Abstract
Concomitant use of tobacco and opioids represents a growing public health concern. In fact, the mortality rate due to smoking-related illness approaches 50% among SUD patients. Cumulative evidence demonstrates that the vulnerability to drugs of abuse is influenced by behavioral, environmental, and genetic factors. This review explores the contribution of genetics and neural mechanisms influencing nicotine and opioid reward, respiration, and antinociception, emphasizing the interaction of cholinergic and opioid receptor systems. Despite the substantial evidence demonstrating nicotine-opioid interactions within the brain and on behavior, the currently available pharmacotherapies targeting these systems have shown limited efficacy for smoking cessation on opioid-maintained smokers. Thus, further studies designed to identify novel targets modulating both nicotinic and opioid receptor systems may lead to more efficacious approaches for co-morbid nicotine dependence and opioid use disorder.
Keywords: Substance use disorder, Opioids, Nicotine, Polydrug abuse, Rodent models, Clinical trials, Genetics, Neurobiology of addiction
1. Introduction
Nicotine dependence (ND) and opioid use disorder (OUD) are primary causes of preventable disease and premature death worldwide. Despite the increased public health campaigns and availability of smoking cessation and OUD aids, the successful smoking cessation rate remains lower than 8% (Creamer et al., 2019), while opioid-related overdose deaths have been escalating (O’Donnell et al., 2020). An overwhelming majority of individuals with opioid use disorder (OUD) report nicotine co-use, with over 80 % of OUD patients reporting cigarette smoking (Kalman et al., 2005). The rate of nicotine-opioid co-use appears even higher in untreated opioid users, with estimated co-use among 92 % of heroin users (Haas et al., 2008) and 91 % of syringe exchange participants not in treatment (Clarke et al., 2001). Furthermore, between 2002 and 2017, the prevalence of OUD has escalated among smokers and former smokers, while the prevalence of OUD has decreased among non-smokers (Parker and Weinberger, 2020). The high prevalence of nicotine-opioid co-use is particularly pressing since their interactions are largely unexplored and co-use may present an even greater risk for negative health effects.
Furthermore, smoking rates are higher in vulnerable drug-using populations, contributing to a 50 % mortality rate due to smoking-related illness among substance use disorder (SUD) patients (Hurt et al., 1996; Hser et al., 1994). For example, the smoking rates among hospitalized smokers with SUD are at least three times greater than hospitalized patients without SUD, particularly in opioid-dependent patients (Kathuria et al., 2019). Conversely, smoking cessation medications are less efficacious among individuals with OUD (Miller and Sigmon, 2015), possibly due to the higher severity of ND in this population (Parker et al., 2018). Moreover, smokers with OUD report low motivation to quit (Kathuria et al., 2019), possibly because they perceive smoking as less harmful and have limited access to treatment programs (Richter et al., 2004). Additionally, smokers with OUD report concerns about opioid relapse, since many use smoking to cope with negative affect and ameliorate other adverse events related to opioid withdrawal, which poses a risk to patient’s adherence and compliance to smoking cessation programs. Chronic pain is also highly prevalent in both smokers and individuals with OUD compared to the general population (Plesner et al., 2016), likely representing a co-morbid driver for dependence. Individuals with chronic pain who are smokers under opioid therapy use more opioids than never smokers and are less sensitive to the adverse effects of opioids. Males with chronic pain consume an increased daily dosage of opioids, and therefore, are particularly vulnerable to misuse prescription opioids and potentially overdose leading to respiratory failure (Young-Wolff et al., 2017). However, while there are epidemiological data showing distinct interactions between ND and OUD, the mechanisms circumscribing this co-dependency are not well understood.
In this review, we will discuss current knowledge and recent developments linking rodent and human observations in nicotine-opioid interaction research, examining evidence of interdigitating roles of nicotine and opioids on reward, antinociception, and respiration. Reward was selected because this process underlies the abuse liability of both nicotine and opioids. Antinociception was selected because (1) nicotinic cholinergic mechanisms are known to be involved and (2) pain relief represents the main clinical use for opioids. Finally, respiration was selected because tobacco inhalation alters respiration rate and respiratory failure represents the main cause of death due to opioid overdose. Through this lens, we will then examine the neural and genetic mechanisms contributing to co-dependency and discuss their salience to current clinical trials data for integration of these mechanisms in evidence-based approaches for individuals suffering from co-morbid ND and OUD.
2. Nicotine dependence and opioid use disorder: reward
2.1. Anatomy of reward pertaining to nicotine and opioid co-use
The mesocorticolimbic or reward pathway is the key neural substrate for reward, including those behaviors underlying drug addiction. Nicotine and opioids are known to enhance the effect of dopaminergic function in the mesocorticolimbic pathway that runs from the midbrain ventral tegmental area (VTA) to medium spiny neurons in nucleus accumbens (NAc) and the prefrontal cortex (PFC). Both nicotine and opioids are known to evoke dopamine release in this pathway (Nisell et al., 1994; Tanda and Di Chiara, 1998), a response known to be a principal neural framework shaping reward-related behaviors. With nicotine, burst firing of dopamine VTA neurons is increased directly via nicotinic acetylcholine receptors (nAChRs) located on cell bodies, as well as indirectly via GABAergic interneurons within VTA (Pistillo et al., 2015; Mansvelder and McGehee, 2002; Faure et al., 2014). Dopamine release is enhanced further via nAChRs in the terminal field (Exley et al., 2008; Lee et al., 2020). With opioids, there is a dopamine-dependent mechanism involving disinhibition of dopamine neurons via GABAergic interneurons in VTA, as well as a dopamine-independent mechanism involving modulation of medium spiny neurons in NAc (Nestler et al., 2020; Hjelmstad and Fields, 2003).
Repeated nicotine or opioid exposure is thought to change the sensitivity of the dopamine reward pathway via extensive glutamatergic input from various structures, including PFC, hippocampus, paraventricular thalamus and amygdala (Kruyer et al., 2020; Scofield et al., 2016). These areas are anatomically and functionally interconnected to exert motivation, reward, cognitive and emotional processing (Goto and Grace, 2008). In addition to reward processing and dependence, additional brain systems mediate drug-induced aversive states that counterbalance activity of the mesolimbic pathway. For example, kappa opioid agonists produce aversion by directly blunting the reward pathway (Knoll and Carlezon, 2010; Robles et al., 2014), whereas nicotine produces aversion by indirectly blunting the reward pathway by activation of the lateral habenula (Fowler and Kenny, 2014; Zuo et al., 2016). Thus, the complex interplay of these various systems broadly demonstrates both the circuitry-level regulation of reward, as well as a dissociations of the discrete symptoms of withdrawal and aversion that ultimately control nicotine and opioid drug-taking.
2.2. Nicotinic contributions to opioid reward
Underlying the co-morbidity of tobacco use and opioid use disorders, preclinical evidence shows a direct interaction between opioid and nicotinic receptor activation in the control of addiction-relevant behaviors measured by either conditioned place preference (CPP) or self-administration (SA). As summarized in Table 1, a number of preclinical studies have examined the role of nAChRs in modulating opioid reward. Most relevant for the co-morbidity of ND and OUD, nicotine pretreatment can have profound effects on opioid reward. When nicotine is administered in the drinking water for 7 weeks prior to conditioning, it sensitizes mice to morphine CPP (Vihavainen et al., 2008a). In contrast, when administered during conditioning, systemic or intra-insular cortex nicotine shifts the dose-effect curve for morphine CPP to the right (Loney et al., 2021), indicative of decreased sensitivity to opioid reward. Similarly, when administered on the test day, systemic and intracerebroventricular nicotine decreases the expression of morphine CPP (Shams et al., 2006). As a compensation for reduced reward sensitivity, acute nicotine pretreatment increases SA of morphine and remifentanil (Loney et al., 2021). Conversely, the use-dependent nicotinic antagonist 2,2,6,6-tetramethyl-4-piperidinyl [BTMPS] decreases morphine SA and seeking during abstinence (Hall et al., 2011). Interestingly, in contrast to nicotine-induced increases in opioid SA, activation of muscarinic cholinergic receptors decreases heroin SA (Zhou et al., 2006).
Table 1.
