes within the human genome that encode mAChRs (abbreviated as; CHRM1, CHRM2, CHRM3, CHRM4 and CHRM5), and 16 genes that encode nAChRs, including multiple alpha and multiple beta subunits (abbreviated as; CHRNA1-7,9,10, CHRNB1-4, CHRNE, CHRND, CHRNG). AChE, is encoded by the Acetylcholinesterase gene, which is abbreviated as AChE, and ChAT is encoded by the Choline O-acetyltransferase gene, which is abbreviated as CHAT. Among the most robust and well-characterized associations based on GWAS is genetic variation within the CHRNA5–A3–B4 gene cluster on chromosome 15 and measures of nicotine intake, such as the number of cigarettes smoked per day and cotinine levels [119, 120]. However, it is unclear whether genetic variation encoded within these genes is related to substance use disorder phenotypes like OUD. It is important to note that sample sizes for genetic studies of OUD have been smaller than many genetic studies of medical traits that have yield highly informative genetic associations. This likely limits the statistically power that is required to detect genetic effects associated with OUD. Also, whether genetic variants in the cholinergic system, (including SNPs with strong statistical links to clinically-relevant traits) affect the response to medication is a topic of ongoing research.

6. Interactions between opioids and ACh

Several lines of evidence suggest a close functional coupling between ACh and opioid transmission. An earlier study demonstrated that morphine administration in mice increased ACh concentration in striatum that coincided with the time-course of the analgesic effects of morphine [121]. Similar increases in ACh levels have also been observed in the spinal cord following morphine administration in monkeys [122]. In another study conducted in rats, ACh administration increased the release of beta-endorphin, Leu-enkephalin and dynorphin in the spinal cord [123]. Consistent with these findings, an increase in ACh level in the CNS produced by systemic administration of acetylcholine esterase inhibitors enhanced the analgesic effects of opioids (see next section for details). In contrast to the actions of MOR agonists on ACh release, N/OFQ reduced ACh release in cortical and hippocampal slices and NOP knock-out mice showed increased ACh release in hippocampus [124, 125]. These findings support the possible role of NOP receptors in hippocampal cholinergic function.

In a recent study, chronic morphine treatment increased ACh transmission in the laterodorsal tegmental nucleus (LDTg)/pedunculopontine tegmentum, which provide stimulatory cholinergic inputs to the ventral tegmental area DA cells [126]. This effect may represent one potential mechanism by which ACh transmission modulates neuroadaptation to chronic morphine exposure. On the other hand, chronic nicotine exposure attenuated the analgesic effects of opioids, suggesting a cross-tolerance between opioids and nicotine. In humans, opioids and nicotine products (e.g., tobacco cigarettes) are commonly abused together and smoking status is an important predictor for using higher doses of prescription opioids and misuse of prescription opioids [127, 128]. Together, these studies suggest that ACh and opioids may play an important role in modulating the pharmacological effects of each other as well as impact ongoing use and addiction to opioids and nicotine. The role of nAChR and mAChR in mediating the analgesic and rewarding effects of opioids remains controversial, partly due to lack of pharmacological specificity of currently available drugs targeting these receptors [126, 129]. The neural circuits mediating the effects ACh on opioid analgesia, reward, withdrawal and behavioral sensitization remain to be elucidated.

7. ACh as treatment target for OUD: Current Evidence

We performed a PubMed search between January to March 2018 to identify preclinical and clinical publications relevant to clarifying the role of the cholinergic system as a potential treatment target for OUD. The search was limited to English language articles. Preclinical studies were included 1) if they included outcomes related to OUD including opioid self-administration, conditioned place preference, opioid sensitization, opioid withdrawal and opioid analgesia and 2) used a cholinergic medication. Clinical studies were included if they examined opioid use, opioid withdrawal and opioid adverse effects. We included studies published between 1999 and 2018, as previous studies have been reviewed elsewhere [130, 131].

Preclinical and clinical studies examining the effects of cholinergic compounds on opioid-related outcomes are summarized in Table 3. The highlights from the table are described below. The majority of studies are preclinical. Regarding opioid reward, morphine self-administration was decreased by arecoline (AREC), a non-selective partial mAChR agonist, and by scopolamine, a mAChRs antagonist [132, 133]. Morphine-induced conditioned place preference was inhibited by donepezil or rivastigmine, which are both AChE inhibitors, and by scopolamine [134136]. Morphine-induced sensitization to locomotor effects was attenuated by mecamylamine, a nAChR antagonist [126], while morphine-induced behavioral sensitization was attenuated by huperzine A, an AChE inhibitor [137]. AREC also reduced reinstatement of drug seeking for morphine [133]. Regarding effects on analgesia, both donepezil or rivastigmine increased morphine’s acute analgesic effects [138]. Donepezil, rivastigmine and scopolamine each attenuated the development of tolerance to morphine’s analgesic effects [134, 138, 139]. In contrast, mecamylamine did not affect tolerance to morphine’s analgesic effects [126].

Table 3:

Clinical and preclinical studies of ACh targeting medications and opioid outcomes.

Study type Reference Drugs Procedures Outcome Key findings
Rodent Bajic et al. 2015 [126] Saline, morphine, mecamylamine The locomotor effects of chronic morphine either paired with saline or mecamylamine: The test day treatments included, saline or morphine paired with either saline or acute mecamylamine. Withdrawal induced by single injection of naloxone (20mg/kg ip) immediately after locomotor testing Antinociceptive effects of morphine compared among the following treatment groups: morphine, mecamylamine, morphine + mecamylamine, and saline. Drug exposure for each group was twice daily for 6.5 days. Sensitization to locomotor effects Mecamylamine (acute and chronic) reduced morphine-induced locomotor sensitization.
Pain perception There was no effect of mecamylamine on morphine induced antinociceptive tolerance.
Withdrawal (naloxone-induced) Mecamylamine did not reduce withdrawal symptoms overall.
Buccafusco et al 2000 [140] Morphine, DFP, echothiophate, AREC, naloxone Morphine dependence induced using morphine paired with saline, DFP (50 or 250 μg/kg daily), AREC (1 mg/kg, twice daily; s.c.), or echothiophate (2.5 μg/kg daily; s.c.). Withdrawal (naloxone-induced) DFP, AREC and echothiophate each separately reduced severity of withdrawal symptoms.
Buccafusco et al. 2007 [133] Morphine, arecoline hydrobromide (AREC), scopolamine Morphin SA and reinstatement during protracted morphine withdrawal comparing the following morphine SA phase treatments: saline, twice daily AREC (0.25 or 1 mg/kg s.c.), twice daily scopolamine (0.5 mg/kg) + AREC (1 mg/kg), and saline (days 1–9) + twice daily AREC (1 mg/kg) starting day 10. Self-administration AREC reduced morphine SA. Scopolamine did not modify the effect of AREC.
Reinstatement of drug-seeking AREC reduced ‘drug seeking’ lever presses during reinstatement. This effect was blocked by scopolamine co-treatment. AREC inhibited withdrawal symptoms (weight loss). This effect was reversed by scopolamine (i.e., scopolamine + AREC was similar to saline).
Gawel K et al. 2014 [136] Donepezil, rivastigmine, mecamylamine, scopolamine The effect of saline, donepezil or rivastigmine on CPP to morphine (5 mg/kg). Motivational Effects of Opioids Donepezil and rivastigmine attenuated acquisition and expression of mCPP and inhibited reinstatement of mCPP. Inhibition of reinstatement was reversed by mecamylamine but not scopolamine.
Gawel K et al. 2017 [138] Donepezil, rivastigmine, mecamylamine, Scopolamine Hot plate test with morphine (5 mg/kg) or saline + treatment with saline, donepezil or rivastigmine Tolerance to antinociceptive effects Donezepil and Rivastigmine each increased morphine’s antinociceptive effects. Effects reversed by scopolamine but not mecamylamine. Not influence analgesia in already morphine tolerant mice.
Hikida et al. 2003 [135] Morphine, donepezil, galantamine CPP and anti-nociceptive effects to morphine (1.0, 5.0 mg/kg), morphine + naloxone, and cocaine. Motivational and anti-nociceptive effects Ablation of cholinergic neurons increased acquisition of morphine CPP, mCPA following naloxone, withdrawal (jumping) after naloxone, anti-nociception effects. Donepezil reduced morphine CPP
Li et al. 2010[132] Morphine, scopolamine, nicotine Morphine (3.2–10 mg/kg), scopolamine (0.032–1.0 mg/kg) and nicotine (0.1–1 mg/kg) were administered alone or in combination to assess effect on lever pressing (previously paired with food reward). Drug reinforcement A single acute dose of scopolamine dose-dependently decreased responding to morphine (only significant at highest 1.0 dose)
Mukae T et al. 2015 [176] Pilocarpine, pirenzepine, donepezil, atropine Thermal paw withdrawal (latency); mechanical paw pressure test; electrical stimulation paw withdrawal Antinociceptive effects Pilocarpine (1 mg/kg) and donepezil (10 μg/kg) increased pain threshold and reduced allodynia. The effects of pilocarpine and donepezil were blocked by pirenzepine and atropine, respectively.
Neugebauer et al. 2013 [142] Saline, nicotine, lobeline, mecamylamine, morphine, and naloxone Morphine-tolerant mice, following their last morphine dose, were administered either saline, nicotine (0.2 or 0.4mg/kg), or lobeline (3 or 10 mg/kg) or mecamylamine (1 mg/kg). Withdrawal (naloxone-induced) Nicotine (0.4 mg/kg) or lobeline (3 mg/kg) decreased naloxone-induced jumping. No effect of mecamylamine.
Sharifipour M et al. 2014 [139] Donepezil The effects of donepezil on tail flick latency in morphine tolerant mice. Tolerance to antinociceptive effects Donepezil delayed tolerance to morphine’s analgesic effects and reduced the number of apoptotic cells on the cortex and lumbar spinal cord.
Sun et al. 2017 [137] huperzine A Effects of huper­zine A (0.2, 0.3 or 0.4 mg/kg) or saline on morphine‑induced behavioral sensitization. Sensitization to locomotor effects High doses of huperzine A when administered daily prior to morphine during the initial development of morphine dependence, attenuated morphine-induced locomotor activity
Xiang et al. 2006 [141] Morphine, scopolamine, naloxone Morphine (10, 20, 30, 40, or 50 mg/kg/injection) for 7 days, with or without scopolamine pre-treatment (0.5 mg/kg), or followed by 7 days of scopolamine (0.5 mg/kg). Withdrawal induced with naloxone. Withdrawal (naloxone-induced) Each scopolamine treatment condition reduced some symptoms of withdrawal. However, an increase in ‘wet dog shakes’ was noted for the pre-treatment condition.
Behavioral effects and drug clearance Each scopolamine treatment condition increased water consumption, urine volume and total volume of morphine excreted in urine.
Zhou et al. 1999 [134] Scopolamine, morphine, naloxone The morphine conditioning phase consisted of morphine (10 mg/kg) daily for 3 days. Animals received scopolamine (0.5 mg/kg) twice daily for 7 days prior to the morphine conditioning phase, or with scopolamine (0.5 mg/kg) 15 min before or 1 h after daily morphine treatment during the conditioning phase. Conditioned place preference Scopolamine, administered prior to or during the morphine conditioning phase, attenuated morphine-induced conditioned place preference
Animals were treated for 9 days with saline, 0.5 mg/kg scopolamine, or Pain perception Scopoloamine, prior to and along-side repeated morphine administrations, attenuated tolerance to morphine’s analgesic effects.
10 mg/kg morphine twice daily, or treated with scopolamine for 7 days prior to morphine treatment at 0.5 mg/kg 15 min before each morphine injection, or at 1.0 mg/kg once daily between two morphine injections.
The effect of pretreatment with scopolamine or saline on Morris Water Maze (prior to test, for 7 consecutive days). Cognition Scopolamine treated animals performed worse on Morris water maze (a measure of spatial learning and memory).
Saline, scopolamine (0.5 mg/kg), or morphine (10 mg/kg) twice daily for 9 days, or scopolamine (0.5 or 1.0 mg/kg) twice daily for 7 days prior to morphine treatment at 0.5 mg/kg 15 min before each morphine injection or at 1.0 mg/kg once daily between morphine injections. Withdrawal (naloxone-induced) Scopolamine attenuated naloxone-induced withdrawal symptoms.
Human Liu S et al. 2013 [145] Scopolamine, chlorpromazine Opioid dependent subjects were randomized to scopolamine detox (SD; n=46, 7 female) or methadone detox (MD, n=45, 5 female). SD given IV scopolamine + chlorpromazine under anesthesia. Withdrawal SD attenuated withdrawal and delayed relapse. The most common SD treatment adverse events was a confused state in 54% of participants.
Hooten WM et al. 2015 [144] Varenicline, placebo Randomized, single blind pilot study including chronic pain patients comparing varenicline to placebo. 11/11 placebo completers and 7/10 varenicline completers. Withdrawal No significant varenicline effects on withdrawal, pain or depression symptoms. Varenicline well-tolerated.
Slatkin NE et al. 2001 [143] Donepezil Cancer patients (n=6) receiving oral morphine (> 200 mg). Antinociceptive effects Donepezil reduced opioid-induced sedation and was well-tolerated without negatively impacting analgesia.
Poling et al. 2010 [166] Varenicline and placebo Cocaine, tobacco and opioid use was evaluated in a sample of 31 methadone-maintained tobacco smokers with opioid and cocaine dependence Tobacco, cocaine and opioid use No effect of varenicline on opioid positive urines compared to placebo. Varenicline reduced the number of cigarettes smoked per day.

AREC, arecoline; DFP, diisopropylflurophosphate; SA, self-administration; CPP, conditioned place preference; mCPP, morphine, conditioned place preference; CPA, conditioned place aversion; mCPA, morphine conditioned place aversion

Naloxone-induced opioid withdrawal symptoms were attenuated by diisopropylflurophosphate (DFP; an AChE inhibitor which acts both centrally and peripherally), echothiophate (a selective peripherally-acting AChE inhibitor), or AREC [140], or scopolamine [141]. Nicotine and the nAChR antagonist, lobeline, attenuated opioid-withdrawal symptoms induced by naloxone, but mecamylamine was not effective [142].

In human studies, donepezil reduced opioid-induced sedation without affecting analgesia in a sample of cancer patients (n=6) that were receiving high doses of opioids [143]. In another study of in-patients undergoing opioid taper, varenicline was well-tolerated. In one clinical study, a non-significant trend for decreased opioid withdrawal symptoms compared to placebo was noted [144]. In a separate clinical study, scopolamine was tested in 91 opioid-dependent patients undergoing opioid taper. Scopolamine, compared to placebo, reduced anxiety, depression, craving and prolonged time to relapse [145]. Overall, there are currently very limited clinical data to support a role of cholinergic agents in treating OUD.

8. Future directions for medication development

The preclinical and clinical studies summarized above support the promise of medications targeting the cholinergic system for the treatment of OUD. These include AChE inhibitors and medications targeting nAChR and mAChR. The adverse effects of opioid agonists (i.e., methadone or buprenorphine) include nausea, vomiting, constipation, endocrine disturbances, decreased libido and increased pain sensitivity or hyperalgesia, and possible cognitive deficits [3134]. Naltrexone’s adverse effects include nausea, headaches, insomnia, injection site pain (injectable form), elevation of transaminases, hypertension, nasopharyngitis, and influenza, possible depression or anhedonia [146, 147]. The most common adverse effects of AChE inhibitors include nausea, vomiting, diarrhea, loss of appetite, headache and dizziness [148]. For intravenous scopolamine, common adverse effects include reduced sweating, dry skin, dry mouth, amnesia, somnolence and less commonly hallucinations and confusion [149]. Overall, cholinesterase inhibitors have a long-established safety profile and their use for OUD is feasible. The more serious adverse effects of scopolamine (e.g., confusion or hallucinations) require closed monitoring of the adverse effects at the time and after infusion. Recent technological advances, such as in genomics, can facilitate the development of new medications by identifying and prioritizing drug targets, helping improve outcomes for established treatments (i.e., by patient stratification), and capturing in-depth treatment responses (e.g. biomarkers) to assist in evaluating efficacy [150, 151]. As such, it is a promising time to develop and evaluate new medications for OUD treatment.