References | Species | Sex | Nicotinic Manipulation | Opioid Reward Test | Result on Opioid Reward |
---|---|---|---|---|---|
(Loney et al., 2021) | Long-Evans rats | male | 1. nicotine 0.4 mg/kg, s.c. | 1. SA FR-3 remifentanil 3.2 μg/kg, i.v. morphine 0.3 mg/kg, i.v. or CPP morphine 5, 10, or 20 mg/kg, s.c. |
1. ↑ SA remifentanil and morphine ↓ CPP morphine 5 and 10 mg/kg ↑ CPP morphine 20 mg/kg |
2. nicotine 4 μg intra-IC | 2. CPP morphine 5, 10 or 20 mg/kg, s.c. | 2. ↓ CPP morphine 5 mg/kg ↑ CPP morphine 10 mg/kg ↔ CPP morphine 20 mg/kg |
|||
(Shams et al., 2006) | Swiss-Webster mice | male | 1. nicotine 0.0006–0.1 mg/kg, i.p. during test | CPP morphine 5 mg/kg, s.c. | 1. ↓ CPP expression |
2. nicotine 0.007–25 ng, i.c.v. during test | 2. ↓ CPP expression | ||||
(Vihavainen et al., 2008a) | NMRI mice | male | nicotine 50–500 μg/mL, drinking water 7 weeks before conditioning | CPP morphine 5 or 10 mg/kg, s.c. | CPP obtained with 5 mg/kg morphine only in nicotine pretreated group, CPP obtained with 10 mg/kg in both nicotine pretreated and control groups |
(Rezayof et al., 2007) | Wistar rats | male | 1. nicotine 0.25, 0.5 or 1 μg, intra-VTA | 1. CPP morphine 0.5 mg/kg, s.c. | 1. morphine CPP obtained only with nicotine 0.5 or 1 μg |
2. mecamylamine 2.5, 5 or 7.5 μg, intra-VTA | 2. CPP morphine 5 mg/kg, s.c. | 2. ↓ morphine CPP with mecamylamine 5 or 7.5 μg | |||
3. nicotine 1 μg + mecamylamine 5, 7.5 or 10 μg, intra-VTA | 3. CPP morphine 0.5 mg/kg, s.c. | 3. morphine CPP obtained only with nicotine 1 μg, reversed by mecamylamine 7.5 or 10 μg | |||
(Rezayof et al., 2006) | Wistar rats | male | 1. nicotine 0.25, 0.5 or 1 μg, intra-dHipp | 1. CPP morphine 0.5 mg/kg, s.c. | 1. morphine CPP obtained only with nicotine 0.75 or 1 μg |
2. mecamylamine 2, 4 or 8 μg, intra-dHipp | 2. CPP morphine 6 mg/kg, s.c. | 2. ↓ morphine CPP with mecamylamine 2, 4 or 8 μg | |||
3. nicotine 0.5, 0.75 or 1 μg + mecamylamine 8 μg, intra-dHipp | 3. CPP morphine 0.5 mg/kg, s.c. | 3. morphine CPP obtained only with nicotine, ↓ by mecamylamine | |||
(Hall et al., 2011) | Wistar rats | male | use-dependent nAChR antagonist BTMPS 0.1 or 0.5 mg/kg, s.c. | SA FR-1 morphine 0.25–1 mg/kg, i. | ↓ SA ↓ morphine seeking during abstinence |
(Zarrindast et al., 2003) | NMRI mice | female | mecamylamine 0.025, 0.05 or 0.1 mg/kg, i.p. | CPP morphine 5 mg/kg, i.p. | ↓ CPP acquisition |
1. nicotine 0.75, 1 or 2 μg, intra-BLA | 1. CPP morphine 0.5 mg/kg, s.c. | 1. morphine CPP obtained only with nicotine 0.75, 1 or 2 μg | |||
2. mecamylamine 1, 3 or 6 μg, intra-BLA | 2. CPP morphine 7.5 mg/kg, s.c. | 2. ↓ morphine CPP with mecamylamine 1, 3 or 6 μg | |||
(Zarrindast et al., 2005) | Wistar rats | male | 3. physostigmine 1, 3 or 5 μg, intra-BLA + atropine 7 μg, intra-BLA or + mecamylamine 6 μg, intra-BLA | 3. CPP morphine 0.5 mg/kg, s.c. | 3. morphine CPP obtained with physostigmine, reversed by atropine but not mecamylamine |
4. nicotine 0.75, 1 or 2 ug, intra-BLA + atropine 7 μg, intra-BLA or + mecamylamine 6 μg, intra-BLA | 4. CPP morphine 0.5 mg/kg, s.c. | 4. morphine CPP obtained with nicotine, reversed by mecamylamine but not atropine | |||
(Feng et al., 2011) | BALB/c mice | male | 1. nicotine alone 0.5 mg/kg, s.c. as reinstating stimulus | CPP morphine 10 mg/kg, s.c. | 1. no reinstatement with nicotine |
2. DHβE 5 mg/kg, s.c. or MLA 4 mg/kg, s.c. during drug prime reinstate physostigmine 0.5 mg/kg, s.c. | 2. ↓ CPP reinstatement with either DHβE or MLA | ||||
(Zhou et al., 2006) | Sprague-Dawley rats | male | + scopolamine (0.5 mg/kg, s.c.) or + mecamylamine (0.5 mg/kg, s.c.) or + saline | SA FR-5 heroin 0.05 mg/kg, i.v. | ↓ SA physostigmine alone, reversed by scopolamine but not by mecamylamine |
(Wright et al., 2019) | 1. C57BL/6 mice | male | |||
Wistar rats | 1. methyllycaconitine, 4 mg/kg, s.c. during conditioning, test or drug prime reinstatement | 1. CPP morphine 10 mg/kg, s.c. | 1. ↔ CPP acquisition ↔ CPP expression ↓ CPP reinstatement |
||
2. methyllycaconitine 6.75 μg intra-dHipp, -vHipp or -mPFC during reinstatement | CPP morphine 5 mg/kg, s.c. | 2. ↓ CPP reinstatement intra-vHipp only |
Abbreviations: BLA=basolateral amygdala; CPP=conditioned place preference; dHipp=dorsal hippocampus; FR=fixed ratio; IC=insular cortex; i.c.v.=intracerebroventricular; i.p.=intraperitoneal; i.v.=intravenous; mPFC=medial prefrontal cortex; nAChR=nicotinic acetylcholine receptor; SA=self-administration; s.c.=subcutaneous; vHipp=ventral hippocampus.
Relative to SA, studies using CPP are more widely represented in the literature, with some results opposing the conclusion drawn above that nicotine decreases opioid reward sensitivity. For example, in contrast to the nicotine-induced decrease in reward sensitivity reported recently (Loney et al., 2021), other reports show increased sensitivity to opioid CPP when nicotine is administered into various brain structures, including VTA, dorsal hippocampus, and basolateral amygdala (Rezayof et al., 2007, 2006; Zarrindast et al., 2005). These latter studies also show that microinjection of mecamylamine (a non-selective nicotinic receptor agonist) into these same regions decreases morphine CPP, an effect that is also obtained with systemic mecamylamine (Zarrindast et al., 2003). It is difficult to reconcile the findings showing decreased sensitivity to opioid reward when nicotine is administered systemically or into insular cortex, but increased sensitivity when nicotine is injected into VTA, dorsal hippocampus and basolateral amygdala. However, it appears that nicotinic receptors from different brain structures exert both excitatory and inhibitory control on opioid reward systems.
There has also been work examining the role of nicotinic receptors in reinstatement of opioid seeking. While one study found that nicotine is ineffective in eliciting morphine seeking following extinction of morphine CPP (Feng et al., 2011), antagonism of either β-containing nAChRs with dihydro-β-erythroidine or α7 homomeric nAChRs with methyllycaconitine reduces drug-primed reinstatement of morphine CPP (Feng et al., 2011; Wright et al., 2019). The effect of methyllycaconitine is mediated, at least in part, by α7 receptors in ventral hippocampus (Wright et al., 2019). These results with reinstatement align more closely with the notion that nicotinic receptors primarily function to increase, rather than decrease, opioid reward sensitivity, although the neurocircuitry involved in excitatory vs. inhibitory nicotinic modulation of opioid reward mechanisms remains to be elucidated.
2.3. Opioid contributions to nicotine reward
As summarized in Table 2, many studies have examined the role of opioid systems in modulating nicotine reward. In mice, development of nicotine CPP is reliably blocked by naloxone (Zarrindast et al., 2003; Walters et al., 2005) and by genetic deletion of β-endorphin production (Trigo et al., 2009) or μ opioid receptor (MOR) expression (Walters et al., 2005; Berrendero et al., 2002), strongly implicating a role for MOR in the control of nicotine reward. Similarly, while some null findings have been reported (Corrigall and Coen, 1991; DeNoble and Mele, 2006), nicotine self-administration in rats is decreased by naloxone and the long-lasting μ1 antagonist naloxonazine (Liu and Jernigan, 2011; Ismayilova and Shoaib, 2010). Reinstatement of nicotine seeking also involves MOR activation since a morphine prime reinstates nicotine CPP and naloxone blocks cue-induced reinstatement of nicotine seeking measured by SA (Biala and Budzynska, 2006; Liu et al., 2009).