8.1. Medications Targeting AChE

AChE inhibitors rivastigmine, donepezil, and galantamine, are marketed for the treatment of dementia [93, 152, 153]. AChE inhibitors differ in their pharmacological profiles although their efficacy for treatment of dementia is comparable [154]. Galantamine is a positive allosteric modulator of nAChRs, resulting in increased synaptic DA and glutamate levels [155, 156]. Donepezil and rivastigmine are more potent AChE inhibitors than galantamine [157, 158]. Rivastigmine may also modulate a glutamatergic transporter [159]. In contrast to donepezil and rivastigmine, galantamine also increases the activity of nAChR via allosteric effects [155]. As summarized in Table 3, in preclinical studies AChE inhibitors attenuated opioid reinforcement, enhanced the analgesic effects of opioids, reduced tolerance to opioid analgesia and attenuate opioid withdrawal. In human studies, galantamine showed promising results as a treatment for alcohol or tobacco use disorder [160, 161]. In a recent randomized clinical trial, galantamine, compared to placebo, reduced cocaine use among cocaine and opioid addictive individuals that were stabilized on methadone [162]. AChE inhibitors have not been examined for the treatment of OUD in humans. Clinical trials testing the potential efficacy of AChE inhibitors on OUD are warranted.

8.2. Medications Targeting nAChR

Although there is some evidence that medications targeting nAChR have efficacy for treating substance use disorders, the evidence specifically for OUD is mixed. In preclinical studies, the nAChR antagonists lobeline and mecamylamine did not show consistent efficacy across outcomes [126, 136, 138, 142]. Varenicline, a partial agonist at the α4β2 nAChR, is marketed for smoking cessation and has also shown promise for alcohol use disorder [163165]. Varenicline also reduced rates of smoking in opioid addicted individuals maintained on methadone, however there was no effect on opioid use [166]. Functional genetic variation encoded in nAChRs, such as the well-characterized and common variants in the CHRNA5–A3–B4 gene cluster, might affect the response to some medications. For smoking cessation, reports on differential responses to varenicline based on CHRNA5–A3–B4 gene cluster variants have been mixed [167169]. As noted above, other nicotinic receptor genes encode variants with strong trait associations (e.g. CHRNA4) that might affect nACHR function and response to treatment. As research in this area progresses, it will be important to consider these known genetic effects and how they might shape the response to medication that targets nAChRs.

8.3. Medications targeting mAChR

As outlined in Table 1, in preclinical studies, both mAChR agonists (e.g., AREC) and the mAChR antagonist scopolamine showed promising effects. AREC reduced both opioid self-administration and reinstatement of opioid self-administration and blocked naloxone-induced opioid withdrawal [133]. Scopolamine also prevented development of tolerance to morphine and attenuated naloxone-induced opioid withdrawal [138]. These findings seem to be contradictory as both an antagonist and an agonist of mAChR have similar effects. It is important to note that AREC is a non-selective partial mAChR agonist, which also has significant agonist effects in multiple nAChR subtypes [170]. Similarly, scopolamine influences nAChR and N-Methyl-D-aspartate (NMDA) receptors in addition to its mAChR antagonist effects [171]. Several mAChR agonists, including AREC, carbachol, and cevimeline have been examined in clinical trials, although not for OUD[172]. Development of these compounds have been abandoned due to their adverse effects, like nausea and diarrhea, which are likely mediated by the drug acting at peripheral M2 and M3 mAChRs. Scopolamine, however, is currently used for post-operative nausea and motion sickness. It has rapid anti-depressant effects with some reports showing efficacy for treating major depressive disorder [173]. It is noteworthy that the Liu et al. study reported that compared to methadone treatment, scopolamine reduced depression and anxiety during opioid tapering [145]. Depression is elevated among those seeking treatment for substance use disorders [174] and common in the population, with 40% of an individual’s risk for depression attributed to their genetics [175]. Scopolamine could be most effective for those that are at increased risk for depression during detoxification, either based on their genetics or a pre-existing condition (e.g. major depressive disorder or bipolar). Further studies testing the efficacy of scopolamine for OUD are warranted.

9. Conclusion

New strategies are needed to meet the challenges associated with the current OUD epidemic facing the United States and other countries. MAT, using opioid-based pharmacotherapies (methadone, buprenorphine and naltrexone), are the only current US Food and Drug Administration-approved treatments for OUD. MAT effectiveness is limited by high drop-out rates and the adverse effects associated with long-term treatment with opioid medications, such as cognitive deficits, endocrine disturbances, decreased libido and increased pain sensitivity or hyperalgesia. The cholinergic system shows promise as a treatment target for OUD. The cholinergic and opioid systems are tightly linked, with ACh involved in CNS functions that are relevant to the development and maintenance of OUD. Several cholinergic medications show promise in clinical and preclinical studies and further studies testing the efficacy of cholinergic medication for OUD are warranted.

Key Points.

  • Opioid use disorder is increasing in the population and new medications could help reduce the negative impact to global health.

  • Medications that target the brain cholinergic system show promise in clinical and preclinical studies of opioids.

  • Further studies on cholinergic medications for opioid use disorder are warranted.

Acknowledgments

Funding

This work was supported by the Veterans Administration (VA) Mental Illness Research, Education and Clinical Center (MIRECC), and NIH grants, P50 DA-009241 and K01 DA039299.

Footnotes

Conflicts of Interest

Kevin P. Jensen, Elise E. DeVito, Sarah Yip, Kathleen M. Carroll and Mehmet Sofuoglu declare no conflicts of interest.

References:

  • 1.Barnett ML, Olenski AR, Jena AB. Opioid-Prescribing Patterns of Emergency Physicians and Risk of Long-Term Use. N Engl J Med. 2017. February 16;376(7):663–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Owens PL, Barrett ML, Weiss AJ, Washington RE, Kronick R. Hospital Inpatient Utilization Related to Opioid Overuse Among Adults, 1993–2012: Statistical Brief #177. Healthcare Cost and Utilization Project (HCUP) Statistical Briefs; Rockville (MD); 2006. [PubMed] [Google Scholar]
  • 3.Degenhardt L, Charlson F, Mathers B, Hall WD, Flaxman AD, Johns N, et al. The global epidemiology and burden of opioid dependence: results from the global burden of disease 2010 study. Addiction. 2014. August;109(8):1320–33. [DOI] [PubMed] [Google Scholar]
  • 4.Center for Behavioral Health Statistics and Quality. Key substance use and mental health indicators in the United States: Results from the 2015 National Survey on Drug Use and Health 2016. [Google Scholar]
  • 5.Rudd RA, Seth P, David F, Scholl L. Increases in Drug and Opioid-Involved Overdose Deaths - United States, 2010–2015. MMWR Morb Mortal Wkly Rep. 2016. December 30;65(5051):1445–52. [DOI] [PubMed] [Google Scholar]
  • 6.Hedegaard H, Warner M, Miniño AM. Drug overdose deaths in the United States, 1999–2016 In: Health NCf, Statistics, editors. Hyattsville, MD; 2017. [Google Scholar]
  • 7.Proctor SL, Copeland AL, Kopak AM, Hoffmann NG, Herschman PL, Polukhina N. Predictors of patient retention in methadone maintenance treatment. Psychol Addict Behav. 2015. December;29(4):906–17. [DOI] [PubMed] [Google Scholar]
  • 8.Carroll KM, Weiss RD. The Role of Behavioral Interventions in Buprenorphine Maintenance Treatment: A Review. Am J Psychiatry. 2017. August 01;174(8):738–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Mattick RP, Breen C, Kimber J, Davoli M. Methadone maintenance therapy versus no opioid replacement therapy for opioid dependence. Cochrane Database Syst Rev. 2009;3(3). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Xu M, Chung S, Zhang S, Zhong P, Ma C, Chang W-C, et al. Basal forebrain circuit for sleep-wake control. Nat Neurosci. 2015;18(11):1641. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Mizuno T, Kimura F. Attenuated stress response of hippocampal acetylcholine release and adrenocortical secretion in aged rats. Neurosci Lett. 1997;222(1):49–52. [DOI] [PubMed] [Google Scholar]
  • 12.Himmelheber AM, Sarter M, Bruno JP. Increases in cortical acetylcholine release during sustained attention performance in rats. Cognitive Brain Research. 2000;9(3):313–25. [DOI] [PubMed] [Google Scholar]
  • 13.Redgrave P, HORRELL RI. Potentiation of central reward by localised perfusion of acetylcholine and 5-hydroxytryptamine. Nature. 1976;262(5566):305. [DOI] [PubMed] [Google Scholar]
  • 14.Janowsky DS, Overstreet DH, Nurnberger JI Jr. Is cholinergic sensitivity a genetic marker for the affective disorders? Am J Med Genet. 1994;54(4):335–44. [DOI] [PubMed] [Google Scholar]
  • 15.Borovikova LV, Ivanova S, Zhang M, Yang H, Botchkina GI, Watkins LR, et al. Vagus nerve stimulation attenuates the systemic inflammatory response to endotoxin. Nature. 2000;405(6785):458. [DOI] [PubMed] [Google Scholar]
  • 16.Hser YI, Evans E, Grella C, Ling W, Anglin D. Long-term course of opioid addiction. Harv Rev Psychiatry. 2015. Mar-Apr;23(2):76–89. [DOI] [PubMed] [Google Scholar]
  • 17.Park TW, Saitz R, Ganoczy D, Ilgen MA, Bohnert AS. Benzodiazepine prescribing patterns and deaths from drug overdose among US veterans receiving opioid analgesics: case-cohort study. BMJ. 2015. June 10;350:h2698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Cicero TJ, Ellis MS, Surratt HL, Kurtz SP. The changing face of heroin use in the United States: a retrospective analysis of the past 50 years. JAMA psychiatry. 2014. July 1;71(7):821–6. [DOI] [PubMed] [Google Scholar]
  • 19.Volkow ND, McLellan AT. Opioid Abuse in Chronic Pain--Misconceptions and Mitigation Strategies. N Engl J Med. 2016. March 31;374(13):1253–63. [DOI] [PubMed] [Google Scholar]
  • 20.Cheng Z, Zhou H, Sherva R, Farrer LA, Kranzler HR, Gelernter J. Genome-wide Association Study Identifies a Regulatory Variant of RGMA Associated With Opioid Dependence in European Americans. Biol Psychiatry. 2018. January 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Nelson EC, Agrawal A, Heath AC, Bogdan R, Sherva R, Zhang B, et al. Evidence of CNIH3 involvement in opioid dependence. Mol Psychiatry. 2016. May;21(5):608–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Gelernter J, Kranzler HR, Sherva R, Koesterer R, Almasy L, Zhao H, et al. Genome-wide association study of opioid dependence: multiple associations mapped to calcium and potassium pathways. Biol Psychiatry. 2014. July 1;76(1):66–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Leeman RF, Sun Q, Bogart D, Beseler CL, Sofuoglu M. Comparisons of Cocaine-Only, Opioid-Only, and Users of Both Substances in the National Epidemiologic Survey on Alcohol and Related Conditions (NESARC). Subst Use Misuse. 2016;51(5):553–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Roncero C, Barral C, Rodriguez-Cintas L, Perez-Pazos J, Martinez-Luna N, Casas M, et al. Psychiatric comorbidities in opioid-dependent patients undergoing a replacement therapy programme in Spain: The PROTEUS study. Psychiatry Res. 2016. September 30;243:174–81. [DOI] [PubMed] [Google Scholar]
  • 25.Barry DT, Cutter CJ, Beitel M, Kerns RD, Liong C, Schottenfeld RS. Psychiatric Disorders Among Patients Seeking Treatment for Co-Occurring Chronic Pain and Opioid Use Disorder. J Clin Psychiatry. 2016. October;77(10):1413–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Schwartz RP, Gryczynski J, O’Grady KE, Sharfstein JM, Warren G, Olsen Y, et al. Opioid agonist treatments and heroin overdose deaths in Baltimore, Maryland, 1995–2009. Am J Public Health. 2013. May;103(5):917–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Comer SD, Sullivan MA, Yu E, Rothenberg JL, Kleber HD, Kampman K, et al. Injectable, sustained-release naltrexone for the treatment of opioid dependence: a randomized, placebo-controlled trial. Arch Gen Psychiatry. 2006. February;63(2):210–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Lobmaier P, Kornor H, Kunoe N, Bjorndal A. Sustained-release naltrexone for opioid dependence. Cochrane Database Syst Rev. 2008. April 16(2):Cd006140. [DOI] [PubMed] [Google Scholar]
  • 29.Lee JD, Nunes EV Jr., Novo P, Bachrach K, Bailey GL, Bhatt S, et al. Comparative effectiveness of extended-release naltrexone versus buprenorphine-naloxone for opioid relapse prevention (X:BOT): a multicentre, open-label, randomised controlled trial. Lancet. 2017. November 14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Jarvis BP, Holtyn AF, Subramaniam S, Tompkins DA, Oga EA, Bigelow GE, et al. Extended-release injectable naltrexone for opioid use disorder: a systematic review. Addiction. 2018. February 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Arout CA, Edens E, Petrakis IL, Sofuoglu M. Targeting Opioid-Induced Hyperalgesia in Clinical Treatment: Neurobiological Considerations. CNS drugs. 2015. June;29(6):465–86. [DOI] [PubMed] [Google Scholar]
  • 32.Rass O, Kleykamp BA, Vandrey RG, Bigelow GE, Leoutsakos JM, Stitzer ML, et al. Cognitive performance in methadone maintenance patients: effects of time relative to dosing and maintenance dose level. Exp Clin Psychopharmacol. 2014. June;22(3):248–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Wang GY, Wouldes TA, Kydd R, Jensen M, Russell BR. Neuropsychological performance of methadone-maintained opiate users. J Psychopharmacol. 2014. August;28(8):789–99. [DOI] [PubMed] [Google Scholar]
  • 34.Rhodin A, Stridsberg M, Gordh T. Opioid endocrinopathy: a clinical problem in patients with chronic pain and long-term oral opioid treatment. Clin J Pain. 2010. June;26(5):374–80. [DOI] [PubMed] [Google Scholar]