Table 2.
References | Species | Sex | Opioid Manipulation | Nicotine Reward Test | Result on Nicotine Reward |
---|---|---|---|---|---|
(Biala and Budzynska, 2006) | Wistar rats | male | morphine 10 mg/kg, i.p. as reinstating stimulus | CPP nicotine 0.5 mg/kg, i.p. | induces reinstatement |
(Zarrindast et al., 2003) | NMRI mice | female | naloxone 0.5, 1 or 2 mg/kg, i.p. | CPP nicotine 1 mg/kg, i.p. | ↓ nicotine CPP, CPA with naloxone alone |
(Corrigall and Coen, 1991) | Long-Evans rats | male | naltrexone 0.1, 1 or 10 mg/kg, s.c. | SA FR-5 nicotine 0.03 mg/kg, i.v. | ↔ SA |
(Liu et al., 2009) | Sprague-Dawley rats | male | naltrexone 0.25,1 or 2 mg/kg, s.c. during SA or cue maintenance in extinction or cue reinstatement | SA FR-5 nicotine 0.03 mg/kg, i.v. | ↔ SA ↓ cue maintenance ↓ cue reinstatement |
(DeNoble and Mele, 2006) | Long-Evans rats | male | naloxone 0.75, 1.5 or 3 mg/kg, i.p. | SA FR-1 nicotine 0.32 mg/kg, i.v. | ↔ SA 1. ↓ CPP ↔ CPA |
Walters et al., 2005 | C57BL6 mice | male | 1. naloxone 1 mg/kg, s.c. during test day |
CPP nicotine 1 mg/kg, s.c. | 1. ↓ CPP ↔ CPA |
2. MOR KO vs WT | CPA nicotine 2 mg/kg, s.c. | 2. CPP in WT only CPA in both KO and WT |
|||
(Trigo et al., 2009) | C57BL/6 J mice | male | β-endorphin KO vs WT | CPP nicotine 0.5 mg/kg, s.c. | CPP in WT only |
(Berrendero et al., 2002) | C57/BL6 mice | female and male | MOR KO vs WT | CPP nicotine 0.5, 0.7 or 1 mg/kg, s.c. | CPP 0.5 or 0.7 mg/kg in WT only |
(Ismayilova and Shoaib, 2010) | Lister rats | male | 1. Naloxone 0.3, 1 or 3 mg/kg, s.c. | SA FR-3 nicotine 0.03 mg/kg, i.v. | 1. ↓ SA 0.3, 1 or 3 mg/kg |
2. DOR antag naltrindole 0.3, 1 or 3 mg/kg, s.c. | 2. ↔ SA | ||||
3. KOR agonist U50,488 0.3, 1 or 3 mg/kg, s.c. | 3. ↓ SA 3 mg/kg | ||||
(Liu and Jernigan, 2011) | Sprague-Dawley rats | male | 1. MOR antag naloxonazine 5 or 15 mg/kg, i.p. | SA FR5 nicotine 0.03 mg/kg, i.v. | 1. ↓ SA |
2. DOR antag naltrindole 0.5 or 5 mg/kg, i.p. | 2. ↔ SA | ||||
3. KOR antag GNTI 0.25 or 1 mg/kg, i.p. | 3. ↔ SA | ||||
Berrendero et al., 2012 | C57BL/6 J mice | male | 1. DOR KO vs. WT | 1. CPP nicotine 0.17 mg/kg, s.c. or SA FR-1 nicotine 0.015 or 0.03 mg/kg, i.v. |
SA DOR < WT acquisition and intake |
2. DOR antag naltrindole 2.5 or 5 mg/kg, i.p. |
2. SA FR-1 and PR nicotine 0.03 mg/kg, i.v. | 2. SA ↓ FR acquisition ↔ PR breakpoint | |||
(Berrendero et al., 2005) | C57BL/6 J mice | female and male | enk KO vs WT | CPP nicotine 0.25, 0.5 or 1 | CPP in WT |
(Al-Hasani et al., 2013) | C57BL/6 mice | male | KOR agonist U50,488 5 mg/kg, i.p. as reinstating stimulus | CPP nicotine 0.5 mg/kg, s.c. | induces reinstatement |
(Grella et al., 2014) | Long Evans rats | male | 1. KOR antag nor-BNI 10 mg/kg, i.p. during yoh reinstatement | SA FR-3 or -5 nicotine 0.03 mg/kg, i.v. | 1. ↓ yoh reinstatement |
2. KOR agonist U50,488 1, 2.5 or 5 mg/kg, i.p. as reinstating stimulus | 2. induces reinstatement | ||||
3. nor-BNI 10 mg/kg, i.p. during cue reinstatement | 3. ↔ cue reinstatement | ||||
(Smith et al., 2012) | C57BL/6 mice | male | 1. KOR antag nor-BNI 10 mg/kg, i.p. during swim stress reinstatement | CPP nicotine 0.5 mg/kg, s.c. | 1. ↓ stress reinstatement |
2. KOR agonist U50,488 2.5, 5 or 10 mg/kg, i.p. as reinstating stimulus + nor-BNI 10 mg/kg, i.p. | 2. U50,488 induces reinstatement, reversed by nor-BNI | ||||
3. U50,488 5 mg/kg, i.p. as reinstating stimulus + nor-BNI 2.5 μg intra-Amyg or intra-VPN | 3. U50,488 induces reinstatement, reversed by nor-BNI intra-Amyg, but not intra-VPN | ||||
(Jackson et al., 2010) | ICR mice | male | KOR antag JDTic 8 or 16 mg/kg, s.c, on test | CPP nicotine 0.5 mg/kg, s.c. | ↔ expression |
Jackson et al., 2013 | ICR mice | male | KOR antag nor-BNI 10 mg/kg, s.c. during drug prime or swim stress reinstatement | CPP nicotine 0.5 mg/kg, s.c. | ↔ drug prime reinstatement ↓ stress reinstatement |
(Galeote et al., 2008) | C57BL/6 J mice | male | proDYN KO vs. WT | CPP nicotine 0.5 mg/kg, s.c. or SA FR-1 or PR nicotine 0.0052-0.0855 mg/kg, i.v. | CPP KO = WT SA acquisition KO > WT at 0.0052 mg/kg dose PR breakpoint KO < WT at 0.0427 mg/kg dose |
(Nygard et al., 2016) | C57BL/6 J mice | male | 1. KOR antag nor-BNI 10 mg/kg, i.p. during footshock, yoh or drug prime reinstatement | CPP nicotine 0.5 mg/kg, s.c. | 1. ↓ footshock reinstatement ↓ yoh reinstatement ↔ drug prime reinstatement |
2. KOR KO vs pre-pro-DYN KO vs WT during yoh or drug prime reinstatement | 2. ↓ yoh reinstatement ↔ drug prime reinstatement each KO vs WT | ||||
3. nor-BNI 2.5 μg, intra-BLA during acquisition or yoh reinstatement or drug prime reinstatement | 3. ↔ acquisition ↔ drug prime reinstatement ↓ yoh reinstatement | ||||
4. KOR cKO AAV-cre-GFP or AAV-GFP, intra BLA footshock and drug prime reinstatement | 4. ↓ footshock reinstatement ↔ drug prime reinstatement AAV-cre-GFP vs AAV-GFP | ||||
5. AAV5-CaMKIIa-hM4D(Gi)-mCitrine or control virus DREADD into BLA, CNO activation as reinstating stimulus | 5. induces nicotine seeking, AAV5-CaMIIa-hM4D(Gi)-mCitrine DREADD only | ||||
(Cippitelli et al., 2016) | Sprague-Dawley rats | male | 1. NOP agonist AT-202 0.3, 1 or 3 mg/kg, i.p. | SA FR-1 co-use nicotine 0.03 mg/kg, i.v. ethanol 10 % oral, 01. mL | 1. ↑ nicotine SA ↔ ethanol SA |
2. NOP antag SB612111 1, 5 or 10 mg/kg, i.p. | 2. ↓ nicotine SA 5 and 10 mg/kg ↓ ethanol SA 10 mg/kg only | ||||
(Sakoori and Murphy, 2009) | C57BL/6 mice | male | NOP receptor KO vs WT | SA 2-bottle choice nicotine 3, 6, 12, 25 or 50 μg/mL, oral | intake KO > WT 3 μg/mL KO = WT 6, 12, 25 or 50 μg/mL |
Abbreviations: AAV=adeno-associated virus; Amyg=amygdala; BLA=basolateral amygdala; CaMKII=calcium/calmodulin-dependent protein kinase II; CNO=clozapine N-oxide; CPA=conditioned place aversion; CPP=conditioned place preference; DOR=delta opioid receptor; DREADD=designer receptor exclusively activated by designer drug; DYN=dynorphin; enk=enkephalin gene; FR=fixed ratio; GFP=green fluorescent protein; i.p.=intraperitoneal; i.v.=intravenous; KO=knockout; KOR=kappa opioid receptor; MOR=mu opioid receptor; NOP=nociception opioid peptide; nor-BNI=nor-binaltorphimine; PR=progressive ratio; SA=self-administration; s.c.=subcutaneous; VPN=ventral posterior thalamic nucleus; WT=wild-type; yoh=yohimbine.
Beyond MOR, genetic evidence also suggests involvement of the δ opioid receptor (DOR) in nicotine reward. In particular, DOR knockout mice fail to develop nicotine CPP and show diminished SA compared to wild-type counterparts (Berrendero et al., 2012). Deletion of enk yields a similar loss of nicotine CPP (Berrendero et al., 2005). Despite this genetic evidence, however, there is only sparse pharmacological data to support a role for DOR in nicotine reward. While one study found that naltrindole slowed acquisition of nicotine SA in mice (Berrendero et al., 2012), this same study found no effect on progressive ratio breakpoint. Moreover, others have reported no effect of naltrindole on nicotine SA in rats (Liu and Jernigan, 2011), bolstering the conclusion that DOR blockade does not alter nicotine reward. Together, these results suggest that genetic deletion of DOR or enk may produce some compensatory neural change in non-DOR systems to alter nicotine reward, which does not translate into pharmacological effects at DOR in wild-types.
Similar to other non-opioid drugs of abuse such as alcohol and cocaine (Funk et al., 2014; Redila and Chavkin, 2008), considerable evidence supports a role for the κ opioid receptor (KOR) in the expression of stress-induced nicotine seeking following a period of extinction. Importantly, prodynorphin KO does not alter acquisition of nicotine CPP (Galeote et al., 2008) and KOR antagonists have no effect on acquisition or expression of nicotine CPP (Nygard et al., 2016; Jackson et al., 2010). In addition, KOR antagonists have no effect on drug- or cue-primed reinstatement of nicotine seeking using CPP (Nygard et al., 2016; Jackson et al., 2013; Grella et al., 2014). Instead, KOR antagonists have a selective ability to decrease stress-induced reinstatement induced by forced swim, footshock or administration of the sympathomimetic yohimbine (Nygard et al., 2016; Jackson et al., 2013; Grella et al., 2014; Smith et al., 2012). The KOR agonist U50,488 also induces reinstatement of nicotine seeking (Al-Hasani et al., 2013) and this effect is reversed by a KOR antagonists delivered either systemically or directly into the amygdala (Smith et al., 2012). Viral activation and genetic knockdown studies also support a specific role of KOR in central nucleus of amygdala in stress-induced nicotine seeking (Nygard et al., 2016).
There is little information on the potential role of nociceptin opioid peptide (NOP) receptors on nicotine reward. NOP receptor KO mice drink more nicotine in a 2-bottle choice (Sakoori and Murphy, 2009), although the effect is modest and occurs only with a dilute nicotine concentration (3 μg/mL) that may not be behaviorally relevant. Nonetheless, a recent study found that nicotine SA is increased by a NOP receptor agonist and decreased by a NOP receptor antagonist (Cippitelli et al., 2016), thus providing an impetus for more work with this opioid receptor subtype. Until then, the main conclusions to be drawn at this time are that: (1) MOR plays a key role in nicotine reward and reinstatement; and (2) KOR plays a specific role in stress-induced reinstatement of nicotine seeking.
3. Nicotine dependence and opioid use disorder: antinociception
3.1. Anatomy of nociception pertaining to nicotine and opioid co-use
Pain signals are detected by nociceptors in the periphery and are transmitted to the dorsal horn of the spinal cord via nociceptive primary afferents where they synapse with projection neurons, which in turn cross the anterior white commissure to ascend the spinothalamic tract for thalamic relay and somatosensory cortical processing. As a result of incoming pain information, opioid interneurons in the periaqueductal grey (PAG) inhibit PAG activity, disinhibiting serotonergic rostral ventral medulla (RVM) neurons, and noradrenergic projections arising from the locus coeruleus (Al-Hasani et al., 2013; Ossipov et al., 2010). The heavy involvement of opioid receptors in the descending and ascending pain pathways allows exogenous opioids, such as morphine, heroin, and fentanyl, to inhibit pain signaling, resulting in a decrease in nociception (Al-Hasani et al., 2013).
Nicotine also works within some of the same pain neurocircuitry components in which opioids act. For example, one study found that intra-vIPAG injection of PHA-543613, an α7 nAChR selective agonist, produced a dose-dependent antinociceptive effect compared to vehicle as determined by a formalin assay in rats, an effect reversed by pre-treatment with an α7 nAChR antagonist (Umana et al., 2017). Additionally, intra-RVM injection of mecamylamine, prevents immobilization stress-induced antinociception in rats following carrageenan-induced mechanical hyperalgesia and formalin-induced paw flinch nociception (Tobaldini et al., 2020). This supports that nicotinic agonists produce antinociception by targeting opioid- and pain-associated brain areas and that nicotine and opioids may interact to alter pain perception.
3.2. Nicotinic modulation of opioid antinociception
Nicotine specifically mediates the antinociceptive properties of opioids by acting as an enhancer of opioid-induced antinociception and increases opioid antinociceptive tolerance when co-used. Fentanyl, oxycodone, and buprenorphine dose-dependently increase squirrel monkey tail-withdrawal latency to hot water, and pre-treatment with nicotine shifts the dose response to the left without altering opioid-induced decreases in milk self-administration (Barreto de Moura et al., 2019). In rodent models, co-administration of the nAChR β2* agonist A85380 (where * indicates the presence of other subunits in addition to the β2) and fentanyl similarly enhances the ability of fentanyl to decrease pain responsivity to thermal plate- and formalin-induced nociception (Ren et al., 2019). Acute nicotine also increases the percent of maximum possible effect (%MPE) of tail flick latency after tail heat exposure in rats given codeine.
In contrast to the enhancement in opioid-induced antinociception observed with acute nicotine, repeated nicotine exposure increases the rate of tolerance for a codeine/nicotine combination (McMillan and Tyndale, 2017). Similarly, human tobacco smokers may be more tolerant to the antinociceptive properties of opioids. A meta-analysis of results from humans suggests that nicotine reduces pain in humans to a small, but significant extent, with more robust effects found in studies using men (Ditre et al., 2016). However, continued nicotine use increases the need for pain relief with opioids after surgery. For example, one study found that smokers deprived of nicotine after coronary artery bypass graft surgery required more opioids for pain relief than non-smokers (Creekmore et al., 2004). A similar effect was found in male smokers following distal gastrectomy with gastroduodenostomy (Kim et al., 2017). Considering the increased rate for OUD among smokers (Kalman et al., 2005), research should examine compatible treatments to reduce the need for increased opioid use in patients with ND.
Overall, the evidence above collectively suggests that nicotine interacts with the opioid system to enhance opioid-induced antinociception when used in combination. However, the complexity of this question is apparent given the dissociable effects of acute and chronic nicotine treatments on opioid-induced analgesia. One potential reason for these observations may be due to the peculiar pharmacology of nicotine action when delivered acutely versus chronically. For example, previous fundamental pharmacological studies both in vitro and in vivo examining the acute and chronic effects of nicotine at the receptor level suggest that while nicotine is a super-agonist at nAChRs, its agonist effects at non-α7 receptors rapidly results in long-lasting desensitization of the receptor (Hulihan-Giblin et al., 1990; Turner et al., 2013a). This effectively results in nicotine acting as a “time-averaged” antagonist. While this has not been directly evaluated in opioid nociceptive studies as yet, utilization of allosteric modulators rather than agonists or partial agonists, may be more effectively clinically.
3.3. Opioid modulation of nicotinic antinociception
Considering the role of endogenous opioids in pain pathway regulation, exogenous opioids are frequently used as treatment for pain in the medical setting (National Academies of Sciences, E. et al., 2017). However, the antinociceptive effects of nicotine may also be modulated by opioid use since opioid receptor manipulation alters nicotine-induced antinociception. When MORs are absent, nAChR agonists no longer produce or enhance antinociception. For example, MOR KO mice show reduced nicotine-induced antinociception compared to wild-type mice tested using either the tail-flick or hotplate assays (Berrendero et al., 2002). Another study found that MOR KO mice express accelerated tolerance to the antinociceptive effects of nicotine compared to wild-type mice (Galeote et al., 2006). However, DOR KO mice show similar tolerance to the antinociceptive effect of chronic nicotine across a 12-day period compared to wild-type mice as determined by tail-immersion tests (Berrendero et al., 2012). While most research examining the impact of opioid system manipulation on nicotine-induced antinociception has been performed in opioid receptor KO rodents, one study found that pretreatment with a MOR, DOR, or KOR-opioid antagonist (β-FNA, naltrindole, or nor-BNI) reduces the antinociceptive effect of i.c.v. nicotine on acetic acid writhing in mice (Kwon et al., 2008). Thus, while the evidence is relatively sparse, it indicates that manipulation of the opioid system may alter nicotine-induced antinociception and should be a target for future work.