  • 35.Cox BM, Christie MJ, Devi L, Toll L, Traynor JR. Challenges for opioid receptor nomenclature: IUPHAR Review 9. Br J Pharmacol. 2015;172(2):317–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.KoSKI G, Klee WA. Opiates inhibit adenylate cyclase by stimulating GTP hydrolysis. Proceedings of the National Academy of Sciences. 1981;78(7):4185–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Sharma SK, Nirenberg M, Klee WA. Morphine receptors as regulators of adenylate cyclase activity. Proceedings of the National Academy of Sciences. 1975;72(2):590–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Johnson SW, North RA. Opioids excite dopamine neurons by hyperpolarization of local interneurons. J Neurosci. 1992. February;12(2):483–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Hughes J, Smith T-W, Kosterlitz H-W, Fothergill LA, Morgan B-A, Morris H. Identification of two related pentapeptides from the brain with potent opiate agonist activity. Nature. 1975;258(5536):577. [DOI] [PubMed] [Google Scholar]
  • 40.Li CH, Chung D. Isolation and structure of an untriakontapeptide with opiate activity from camel pituitary glands. Proceedings of the National Academy of Sciences. 1976;73(4):1145–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Day R, Lazure C, Basak A, Boudreault A, Limperis P, Dong W, et al. Prodynorphin processing by proprotein convertase 2 cleavage at single basic residues and enhanced processing in the presence of carboxypeptidase activity. J Biol Chem. 1998;273(2):829–36. [DOI] [PubMed] [Google Scholar]
  • 42.Nothacker H-P, Reinscheid RK, Mansour A, Henningsen RA, Ardati A, Monsma FJ, et al. Primary structure and tissue distribution of the orphanin FQ precursor. Proceedings of the National Academy of Sciences. 1996;93(16):8677–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Valentino RJ, Van Bockstaele E. Endogenous opioids: opposing stress with a cost. F1000Prime Rep. 2015;7:58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Miczek KA, Thompson ML, Shuster L. Opioid-like analgesia in defeated mice. Science. 1982. March 19;215(4539):1520–2. [DOI] [PubMed] [Google Scholar]
  • 45.Reed B, Butelman ER, Kreek MJ. Endogenous opioid system in addiction and addiction-related behaviors. Current opinion in behavioral sciences. 2017;13:196–202. [Google Scholar]
  • 46.Matthes HW, Maldonado R, Simonin F, Valverde O, Slowe S, Kitchen I, et al. Loss of morphine-induced analgesia, reward effect and withdrawal symptoms in mice lacking the mu-opioid-receptor gene. Nature. 1996. October 31;383(6603):819–23. [DOI] [PubMed] [Google Scholar]
  • 47.Sora I, Elmer G, Funada M, Pieper J, Li XF, Hall FS, et al. Mu opiate receptor gene dose effects on different morphine actions: evidence for differential in vivo mu receptor reserve. Neuropsychopharmacology. 2001. July;25(1):41–54. [DOI] [PubMed] [Google Scholar]
  • 48.Becker A, Grecksch G, Kraus J, Loh HH, Schroeder H, Hollt V. Rewarding effects of ethanol and cocaine in mu opioid receptor-deficient mice. Naunyn Schmiedebergs Arch Pharmacol. 2002. April;365(4):296–302. [DOI] [PubMed] [Google Scholar]
  • 49.Berrendero F, Kieffer BL, Maldonado R. Attenuation of nicotine-induced antinociception, rewarding effects, and dependence in mu-opioid receptor knock-out mice. J Neurosci. 2002. December 15;22(24):10935–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Ghozland S, Matthes HW, Simonin F, Filliol D, Kieffer BL, Maldonado R. Motivational effects of cannabinoids are mediated by mu-opioid and kappa-opioid receptors. J Neurosci. 2002. February 1;22(3):1146–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Klitenick MA, DeWitte P, Kalivas PW. Regulation of somatodendritic dopamine release in the ventral tegmental area by opioids and GABA: an in vivo microdialysis study. J Neurosci. 1992. July;12(7):2623–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Yeomans MR, Gray RW. Effects of naltrexone on food intake and changes in subjective appetite during eating: evidence for opioid involvement in the appetizer effect. Physiol Behav. 1997;62(1):15–21. [DOI] [PubMed] [Google Scholar]
  • 53.Daniel M, Martin A, Carter J. Opiate receptor blockade by naltrexone and mood state after acute physical activity. Br J Sports Med. 1992;26(2):111–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Mallik A, Chanda ML, Levitin DJ. Anhedonia to music and mu-opioids: Evidence from the administration of naltrexone. Sci Rep. 2017. February 8;7:41952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Sofuoglu M, Singha A, Kosten TR, McCance-Katz FE, Petrakis I, Oliveto A. Effects of naltrexone and isradipine, alone or in combination, on cocaine responses in humans. Pharmacol Biochem Behav. 2003. July;75(4):801–8. [DOI] [PubMed] [Google Scholar]
  • 56.Knott VJ, Fisher DJ. Naltrexone alteration of the nicotine-induced EEG and mood activation response in tobacco-deprived cigarette smokers. Exp Clin Psychopharmacol. 2007. August;15(4):368–81. [DOI] [PubMed] [Google Scholar]
  • 57.Le Merrer J, Plaza-Zabala A, Del Boca C, Matifas A, Maldonado R, Kieffer BL. Deletion of the δ opioid receptor gene impairs place conditioning but preserves morphine reinforcement. Biol Psychiatry. 2011;69(7):700–3. [DOI] [PubMed] [Google Scholar]
  • 58.Filliol D, Ghozland S, Chluba J, Martin M, Matthes HW, Simonin F, et al. Mice deficient for δ-and μ-opioid receptors exhibit opposing alterations of emotional responses. Nat Genet. 2000;25(2):195. [DOI] [PubMed] [Google Scholar]
  • 59.Spanagel R, Herz A, Shippenberg TS. The effects of opioid peptides on dopamine release in the nucleus accumbens: an in vivo microdialysis study. J Neurochem. 1990;55(5):1734–40. [DOI] [PubMed] [Google Scholar]
  • 60.Clasen MM, Flax SM, Hempel BJ, Cheng K, Rice KC, Riley AL. Antagonism of the kappa opioid receptor attenuates THC-induced place aversions in adult male Sprague-Dawley rats. Pharmacology Biochemistry and Behavior. 2017;163:30–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Walker BM, Zorrilla EP, Koob GF. Systemic κ‐opioid receptor antagonism by nor‐binaltorphimine reduces dependence‐induced excessive alcohol self‐administration in rats. Addict Biol. 2011;16(1):116–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Grella SL, Funk D, Coen K, Li Z, Lê A. Role of the kappa-opioid receptor system in stress-induced reinstatement of nicotine seeking in rats. Behav Brain Res. 2014;265:188–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Mollereau C, Parmentier M, Mailleux P, Butour J-L, Moisand C, Chalon P, et al. ORL1, a novel member of the opioid receptor family. FEBS Lett. 1994;341(1):33–8. [DOI] [PubMed] [Google Scholar]
  • 64.Yu TP, Fein J, Phan T, Evans CJ, Xie CW. Orphanin FQ inhibits synaptic transmission and long‐term potentiation in rat hippocampus. Hippocampus. 1997;7(1):88–94. [DOI] [PubMed] [Google Scholar]
  • 65.King M, Chang A, Pasternak GW. Functional blockade of opioid analgesia by orphanin FQ/nociceptin. Biochem Pharmacol. 1998;55(9):1537–40. [DOI] [PubMed] [Google Scholar]
  • 66.King MA, Rossi GC, Chang AH, Williams L, Pasternak GW. Spinal analgesic activity of orphanin FQ/nociceptin and its fragments. Neurosci Lett. 1997;223(2):113–6. [DOI] [PubMed] [Google Scholar]
  • 67.Williams JT, Ingram SL, Henderson G, Chavkin C, von Zastrow M, Schulz S, et al. Regulation of μ-opioid receptors: desensitization, phosphorylation, internalization, and tolerance. Pharmacol Rev. 2013;65(1):223–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Williams JT, Christie MJ, Manzoni O. Cellular and synaptic adaptations mediating opioid dependence. Physiol Rev. 2001;81(1):299–343. [DOI] [PubMed] [Google Scholar]
  • 69.Koch T, Hollt V. Role of receptor internalization in opioid tolerance and dependence. Pharmacol Ther. 2008. February;117(2):199–206. [DOI] [PubMed] [Google Scholar]
  • 70.Bagley EE, Chieng BC, Christie MJ, Connor M. Opioid tolerance in periaqueductal gray neurons isolated from mice chronically treated with morphine. Br J Pharmacol. 2005. September;146(1):68–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Sharma SK, Klee WA, Nirenberg M. Dual regulation of adenylate cyclase accounts for narcotic dependence and tolerance. Proc Natl Acad Sci U S A. 1975. August;72(8):3092–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Kanof PD, Handelsman L, Aronson MJ, Ness R, Cochrane KJ, Rubinstein KJ. Clinical characteristics of naloxone-precipitated withdrawal in human opioid-dependent subjects. J Pharmacol Exp Ther. 1992;260(1):355–63. [PubMed] [Google Scholar]