4. Nicotine dependence and opioid use disorder: respiration
4.1. Anatomy of respiration pertaining to nicotine and opioid co-use
The neural control of respiration involves multiple neurotransmitters emanating from anatomically distinct clusters of brainstem neurons that descend the spinal cord to synchronize breathing. While a comprehensive description is beyond the scope of this review, as described in detail previously (McCrimmon et al., 2008), there are three main groups of respiratory neurons. The first is the dorsal respiratory group located in the nucleus of the solitary tract, which are primarily inspiratory neurons under glutamatergic control. The second is contained within the ventrolateral medullar column that extends from the spinal-medulla junction to the facial nucleus, which are both inspiratory and expiratory neurons that utilize glutamate, GABA and glycine. The third is the pontine respiratory group within the dorsolateral pons, which are heterogeneous neurons utilizing glutamate, enkephalin and catecholamines. These three groups receive top-down modulation by serotonergic and catecholaminergic input from midbrain periaqueductal gray, hypothalamus and cortex. In addition, most relevant for this review, there is also modulatory control of medullary respiratory neurons through stimulation of α4β2 nicotinic receptors (Ren et al., 2019), as well as through inhibition via MOR of both the medullary and pontine respiratory groups (Varga et al., 2019).
4.2. Nicotinic modulation of opioid effects on respiration
Heroin is known to produce profound life-threatening respiratory depression that is reversed by naloxone (Jolley et al., 2015; Strang et al., 2016). Moreover, the recent accelerating opioid overdose crisis is related primarily to the introduction of high potency opioids such as fentanyl and carfentanil into the illicit drug market (Volkow et al., 2019). In addition to producing profound respiration depression similar to heroin, high-potency fentanyl analogues also induce vocal chord closure and wooden chest syndrome that contributes to lethality (Torralva and Janowsky, 2019) and all of these untoward effects on breathing involve multiple mechanisms beyond mere occupation of opioid receptors (Torralva et al., 2020).
Stimulation of nicotinic receptors has recently been advanced as a potential intervention for countering opioid-induced respiratory depression (Imam et al., 2020). Using whole-body plethysmography in anesthetized rats, recent studies found that fentanyl-induced respiratory depression is reversed by nicotine or A85380 (Ren et al., 2019), as well as by partial agonists such as varenicline (Ren et al., 2020). Interestingly, the reversal of respiration depression is not accompanied by a reversal of the motoric rigidity that also accompanies high-dose fentanyl exposure. The dissociation between respiratory and motoric depression likely reflects that nicotinic receptors modulate MOR in respiratory brainstem nuclei (Ren et al., 2019), but not in motoric striatal circuits (Havemann and Kuschinsky, 1981). Since the α7 agonist PNU282987 has no effect on fentanyl-induced suppression of respiratory circuits (Ren et al., 2019), β-containing nAChRs play the key role. However, some caution is needed in interpreting these results using an anesthetized rat preparation because a recent study using conscious rats failed to find an effect of A85380 in reversing sufentanil-induced respiratory depression (Dandrea and Cotten, 2021), thus begging for further work on this vital clinical problem related to opioid overdose.
4.3. Opioid modulation of nicotinic effects on respiration
Most work assessing the effects of nicotine on respiration in humans has used tobacco cigarettes as the delivery device, which can lead to either decreases or increases in respiration based on individual differences (Jones, 1987). Similarly, different inbred mouse strains show variable respiratory responses to nicotine, with dose- and time-dependent decreases or increases occurring based on the genetic strain assessed (Marks et al., 1989). In general, however, preclinical studies in rodents and monkeys indicate that exposure to either tobacco smoke or systemic nicotine initially inhibits breathing, followed by a rebound tachypnea (Bloom, 2019; Howell, 1995; Lee et al., 1990). In dogs, nicotine-induced tachypnea is potentiated by naloxone pretreatment (Kamerling et al., 1982), indicating the involvement of opioid receptors
Evidence also indicates that the initial respiratory depressant effect of nicotine is modulated by opioid receptors. For example, the respiratory depression produced by acetylcholinesterase inhibition in rats, while reversed by muscarinic antagonists, also involves a significant nicotinic receptor component that is sensitive to naloxone (Skrbic et al., 2017). Even more directly relevant, naltrexone administration prevents the respiratory depressant and lethal effects of intravenous nicotine in anesthetized rats (Sloan et al., 1989), supporting the notion that nicotine depresses respiration via central opioid mechanisms (Krause et al., 2018).
5. Neural mechanisms contributing to overlapping nicotine and opioid co-use
Several lines of evidence suggest a functional interaction of the nicotinic and opioidergic systems in the development and maintenance of drug-seeking. Human and rodent studies indicate a bidirectional relationship between nicotine and opioid systems that is critical for the development and establishment of their drug dependence (Young-Wolff et al., 2017; Krause et al., 2018; Almeida et al., 2000; Trigo et al., 2009); W For example, nicotine modulates opioid actions in the mesolimbic circuit by enhancing the anxiolytic effects (Zarrindast et al., 2008) and rewarding properties of MOR agonists (Rezayof et al., 2006; Zarrindast et al., 2003; Berrendero et al., 2002; Trigo et al., 2009),which are heavily implicated in relapse and the development of dependence. Similarly, a human laboratory study showed that opioid receptor non-selective antagonism with naloxone challenge dose-dependently promotes nicotine withdrawal signs and alters craving in nicotine-dependent subjects (Krause et al., 2018). This portion of the review will examine the interaction of these drugs at the receptor and circuitry levels, with some integration of behavioral endpoints for functional emphasis.
5.1. Nicotinic and opioidergic interactions: receptors and downstream signaling
Nicotinic and opioidergic receptors are often tandem co-regulators, whether on the same cell or on different cells within the same circuit. In fact, these receptors can even be activated by the converse drug; for example, an in vitro study in human cell lines suggests that morphine acts directly as a partial agonist of α4β2 nAChRs (Talka et al., 2013). However, the predominant observations at the receptor level relate to (1) expression changes of the receptors due to exposure to the converse drug or (2) alterations in common downstream signaling mechanisms. For example, nicotine treatment in mice lacking MOR does not activate the transcriptional factor CREB in the VTA, and, similarly, naloxone precludes nicotine-induced phosphorylation of CREB (Walters et al., 2005).
Notably, the findings regarding the effect of nicotine on MOR expression are are are mixed. For example, in rodents, while chronic high doses of nicotine (5 mg/kg, twice daily) decrease MOR density in the mice NAc and dorsal striatum (Galeote et al., 2006), other studies using lower doses (0.3 mg/kg, twice daily) show increased MOR expression in rat striatum (Wewers et al., 1999). This is also true in the human literature, as a series of studies using positron emission tomography (PET) imaging to assess MOR binding potential (BP) availability after smoking or placebo have yielded mixed results. For instance, some studies showed increased C-carfentanil BP after smoking in areas such as the Nac, amygdala, and putamen, while decreased BP was observed in Nac, ACC, striatum, PFC, and, hippocampus, possibly indicating MOR up- or down-regulation according to the region (Domino et al., 2015; Nuechterlein et al., 2016; Scott et al., 2007). Moreover, some studies did not identify any differences in MOR binding between placebo and active cigarette groups in any region examined (Ray et al., 2007; Kuwabara et al., 2014). Despite these discrepant findings, one study in human smokers observed that MOR availability in reward-related brain areas was negatively correlated to Fagerstrom Test for Nicotine Dependence (FTND) scores (Domino and Hirasawa-Fujita, 2019). This suggests that inter-individual variability may play a role in the discrete regulation of MORs by nicotine and that the expression levels of MORs may directly impact nicotine dependence and withdrawal behavioral phenotypes. This is supported by the finding that expression of MORs and endogenous opioids are required for nicotine reward and dependence (Walters et al., 2005).
Conversely, opioids can also regulate nicotinic receptor expression. For example, naloxone decreases the activity and expression of α4β2 and α7 nAChRs induced by nicotine treatment (Almeida et al., 2000) Studies have also found that naltrexone inhibits nicotine-induced up-regulation of nAChRs, a hallmark of nicotine exposure; this may indicate that opioid antagonism exerts direct effects on nAChRs, thus leading to decreased activity in the mesolimbic pathway and precipitation of withdrawal (Kenny and Markou, 2001). However, these effects may be MOR-specific, as a recent multicenter human laboratory study assessing the efficacy of a short-acting KOR antagonist in smokers found that the administration of a KOR antagonist was ineffective in altering smoking latency, consumption, and withdrawal, as well as any affective measures of smoking, compared to placebo (Jones et al., 2020).