  • 73.Tompkins DA, Smith MT, Mintzer MZ, Campbell CM, Strain EC. A double blind, within subject comparison of spontaneous opioid withdrawal from buprenorphine versus morphine. J Pharmacol Exp Ther. 2014;348(2):217–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.MacArthur J, Bowler E, Cerezo M, Gil L, Hall P, Hastings E, et al. The new NHGRI-EBI Catalog of published genome-wide association studies (GWAS Catalog). Nucleic Acids Res. 2017. January 4;45(D1):D896–D901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Jensen KP. A Review of Genome-Wide Association Studies of Stimulant and Opioid Use Disorders. Mol Neuropsychiatry. 2016. May;2(1):37–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Crist RC, Clarke TK, Berrettini WH. Pharmacogenetics of Opioid Use Disorder Treatment. CNS drugs. 2018. April 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Berrettini W A brief review of the genetics and pharmacogenetics of opioid use disorders. Dialogues Clin Neurosci. 2017. September;19(3):229–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Befort K, Filliol D, Decaillot FM, Gaveriaux-Ruff C, Hoehe MR, Kieffer BL. A single nucleotide polymorphic mutation in the human mu-opioid receptor severely impairs receptor signaling. J Biol Chem. 2001. February 2;276(5):3130–7. [DOI] [PubMed] [Google Scholar]
  • 79.Beyer A, Koch T, Schroder H, Schulz S, Hollt V. Effect of the A118G polymorphism on binding affinity, potency and agonist-mediated endocytosis, desensitization, and resensitization of the human mu-opioid receptor. J Neurochem. 2004. May;89(3):553–60. [DOI] [PubMed] [Google Scholar]
  • 80.Zhang Y, Wang D, Johnson AD, Papp AC, Sadee W. Allelic expression imbalance of human mu opioid receptor (OPRM1) caused by variant A118G. J Biol Chem. 2005. September 23;280(38):32618–24. [DOI] [PubMed] [Google Scholar]
  • 81.Hancock DB, Levy JL, Gaddis NC, Glasheen C, Saccone NL, Page GP, et al. Cis-Expression Quantitative Trait Loci Mapping Reveals Replicable Associations with Heroin Addiction in OPRM1. Biol Psychiatry. 2015. October 1;78(7):474–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Smith AH, Jensen KP, Li J, Nunez Y, Farrer LA, Hakonarson H, et al. Genome-wide association study of therapeutic opioid dosing identifies a novel locus upstream of OPRM1. Mol Psychiatry. 2017. March;22(3):346–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Crist RC, Doyle GA, Nelson EC, Degenhardt L, Martin NG, Montgomery GW, et al. A polymorphism in the OPRM1 3’-untranslated region is associated with methadone efficacy in treating opioid dependence. Pharmacogenomics J. 2018. January;18(1):173–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Crist RC, Clarke TK, Ang A, Ambrose-Lanci LM, Lohoff FW, Saxon AJ, et al. An intronic variant in OPRD1 predicts treatment outcome for opioid dependence in African-Americans. Neuropsychopharmacology. 2013. September;38(10):2003–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Nishizawa D, Fukuda K, Kasai S, Hasegawa J, Aoki Y, Nishi A, et al. Genome-wide association study identifies a potent locus associated with human opioid sensitivity. Mol Psychiatry. 2014. January;19(1):55–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Mark GP, Rada P, Pothos E, Hoebel BG. Effects of feeding and drinking on acetylcholine release in the nucleus accumbens, striatum, and hippocampus of freely behaving rats. J Neurochem. 1992;58(6):2269–74. [DOI] [PubMed] [Google Scholar]
  • 87.Wilson MC, Schuster CR. Cholinergic influence on intravenous cocaine self-administration by rhesus monkeys. Pharmacology Biochemistry and Behavior. 1973;1(6):643–9. [DOI] [PubMed] [Google Scholar]
  • 88.Zhou W, Liu H, Zhang F, Tang S, Zhu H, Lai M, et al. Role of acetylcholine transmission in nucleus accumbens and ventral tegmental area in heroin-seeking induced by conditioned cues. Neuroscience. 2007;144(4):1209–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Rada PV, Mark GP, Taylor KM, Hoebel BG. Morphine and naloxone, ip or locally, affect extracellular acetylcholine in the accumbens and prefrontal cortex. Pharmacology Biochemistry and Behavior. 1996;53(4):809–16. [DOI] [PubMed] [Google Scholar]
  • 90.Rada P, Jensen K, Hoebel BG. Effects of nicotine and mecamylamine-induced withdrawal on extracellular dopamine and acetylcholine in the rat nucleus accumbens. Psychopharmacology (Berl). 2001;157(1):105–10. [DOI] [PubMed] [Google Scholar]
  • 91.Rada P, Johnson D, Lewis M, Hoebel B. In alcohol-treated rats, naloxone decreases extracellular dopamine and increases acetylcholine in the nucleus accumbens: evidence of opioid withdrawal. Pharmacology Biochemistry and Behavior. 2004;79(4):599–605. [DOI] [PubMed] [Google Scholar]
  • 92.Potter L Synthesis, storage and release of [14C] acetylcholine in isolated rat diaphragm muscles. The Journal of physiology. 1970;206(1):145–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Giacobini E Cholinesterase inhibitors: new roles and therapeutic alternatives. Pharmacol Res. 2004. October;50(4):433–40. [DOI] [PubMed] [Google Scholar]
  • 94.Selden NR, Gitelman DR, Salamon-Murayama N, Parrish TB, Mesulam M-M. Trajectories of cholinergic pathways within the cerebral hemispheres of the human brain. Brain: a journal of neurology. 1998;121(12):2249–57. [DOI] [PubMed] [Google Scholar]
  • 95.Kimura M, Rajkowski J, Evarts E. Tonically discharging putamen neurons exhibit set-dependent responses. Proceedings of the National Academy of Sciences. 1984;81(15):4998–5001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Saper CB, Chou TC, Scammell TE. The sleep switch: hypothalamic control of sleep and wakefulness. Trends Neurosci. 2001. December;24(12):726–31. [DOI] [PubMed] [Google Scholar]
  • 97.Bigl V, Woolf NJ, Butcher LL. Cholinergic projections from the basal forebrain to frontal, parietal, temporal, occipital, and cingulate cortices: a combined fluorescent tracer and acetylcholinesterase analysis. Brain Res Bull. 1982;8(6):727–49. [DOI] [PubMed] [Google Scholar]
  • 98.Mesulam MM, Mufson EJ, Levey AI, Wainer BH. Cholinergic innervation of cortex by the basal forebrain: cytochemistry and cortical connections of the septal area, diagonal band nuclei, nucleus basalis (substantia innominata), and hypothalamus in the rhesus monkey. J Comp Neurol. 1983;214(2):170–97. [DOI] [PubMed] [Google Scholar]
  • 99.Levey A, Kitt C, Simonds W, Price D, Brann M. Identification and localization of muscarinic acetylcholine receptor proteins in brain with subtype-specific antibodies. J Neurosci. 1991;11(10):3218–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Caulfield MP, Birdsall NJ. International Union of Pharmacology. XVII. Classification of muscarinic acetylcholine receptors. Pharmacol Rev. 1998. June;50(2):279–90. [PubMed] [Google Scholar]
  • 101.Oki T, Takagi Y, Inagaki S, Taketo MM, Manabe T, Matsui M, et al. Quantitative analysis of binding parameters of [3H]N-methylscopolamine in central nervous system of muscarinic acetylcholine receptor knockout mice. Brain Res Mol Brain Res. 2005. January 5;133(1):6–11. [DOI] [PubMed] [Google Scholar]
  • 102.Wei J, Walton EA, Milici A, Buccafusco JJ. m1–m5 muscarinic receptor distribution in rat CNS by RT‐PCR and HPLC. J Neurochem. 1994;63(3):815–21. [DOI] [PubMed] [Google Scholar]
  • 103.Forster GL, Yeomans JS, Takeuchi J, Blaha CD. M5 muscarinic receptors are required for prolonged accumbal dopamine release after electrical stimulation of the pons in mice. J Neurosci. 2002;22(1):RC190-RC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Felder CC, Bymaster FP, Ward J, DeLapp N. Therapeutic opportunities for muscarinic receptors in the central nervous system. J Med Chem. 2000. November 16;43(23):4333–53. [DOI] [PubMed] [Google Scholar]
  • 105.Tzavara ET, Bymaster FP, Davis RJ, Wade MR, Perry KW, Wess J, et al. M4 muscarinic receptors regulate the dynamics of cholinergic and dopaminergic neurotransmission: relevance to the pathophysiology and treatment of related CNS pathologies. FASEB J. 2004. September;18(12):1410–2. [DOI] [PubMed] [Google Scholar]
  • 106.Hasselmo ME, Schnell E. Laminar selectivity of the cholinergic suppression of synaptic transmission in rat hippocampal region CA1: computational modeling and brain slice physiology. J Neurosci. 1994;14(6):3898–914. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Herrera‐Morales W, Mar I, Serrano B, Bermúdez‐Rattoni F. Activation of hippocampal postsynaptic muscarinic receptors is involved in long‐term spatial memory formation. Eur J Neurosci. 2007;25(5):1581–8. [DOI] [PubMed] [Google Scholar]
  • 108.Leaderbrand K, Chen HJ, Corcoran KA, Guedea AL, Jovasevic V, Wess J, et al. Muscarinic acetylcholine receptors act in synergy to facilitate learning and memory. Learn Mem. 2016;23(11):631–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Champtiaux N, Gotti C, Cordero-Erausquin M, David DJ, Przybylski C, Léna C, et al. Subunit composition of functional nicotinic receptors in dopaminergic neurons investigated with knock-out mice. J Neurosci. 2003;23(21):7820–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Gotti C, Moretti M, Zanardi A, Gaimarri A, Champtiaux N, Changeux JP, et al. Heterogeneity and selective targeting of nAChR subtypes expressed on retinal afferents of the superior colliculus and lateral geniculate nucleus. Identification of a new native nAChR subtype α3β2 (α5 or β3) enriched in retinocollicular afferents. Mol Pharmacol. 2005. [DOI] [PubMed] [Google Scholar]
  • 111.Zoli M, Moretti M, Zanardi A, McIntosh JM, Clementi F, Gotti C. Identification of the nicotinic receptor subtypes expressed on dopaminergic terminals in the rat striatum. J Neurosci. 2002;22(20):8785–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Wonnacott S, Kaiser S, Mogg A, Soliakov L, Jones IW. Presynaptic nicotinic receptors modulating dopamine release in the rat striatum. Eur J Pharmacol. 2000;393(1–3):51–8. [DOI] [PubMed] [Google Scholar]
  • 113.Grilli M, Zappettini S, Raiteri L, Marchi M. Nicotinic and muscarinic cholinergic receptors coexist on GABAergic nerve endings in the mouse striatum and interact in modulating GABA release. Neuropharmacology. 2009;56(3):610–4. [DOI] [PubMed] [Google Scholar]
  • 114.Rodrigues RJ, Almeida T, de Mendonça A, Cunha RA. Interaction between P2X and nicotinic acetylcholine receptors in glutamate nerve terminals of the rat hippocampus. J Mol Neurosci. 2006;30(1–2):173–6. [DOI] [PubMed] [Google Scholar]
  • 115.Wilkie G, Hutson P, Sullivan J, Wonnacott S. Pharmacological characterization of a nicotinic autoreceptor in rat hippocampal synaptosomes. Neurochem Res. 1996;21(9):1141–8. [DOI] [PubMed] [Google Scholar]
  • 116.Picciotto MR, Corrigall WA. Neuronal systems underlying behaviors related to nicotine addiction: neural circuits and molecular genetics. J Neurosci. 2002;22(9):3338–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Picciotto MR, Zoli M, Lena C, Bessis A, Lallemand Y, Le Novere N, et al. Abnormal avoidance learning in mice lacking functional high-affinity nicotine receptor in the brain. Nature. 1995. March 2;374(6517):65–7. [DOI] [PubMed] [Google Scholar]
  • 118.Epping-Jordan MP, Watkins SS, Koob GF, Markou A. Dramatic decreases in brain reward function during nicotine withdrawal. Nature. 1998. May 7;393(6680):76–9. [DOI] [PubMed] [Google Scholar]
  • 119.Tobacco Genetics Consortium. Genome-wide meta-analyses identify multiple loci associated with smoking behavior. Nat Genet. 2010. May;42(5):441–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Ware JJ, Chen X, Vink J, Loukola A, Minica C, Pool R, et al. Genome-Wide Meta-Analysis of Cotinine Levels in Cigarette Smokers Identifies Locus at 4q13.2. Sci Rep. 2016;6:20092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Green JP, Glick SD, Crane AM, Szilagyi PI. Acute effects of morphine on regional brain levels of acetylcholine in mice and rats. Eur J Pharmacol. 1976;39(1):91–9. [DOI] [PubMed] [Google Scholar]
  • 122.Gage HD, Gage JC, Tobin JR, Chiari A, Tong C, Xu Z-M, et al. Morphine-induced spinal cholinergic activation: in vivo imaging with positron emission tomography. Pain. 2001;91(1–2):139–45. [DOI] [PubMed] [Google Scholar]
  • 123.Yang J, Zhao Y, Pan Y, Lu G, Lu L, Wang D, et al. Acetylcholine participates in pain modulation by influencing endogenous opiate peptides in rat spinal cord. World Journal of Neuroscience. 2012;2(01):15. [Google Scholar]
  • 124.Cavallini S, Marino S, Beani L, Bianchi C, Siniscalchi A. Nociceptin inhibition of acetylcholine efflux from different brain areas. Neuroreport. 2003;14(17):2167–70. [DOI] [PubMed] [Google Scholar]
  • 125.Uezu K, Sano A, Sei H, Toida K, Houtani T, Sugimoto T, et al. Enhanced hippocampal acetylcholine release in nociceptin-receptor knockout mice. Brain Res. 2005;1050(1–2):118–23. [DOI] [PubMed] [Google Scholar]
  • 126.Bajic D, Soiza-Reilly M, Spalding AL, Berde CB, Commons KG. Endogenous cholinergic neurotransmission contributes to behavioral sensitization to morphine. PLoS One. 2015;10(2):e0117601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Hudson TJ, Painter JT, Martin BC, Austen MA, Williams JS, Fortney JC, et al. Pharmacoepidemiologic analyses of opioid use among OEF/OIF/OND veterans. Pain. 2017;158(6):1039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Young-Wolff KC, Klebaner D, Weisner C, Von Korff M, Campbell CI. Smoking status and opioid-related problems and concerns among men and women on chronic opioid therapy. The Clinical journal of pain. 2017;33(8):730–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Schmidt B, Tambeli C, Gear R, Levine J. Nicotine withdrawal hyperalgesia and opioid-mediated analgesia depend on nicotine receptors in nucleus accumbens. Neuroscience. 2001;106(1):129–36. [DOI] [PubMed] [Google Scholar]
  • 130.Buccafusco JJ. Neuropharmacologic and behavioral actions of clonidine: interactions with central neurotransmitters. Int Rev Neurobiol. 1992;33:55–107. [DOI] [PubMed] [Google Scholar]
  • 131.Redmond DE Jr., Krystal JH. Multiple mechanisms of withdrawal from opioid drugs. Annu Rev Neurosci. 1984;7:443–78. [DOI] [PubMed] [Google Scholar]
  • 132.Li X, Li JX, France CP. Interactions between morphine, scopolamine and nicotine: schedule-controlled responding in rats. Pharmacol Biochem Behav. 2010. July;96(1):91–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Buccafusco JJ, Bain JN. A 24-h access I.V. self-administration schedule of morphine reinforcement and the estimation of recidivism: Pharmacological modification by arecoline. Neuroscience. 2007. November 9;149(3):487–98. [DOI] [PubMed] [Google Scholar]
  • 134.Zhou H, Ge X, Wang LZ, Ma L, Pei G. Attenuation of morphine tolerance and dependence in scopolamine-treated rats. Neuroreport. 1999. July 13;10(10):2007–10. [DOI] [PubMed] [Google Scholar]
  • 135.Hikida T, Kitabatake Y, Pastan I, Nakanishi S. Acetylcholine enhancement in the nucleus accumbens prevents addictive behaviors of cocaine and morphine. Proc Natl Acad Sci U S A. 2003. May 13;100(10):6169–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Gawel K, Labuz K, Jenda M, Silberring J, Kotlinska JH. Influence of cholinesterase inhibitors, donepezil and rivastigmine on the acquisition, expression, and reinstatement of morphine-induced conditioned place preference in rats. Behav Brain Res. 2014. July 15;268:169–76. [DOI] [PubMed] [Google Scholar]
  • 137.Sun J, Tian L, Cui R, Li X. Huperzine A inhibits immediate addictive behavior but not behavioral sensitization following repeated morphine administration in rats. Exp Ther Med. 2017. April;13(4):1584–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Gawel K, Gibula-Bruzda E, Dziedzic M, Jenda-Wojtanowska M, Marszalek-Grabska M, Silberring J, et al. Cholinergic activation affects the acute and chronic antinociceptive effects of morphine. Physiol Behav. 2017. February 1;169:22–32. [DOI] [PubMed] [Google Scholar]
  • 139.Sharifipour M, Izadpanah E, Nikkhoo B, Zare S, Abdolmaleki A, Hassanzadeh K, et al. A new pharmacological role for donepezil: attenuation of morphine-induced tolerance and apoptosis in rat central nervous system. J Biomed Sci. 2014. 01/2309/17/received 01/20/accepted; 21(1):6-. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Buccafusco JJ, Zhang LC, Shuster LC, Jonnala RR, Gattu M. Prevention of precipitated withdrawal symptoms by activating central cholinergic systems during a dependence-producing schedule of morphine in rats. Brain Res. 2000. January 3;852(1):76–83. [DOI] [PubMed] [Google Scholar]
  • 141.Xiang X-H, Wang H-L, Wu W-R, Guo Y, Cao D-Y, Wang H-S, et al. Ethological analysis of scopolamine treatment or pretreatment in morphine dependent rats. Physiol Behav. 2006;88:183–90. [DOI] [PubMed] [Google Scholar]