In addition to the effects of nicotine and opioids on receptor expression, these drugs seem to also converge on common intracellular signaling pathways. As an example, the extracellular signal-regulated kinase (ERK) is a signaling pathway involved in the neuroadaptations caused by drugs of abuse. Both nicotine and morphine administration lead to increased ERK expression and activity in various regions within the mesolimbic system (for review, see Zhai et al., 2008). Once activated via phosphorylation, pERK translocates to the nucleus (Chen et al., 1992) and indirectly regulates several transcription factors, including the canonical transcription factor cAMP Response Element-Binding Protein (CREB) (Brami-Cherrier, 2007; Xing et al., 1998). In the nucleus, CREB activation culminates in the transcription of several genes, such as NRG3 (Turner et al., 2013b), that have been implicated in addiction processes (Fisher et al., 2017; Portugal et al., 2012). Interestingly, blood samples from smokers exhibit higher levels of both phosphorylated ERK (pERK) and CREB (pCREB) compared to never smokers (Lenz, 2012; Lenz et al., 2010, 2012). It remains to be determined if similar changes in blood occur among individuals with OUD.
5.2. Nicotinic and opioidergic interactions: regulation of dopamine and other neurotransmitter release
Extending these observations from the receptor level to the circuit level, both nicotinic and opioidergic receptors also contribute to dopaminergic signaling, which is critically important for drug reward. While both receptors can regulate the release of other neurotransmitters, this function is a canonical role for nicotinic receptors, which are predominantly found presynaptically or preterminally in the central nervous system. Due to this positioning, their activation can elicit neurotransmitter release from the synaptic terminal without depolarization of the entire neuron.
With regards to dopaminergic co-regulation by nicotine and opioids, the predominant regions involved are within the mesolimbic circuitry, namely the nucleus accumbens (NAc) and the ventral tegmental area (VTA). There is considerable evidence that both nicotine and opioid agonists trigger the release of dopamine from terminals in the NAc and, more broadly, from striatal terminals, both of which have behavioral consequences (Di Chiara and Imperato, 1988). This is thought to be due to the effects of nicotine and opioids signaling in both the NAc, as well as the VTA. For example, microinfusion studies show direct effects of nicotine on dopamine release in both the VTA and NAc (Pistillo et al., 2015). In both the mesolimbic and nigrostriatal terminal fields, nicotine appears to modulate DA via activation of presynaptic β2-containing nAChRs (Mamaligas et al., 2016).
In addition to these isolated effects of nicotine on dopamine signaling, there is considerable evidence of overlapping regulatory mechanisms by both nicotine and opioids. For example, proenkephalin (PENK) genetic deletion reduces nicotine-induced dopamine release in the NAc, suggesting that nicotine’s effects can also act indirectly via MOR activation (Berrendero et al., 2005). These effects may also encompass metabolic effects of nicotine, as neurochemical studies indicate that chronic nicotine sensitizes dopaminergic pathways to morphine, which augments dopamine turnover and metabolism in the striatum (Vihavainen et al., 2006).
However, there is cumulative evidence that synchronized action of nAChRs and opioid receptors results in more homeostatic modulation of dopamine outflow in the striatum. For example, electrochemical studies suggest that endogenous opioids are released in the NAc to modulate dopamine neurotransmission via MOR activation on NAc cholinergic interneurons (Britt and McGehee, 2008). This results in effective modulation of striatal dopamine by inhibiting acetylcholine release and reducing nAChR-mediated dopamine release (Britt and McGehee, 2008). This may be a salient point for clinical applications, as cigarette smoking produces dose-related increases of circulating β-endorphin in human subjects (Pomerleau et al., 1983). In addition to the opioid effects through MOR activation, KOR activation by opioids can have both direct effects by triggering dopamine release from presynaptic neurons, as well as indirectly by their inhibitory effects on cholinergic interneurons in the NAc (Mamaligas et al., 2016; Britt and McGehee, 2008).
The actions of nicotine and opioids within the VTA regulate the overall firing of dopaminergic neurons in the mesolimbic pathway, with MOR and nicotinic agonists enhancing net excitatory activity by disinhibition of dopaminergic neurons. For example, nicotine microinjection in the VTA enhances dopamine overflow in the NAc, an effect that is reversed by nAChR antagonism (Nisell et al., 1994). Further, activation of MORs in VTA GABAergic interneurons result in inhibited GABA release and subsequently increases dopamine firing and release in the NAc, an effect that is precluded by opioid antagonists (Tanda and Di Chiara, 1998). Additionally, chronic nicotine exposure and withdrawal alters GABAergic transmission in the VTA and GABA response to morphine, possibly underlying the potentiation of locomotor behaviors (Vihavainen et al., 2008b)..
6. Genetic factors impacting both nicotine and opioid dependence
An intricate relationship of polygenic and environmental factors underlies SUD phenotypic manifestations (Walker and Nestler, 2018). For example, heritability accounts for upwards of 70 % of ND (Lessov et al., 2004), with data from familial and twin studies indicating that genetic liability for smoking initiation is estimated at 60 %. In comparison, the other 40 % of variance pertains to the influence of environmental factors due to individual characteristics or features of the twin pair (Sullivan and Kendler, 1999). Likewise, for OUD, genetic liability is estimated at around 70 % (Goldman et al., 2005). In this section, we will discuss some of the genetic contributors in common between ND and OUD, with an emphasis on variants in the nAChRs or MORs themselves. Furthermore, while the section will focus on tobacco and opioid use disorders, variation within these targets have also been implicated in SUDs broadly (Arias et al., 2006; Bart et al., 2005; Brynildsen and Blendy, 2021; Ide et al., 2004).
6.1. Polymorphisms in nAChR subunits and effects on nicotine and opioid dependence
Genetic association (GWAS) studies have identified numerous risk alleles for nicotine and opioid addiction. Among these genes, the most well described haplotype is in the nAChR gene cluster (CHRNA5–CHRNA3–CHRNB4 locus), encoding the α5, α3, and β4 nicotinic subunits. Variants in this nAChR gene cluster, particularly CHRNA5 (rs16969968), have been implicated in the addiction vulnerability across various addictive substances, including nicotine and opioid addiction (for review, see Brynildsen and Blendy, 2021). For example, SNPs in CHRNA5 are linked to both lifetime ND and OUD in European and Afro-American families (Sherva et al., 2010). For example, the A allele of the rs16969968 SNP within CHRNA5 is associated with a more severe OUD presentation (Erlich et al., 2010) and mechanistic fMRI studies of this variant found higher connectivity in the habenula-caudate circuitry in opioid users carrying the CHRNA5 G allele. Considering that the habenula is (1) enriched in both nAChRs and opioid receptors and (2) is known to modulate aversion, negative reinforcement and drug withdrawal, it was suggested that high-risk alleles might confer lower sensitivity to the aversive properties of opioids, increasing the susceptibility and severity of OUD (Curtis et al., 2017).
With respect to nicotine-specific endophenotypes, SNPs within the CHRNA5–CHRNA3–CHRNB4 cluster have been implicated in heavy smoking, the predisposition for ND, and smoking-related diseases (Bierut et al., 2008; Amos et al., 2008), nicotine withdrawal, craving, and inability to quit (Baker et al., 2009; Quach et al., 2020; Chen et al., 2020). Interestingly, the CHRNA5 risk allele attenuates the aversive properties of nicotine in both rodents (Fowler et al., 2013) and humans (Jensen et al., 2015), possibly increasing smoking consumption. Moreover, these CHRNA5 risk alleles also predict the failure to quit among untreated patients (Chen et al., 2012) or after discontinuation of pharmacotherapy (Sarginson et al., 2011). These findings highlight the important role of genetic variation in the nicotinic receptor family to contribute to nicotine dependence and its associated endophenotypes.