  • 142.Neugebauer NM, Einstein EB, Lopez MB, McClure-Begley TD, Mineur YS, Picciotto MR. Morphine dependence and withdrawal induced changes in cholinergic signaling. Pharmacol Biochem Behav. 2013. August;109:77–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Slatkin NE, Rhiner M, Bolton TM. Donepezil in the treatment of opioid-induced sedation: report of six cases. J Pain Symptom Manage. 2001. May;21(5):425–38. [DOI] [PubMed] [Google Scholar]
  • 144.Hooten WM, Warner DO. Varenicline for opioid withdrawal in patients with chronic pain: a randomized, single-blinded, placebo controlled pilot trial. Addict Behav. 2015. March;42:69–72. [DOI] [PubMed] [Google Scholar]
  • 145.Liu S, Li L, Shen W, Shen X, Yang G, Zhou W. Scopolamine detoxification technique for heroin dependence: a randomized trial. CNS drugs. 2013. December;27(12):1093–102. [DOI] [PubMed] [Google Scholar]
  • 146.Dean AJ, Saunders JB, Jones RT, Young RM, Connor JP, Lawford BR. Does naltrexone treatment lead to depression? Findings from a randomized controlled trial in subjects with opioid dependence. J Psychiatry Neurosci. 2006. January;31(1):38–45. [PMC free article] [PubMed] [Google Scholar]
  • 147.Miotto K, McCann M, Basch J, Rawson R, Ling W. Naltrexone and dysphoria: fact or myth? The American journal on addictions. 2002;11(2):151–60. [DOI] [PubMed] [Google Scholar]
  • 148.Kavirajan H, Schneider LS. Efficacy and adverse effects of cholinesterase inhibitors and memantine in vascular dementia: a meta-analysis of randomised controlled trials. Lancet Neurol. 2007. September;6(9):782–92. [DOI] [PubMed] [Google Scholar]
  • 149.Product Information: TRANSDERM SCOP(R) transdermal patch, scopolamine transdermal patch. Baxter Healthcare Corporation (per DailyMed), Deerfield, IL; 2010. [Google Scholar]
  • 150.Evans WE, Relling MV. Pharmacogenomics: translating functional genomics into rational therapeutics. Science. 1999;286(5439):487–91. [DOI] [PubMed] [Google Scholar]
  • 151.Torkamani A, Andersen KG, Steinhubl SR, Topol EJ. High-Definition Medicine. Cell. 2017. August 24;170(5):828–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Birks J Cholinesterase inhibitors for Alzheimer’s disease. Cochrane Database Syst Rev. 2006(1):CD005593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Farlow M A clinical overview of cholinesterase inhibitors in Alzheimer’s disease. Int Psychogeriatr. 2002;14 Suppl 1:93–126. [DOI] [PubMed] [Google Scholar]
  • 154.Trinh NH, Hoblyn J, Mohanty S, Yaffe K. Efficacy of cholinesterase inhibitors in the treatment of neuropsychiatric symptoms and functional impairment in Alzheimer disease: a meta-analysis. JAMA. 2003. January 8;289(2):210–6. [DOI] [PubMed] [Google Scholar]
  • 155.Schilstrom B, Ivanov VB, Wiker C, Svensson TH. Galantamine enhances dopaminergic neurotransmission in vivo via allosteric potentiation of nicotinic acetylcholine receptors. Neuropsychopharmacology. 2007. January;32(1):43–53. [DOI] [PubMed] [Google Scholar]
  • 156.Penner J, Rupsingh R, Smith M, Wells JL, Borrie MJ, Bartha R. Increased glutamate in the hippocampus after galantamine treatment for Alzheimer disease. Prog Neuropsychopharmacol Biol Psychiatry. 2010;34(1):104–10. [DOI] [PubMed] [Google Scholar]
  • 157.Zhao Q, Tang XC. Effects of huperzine A on acetylcholinesterase isoforms in vitro: comparison with tacrine, donepezil, rivastigmine and physostigmine. Eur J Pharmacol. 2002;455(2–3):101–7. [DOI] [PubMed] [Google Scholar]
  • 158.Geerts H, Guillaumat P-O, Grantham C, Bode W, Anciaux K, Sachak S. Brain levels and acetylcholinesterase inhibition with galantamine and donepezil in rats, mice, and rabbits. Brain Res. 2005;1033(2):186–93. [DOI] [PubMed] [Google Scholar]
  • 159.Andin J, Enz A, Gentsch C, Marcusson J. Rivastigmine as a modulator of the neuronal glutamate transporter rEAAC1 mRNA expression. Dement Geriatr Cogn Disord. 2005;19(1):18–23. [DOI] [PubMed] [Google Scholar]
  • 160.Diehl A, Nakovics H, Croissant B, Smolka MN, Batra A, Mann K. Galantamine reduces smoking in alcohol-dependent patients: a randomized, placebo-controlled trial. Int J Clin Pharmacol Ther. 2006. December;44(12):614–22. [DOI] [PubMed] [Google Scholar]
  • 161.Mann K, Ackermann K, Diehl A, Ebert D, Mundle G, Nakovics H, et al. Galantamine: a cholinergic patch in the treatment of alcoholism: a randomized, placebo-controlled trial. Psychopharmacology (Berl). 2006;184(1):115–21. [DOI] [PubMed] [Google Scholar]
  • 162.Carroll KM, Nich C, DeVito EE, Shi JM, Sofuoglu M. Galantamine and Computerized Cognitive Behavioral Therapy for Cocaine Dependence: A Randomized Clinical Trial. The Journal of clinical psychiatry. 2017;79(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.de Bejczy A, Lof E, Walther L, Guterstam J, Hammarberg A, Asanovska G, et al. Varenicline for treatment of alcohol dependence: a randomized, placebo-controlled trial. Alcohol Clin Exp Res. 2015. November;39(11):2189–99. [DOI] [PubMed] [Google Scholar]
  • 164.McKee SA, Harrison EL, O’Malley SS, Krishnan-Sarin S, Shi J, Tetrault JM, et al. Varenicline reduces alcohol self-administration in heavy-drinking smokers. Biol Psychiatry. 2009. July 15;66(2):185–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Mitchell JM, Teague CH, Kayser AS, Bartlett SE, Fields HL. Varenicline decreases alcohol consumption in heavy-drinking smokers. Psychopharmacology (Berl). 2012. October;223(3):299–306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Poling J, Rounsaville B, Gonsai K, Severino K, Sofuoglu M. The safety and efficacy of varenicline in cocaine using smokers maintained on methadone: a pilot study. Am J Addict. 2010. Sep-Oct;19(5):401–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Chen LS, Baker TB, Piper ME, Breslau N, Cannon DS, Doheny KF, et al. Interplay of genetic risk factors (CHRNA5-CHRNA3-CHRNB4) and cessation treatments in smoking cessation success. Am J Psychiatry. 2012. July 1;169(7):735–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.King DP, Paciga S, Pickering E, Benowitz NL, Bierut LJ, Conti DV, et al. Smoking cessation pharmacogenetics: analysis of varenicline and bupropion in placebo-controlled clinical trials. Neuropsychopharmacology. 2012. February;37(3):641–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Tyndale RF, Zhu AZ, George TP, Cinciripini P, Hawk LW Jr., Schnoll RA, et al. Lack of Associations of CHRNA5–A3–B4 Genetic Variants with Smoking Cessation Treatment Outcomes in Caucasian Smokers despite Associations with Baseline Smoking. PLoS One. 2015;10(5):e0128109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Papke RL, Horenstein NA, Stokes C. Nicotinic activity of arecoline, the psychoactive element of” Betel Nuts”, suggests a basis for habitual use and anti-inflammatory activity. PLoS One. 2015;10(10):e0140907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Falsafi SK, Deli A, Höger H, Pollak A, Lubec G. Scopolamine administration modulates muscarinic, nicotinic and NMDA receptor systems. PLoS One. 2012;7(2):e32082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Langmead CJ, Watson J, Reavill C. Muscarinic acetylcholine receptors as CNS drug targets. Pharmacol Ther. 2008. February;117(2):232–43. [DOI] [PubMed] [Google Scholar]
  • 173.Drevets WC, Furey ML. Replication of scopolamine’s antidepressant efficacy in major depressive disorder: a randomized, placebo-controlled clinical trial. Biol Psychiatry. 2010. March 1;67(5):432–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Chan YF, Dennis ML, Funk RR. Prevalence and comorbidity of major internalizing and externalizing problems among adolescents and adults presenting to substance abuse treatment. J Subst Abuse Treat. 2008. January;34(1):14–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Kendler KS, Gardner CO, Neale MC, Prescott CA. Genetic risk factors for major depression in men and women: similar or different heritabilities and same or partly distinct genes? Psychol Med. 2001. May;31(4):605–16. [DOI] [PubMed] [Google Scholar]
  • 176.Mukae T, Uchida H, Ueda H. Donepezil reverses intermittent stress-induced generalized chronic pain syndrome in mice. J Pharmacol Exp Ther. 2015. June;353(3):471–9. [DOI] [PubMed] [Google Scholar]

RESOURCES