6.2. Polymorphisms in MOR and effects on nicotine and opioid dependence
Evidence from transgenic mouse studies demonstrates that MORs are necessary for the somatic manifestation of nicotine- and morphine-induced rewarding and antinociceptive phenotypes (Berrendero et al., 2002; Matthes et al., 1996). This is in line with a number of human genetic studies implicating variants within the MOR gene (OPRM1) with both ND and OUD outcomes. Studies examining the well-described nonsynonymous OPRM1 SNP (termed A118G or rs10485057) have associated it with both ND (Zhang et al., 2006) and OUD (Tan et al., 2003; Bart et al., 2004; Kapur et al., 2007). This functional OPRM1 SNP (A118G) markedly increases the binding affinity of MOR to its ligand β-endorphin (Bond et al., 1998), while at the same time reduces mRNA and protein expression of MORs in rodent models (Mague et al., 2009). These complex effects result in the functional attenuation of the rewarding and antinociceptive effects of morphine (Mague et al., 2009). This rodent work has been replicated in human imaging studies, demonstrating reduced MOR binding in mesolimbic areas of G-carriers of the A118G polymorphism (Ray et al., 2011), and may underlie it’s association with OUD (Tan et al., 2003; Bart et al., 2004; Kapur et al., 2007).
With regards to its contribution to nicotine dependence phenotypes, both GWAS and twin studies implicate polymorphisms in the OPRM1 gene in smoking susceptibility, ND, and relapse. For example, analyses from individuals recruited from a twin study database revealed a significant association of a three marker (rs2075572, rs10485057, and rs10485058) haplotype block within the OPRM1 gene and smoking initiation (Zhang et al., 2006). Additionally, the A118G polymorphism has also been associated with ND endophenotypes, such as the rewarding properties of tobacco smoking induced by negative mood, (Perkins et al., 2008), alterations in the self-reported rewarding effects of nicotine (Ray et al., 2011) and general nicotine reward in male mice and human subjects (Bernardi et al., 2016). Additionally, a recent GWAS study in individuals with or without an OUD diagnosis found that the A118G variant associated was associated with nicotine dependence in individuals with OUD (Zhou et al., 2020). However, the precise mechanism by which this variant contributes to increased nicotine dependence in this population is unknown.
7. Clinical trials and future directions for co-dependency of nicotine and opioids
Concurrent use of tobacco products and opioids is mutually reinforcing Story and Stark, 1991). (Clinical observations show that tobacco use promotes the escalation of methadone regimens for OUD patients (Schmitz et al., 1994). Conversely, methadone dose-dependently increases cigarette consumption. Thus, smoking rates are overwhelmingly high in OUD individuals relative to the general population. Despite the intent to quit, successful attempts at smoking cessation are markedly reduced in OUD individuals, rendering them vulnerable to health-related complications from tobacco use. In fact, the mortality rate due to smoking-related illness approaches 50 % among SUD patients (Hurt et al., 1996; Hser et al., 1994). Moreover, the efficacy of the currently available medications for tobacco cessation is modest in opioid co-users (Miller and Sigmon, 2015), underscoring the critical gap in effective treatments for these individuals. This has led to a series of randomized controlled trials assessing pharmacological and multimodal interventions for smoking cessation in opioid-maintained patients has been conducted, with biochemically validated abstinence measures as the primary outcome in patients enrolled in opioid treatment programs (Table 3). We will briefly describe a subset of these studies below.
Table 3.
References | Participants | Intervention | Assessment:Primary and Secondary Outcomes |
Follow-up time point for abstinence |
Results |
---|---|---|---|---|---|
(Hall et al., 2018) | Buprenorphine-maintained smokers | 1. Skills training + NRT (patch 7 mg, 14 mg or 21 mg), (lozenges 2 mg combination or varenicline + CBT) | 1. CPD and biochemical: CO + urine | 3, 6, 12, and 18 months | The interventional group had higher abstinence rates than controls at 3-months FU. |
N = 175 | 2. Information (control) 12 weeks | 2. Questionnaires: FTND and others. | CPD and cannabis a month prior to FU as a predictor of smoking continuance | ||
(Nahvi et al., 2014) | Methadone-maintained Smokers | 1. Varenicline (2 mg) | 1. CO abstinence at 12 weeks FU | 2, 4, 8, 12, and 24 months | Varenicline higher abstinence rates at 12-weeks FU |
N = 112 | 2. Placebo 12 weeks | 2. CO abstinence each FU, CPD, quit attempts >24 h, psychiatric or cardiac adverse events | Reduced CPD at 12 weeks | ||
No differences in psychiatric or adverse events between groups | |||||
(Rohsenow et al., 2017) | SUD, including OUD outpatient smokers | 1. Varenicline (2 mg) or placebo | 1. CO + urine cotinine | 3, 6 months | Varenicline was more effective than NRT in promoting abstinence at a 3-months timepoint. |
N = 137 | 2. NRT (patch 7 mg, 14 mg or 21 mg) or placebo 12 weeks | 2. CPD, or continuous abstinence assessment | |||
(Stein et al., 2013) | Methadone-maintained Smokers | 1. Varenicline (2 mg) | 1. CO + salivary cotinine at 3months | 6 months | Varenicline was not more effective than placebo in promoting abstinence at a 6-month time point. |
N = 315 | 2. NRT combination (patch 21 mg or 42 mg; 4 mg gum) | 2. CO at 6 months, and urine test, CPD, self-report abstinence for drugs | |||
(Reid et al., 2008) | Methadone-maintained Smokers | 1. NTR (14 mg or 21 mg) + CBT | 1. CO + urine | 1–9, 13, and 26 weeks | NRT + CBT promotes a small but significant reduction of CPD and abstinence during the treatment period compared to the control group. |
N = 225 | 2. Treatment as usual 9 weeks | 2.CPD | |||
(Martin et al., 2019) | OUD vs. non-OUD outpatient smokers | 1. Varenicline (2 mg) or placebo | 1. CO + cotinine saliva | 3, 6 months | Smoking abstinence and CPD were not significantly different between OUD vs. NOUD at 3- and 6 months time points. |
N = 137 (drawn from Rohsenow, 2017) | 2. NRT (patch 7 mg, 14 mg or 21 mg) or placebo 12 weeks | 2. CO at 6 months, and urine test, CPD, self-report abstinence for drugs | |||
(Cooperman et al., 2018) | Methadone-maintained Smokers | 1. IMB + NRT | 1. CO abstinence | 3, 6 months | The interventional group reported less CPD at 3 and 6months compared to the control. |
N = 83 | 2. Referral to Quitline (Control) 3 months | 2. CPD, self-report quit attempts, number of days of abstinence | IMB + NRT was not more effective than placebo for abstinence at 3 or 6 months. | ||
(Mooney et al., 2008) | Buprenorphine-maintained smokers | 1. Bupropion (300 mg) | 1. Combined nicotine and illicit drugs abstinence (CO and urine). | 2, 4, 6, 8, and 10 weeks | Bupropion was not more effective than placebo for smoking and opioids abstinence. |
N = 40 | 2. Placebo 10 weeks |
2. Nicotine and Opioid WD |
Abbreviations: CO= carbon monoxide, CBT=cognitive behavioral therapy; CPD=cigarettes per day; FTND= Fagerstrom Test for Nicotine Dependence, FU=follow-up; IMB=model-based intervention, NRT=nicotine replacement therapy; WD=withdrawal.
In a recent study, Hall and colleagues compared the effect of combined extended NRT with cognitive-behavioral therapy (CBT) and motivational intervention to standard control (counseling only) in buprenorphine-maintained smokers. Combined therapy showed only short-term efficacy for abstinence. However, it was superior to control treatment in providing motivation to quit smoking (Hall et al., 2018). Their findings corroborate previous studies indicating that smoking cessation approaches in opioid-maintained patients are only modestly efficacious in the first months of intervention. For example, one study in methadone-maintained smokers found a modest abstinence rate of 10.5 % and reduced daily cigarette consumption in varenicline-treated subjects compared to placebo. Abstinence in this study lasted for only three months, and it was not sustained beyond the intervention period (Nahvi et al., 2014). Varenicline displayed higher efficacy than NRT in promoting abstinence 3 months after treatment initiation in SUD patients (Rohsenow et al., 2017). Still, at the 6 months timepoint, cessation rates were similar between varenicline, NRT, and placebo (Stein et al., 2013). Similarly, another study evaluating the efficacy of combined CBT and NRT in methadone-treated smokers found a similar low, albeit significant abstinence rate (10 %) at three months of therapy that was no longer significantly different than placebo after treatment discontinuation (Reid et al., 2008). A study by Martin and colleagues provides a possible explanation for the lack of long-term efficacy of smoking cessation aids in OUD smokers. They found that smoking relapse may be partly attributable to inadequate medication and treatment adherence, decreased tolerance to physical discomfort, and escalated drug use while receiving varenicline compared to smokers with other SUDs (Martin et al., 2019).
Recent trials have focused on implementing individualized therapies in an attempt to achieve long-term tobacco abstinence among individuals with OUD. Cooperman et al. compared two interventions to improve smoking cessation outcomes. One consisted of eight sessions of Intervention-Motivation-Behavioral (IMB) model-based skills combined with NRT or a referral to a telephone-based service for follow-up as a control. Only a small subset of the IMB group achieved abstinence during the 6 months follow-up. Similar to other treatment strategies, IMB reduced the daily cigarette consumption, but the effects were short-lived (Cooperman et al., 2018).
Besides pharmacotherapies targeting nAChRs, other drug modalities have been explored as alternatives to improve tobacco cessation rates among OUD smokers. For example, individuals under a buprenorphine maintenance program underwent a 10-week regimen of bupropion treatment or placebo; participants also received contingency management to abstain from tobacco and illicit drugs during the intervention period. Bupropion was not well-tolerated and led to low adherence to treatment. Moreover, smoking and opioid abstinence rates were similar between the experimental and control groups (Mooney et al., 2008). In another study, however, patients not allowed to smoke during treatment of opioid withdrawal using low-dose naltrexone and/or clonidine showed enhanced abstinence from cigarettes, but not from opioids, at a 1-week follow-up (Mannelli et al., 2013). Using a laboratory-based approach, it has been found that the severity of opioid craving and acute withdrawal correlates with ND and negative affect (Streck et al., 2020). This latter study suggests that opioid-maintained smokers may positively respond to a national nicotine reduction policy, such as increased taxation, for reducing smoking-related consequences.
Based on these clinical trials, it can be concluded that current pharmacotherapeutic approaches approved to treat nicotine and opioid addiction that target nAChR or MOR independently are of limited effectiveness in managing polydrug abuse. Further investigations designed a priori to identify unique mechanisms dictating nicotine and opioid co-abuse may lead to efficacious treatment for co-morbid ND and OUD. Increased coordinated translational efforts, spanning model systems to clinical trials, are needed for the development of individualized therapies to improve both smoking cessation and opioid discontinuation rates.
8. Summary
In summary, the evidence collected above indicates that nicotine-opioid interactions exist in reward, antinociception, and respiration. The rewarding effects of nicotine are moderated by manipulation of opioid systems, and nicotine amplifies opioid reward in CPP and self-administration models. Nicotine additionally produces opioid system-mediated antinociception and enhances the acute antinociceptive effect of opioids via nAChR activation. These interactions extend to respiration, where nicotine and other nAChR agonists may ameliorate opioid-induced respiratory depression. Conversely, however, nicotine alone generally produces transient respiratory depression, an effect modulated by opioid-receptors. Direct examination of these opioid-nicotine interactions at the cellular level indicates that nicotinic and opioidergic systems work bidirectionally and explain, at least in part, the behavioral and physiological effects observed with co-administration. Variants of genes encoding opioid and nicotinic receptors also underlie ND and OUD comorbidity, with the behavior observed in preclinical rodent models generally mimicking that found in human models. Despite this confluence of preclinical and clinical results, there is limited success in the treatment of nicotine-opioid co-use in clinical trials. Future research should delve into nicotine-opioid interactions further, considering their intricate cellular- and behavioral-level interconnectivity. Clinical trials that target nicotine-opioid co-use need to be continued, especially those intended to improve smoking cessation success among individuals with OUD.
Funding
This work was supported by the following NIH grants: R01DA044311 for JRT and R01DA044311-S1 for LC, JRT PI, R01DA0570 for MTB and JRT, and T32DA035200 for SM.
Footnotes
Declaration of Competing Interest
The authors report no declarations of interest.
References
- Al-Hasani R, McCall JG, Bruchas MR, 2013. Exposure to chronic mild stress prevents kappa opioid-mediated reinstatement of cocaine and nicotine place preference. Front. Pharmacol 4 10.3389/fphar.2013.00096, 96–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Almeida LE, et al. , 2000. The opioid antagonist naltrexone inhibits activity and alters expression of alpha7 and alpha4beta2 nicotinic receptors in hippocampal neurons: implications for smoking cessation programs. Neuropharmacology 39, 2740–2755. 10.1016/s0028-3908(00)00157-x. [DOI] [PubMed] [Google Scholar]
- Amos CI, et al. , 2008. Genome-wide association scan of tag SNPs identifies a susceptibility locus for lung cancer at 15q25.1. Nat. Genet 40, 616–622. 10.1038/ng.109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arias A, Feinn R, Kranzler HR, 2006. Association of an Asn40Asp (A118G) polymorphism in the mu-opioid receptor gene with substance dependence: a meta-analysis. Drug Alcohol Depend. 83, 262–268. [DOI] [PubMed] [Google Scholar]
- Baker TB, et al. , 2009. Human neuronal acetylcholine receptor A5-A3-B4 haplotypes are associated with multiple nicotine dependence phenotypes. Nicotine Tob. Res 11, 785–796. 10.1093/ntr/ntp064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barreto de Moura F, Withey SL, Bergman J, 2019. Enhancement of opioid antinociception by nicotine. J. Pharmacol. Exp. Ther 371, 624–632. 10.1124/jpet.119.261438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bart G., et al. , 2004. Substantial attributable risk related to a functional mu-opioid receptor gene polymorphism in association with heroin addiction in central Sweden. Mol. Psychiatry 9, 547–549. 10.1038/sj.mp.4001504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bart G., et al. , 2005. Increased attributable risk related to a functional mu-opioid receptor gene polymorphism in association with alcohol dependence in central Sweden. Neuropsychopharmacology 30, 417–422. [DOI] [PubMed] [Google Scholar]
- Bernardi RE, et al. , 2016. A gene-by-sex interaction for nicotine reward: evidence from humanized mice and epidemiology. Transl. Psychiatry 6, e861. 10.1038/tp.2016.132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Berrendero F, Kieffer BL, Maldonado R, 2002. Attenuation of nicotine-induced antinociception, rewarding effects, and dependence in mu-opioid receptor knock-out mice. J. Neurosci 22, 10935–10940. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Berrendero F., et al. , 2005. Nicotine-induced antinociception, rewarding effects, and physical dependence are decreased in mice lacking the preproenkephalin gene. J. Neurosci 25 (5), 1103–1112. 10.1523/jneurosci.3008-04.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Berrendero F., et al. , 2012. Influence of δ-Opioid Receptors in the Behavioral Effects of Nicotine. Neuropsychopharmacology 37 (10), 2332–2344. 10.1038/npp.2012.88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Biala G, Budzynska B, 2006. Reinstatement of nicotine-conditioned place preference by drug priming: effects of calcium channel antagonists. Eur. J. Pharmacol 537 (1–3), 85–93. 10.1016/j.ejphar.2006.03.017. [DOI] [PubMed] [Google Scholar]
- Bierut LJ, et al. , 2008. Variants in nicotinic receptors and risk for nicotine dependence. Am. J. Psychiatry 165, 1163–1171. 10.1176/appi.ajp.2008.07111711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bloom AJ, 2019. Mouse strain-specific acute respiratory effects of nicotine unrelated to nicotine metabolism. Toxicol. Mech. Methods 29, 542–548. 10.1080/15376516.2019.1628141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bond C., et al. , 1998. Single-nucleotide polymorphism in the human mu opioid receptor gene alters beta-endorphin binding and activity: possible implications for opiate addiction. Proc Natl Acad Sci U S A 95, 9608–9613. 10.1073/pnas.95.16.9608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brami-Cherrier K., et al. , 2007. Glutamate induces histone H3 phosphorylation but not acetylation in striatal neurons: role of mitogen- and stress-activated kinase-1. J Neurochem 101 (3), 697–708. 10.1111/j.1471-4159.2006.04352. [DOI] [PubMed] [Google Scholar]
- Britt JP, McGehee DS, 2008. Presynaptic opioid and nicotinic receptor modulation of dopamine overflow in the nucleus accumbens. J. Neurosci 28, 1672–1681. 10.1523/JNEUROSCI.4275-07.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brynildsen JK, Blendy JA, 2021. Linking the CHRNA5 SNP to drug abuse liability: from circuitry to cellular mechanisms. Neuropharmacology 186, 108480. 10.1016/j.neuropharm.2021.108480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen LS, et al. , 2012. Interplay of genetic risk factors (CHRNA5-CHRNA3-CHRNB4) and cessation treatments in smoking cessation success. Am. J. Psychiatry 169, 735–742. 10.1176/appi.ajp.2012.11101545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen LS, et al. , 2020. Genetic variant in CHRNA5 and response to Varenicline and combination nicotine replacement in a randomized placebo-controlled trial. Clin. Pharmacol. Ther 108, 1315–1325. 10.1002/cpt.1971. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen RH, Sarnecki C, Blenis J, 1992. Nuclear localization and regulation of erk- and rsk-encoded protein kinases. Mol Cell Biol 12 (3), 915–927. 10.1128/mcb.12.3.915-927.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cippitelli A., et al. , 2016. A key role for the N/OFQ-NOP receptor system in modulating nicotine taking in a model of nicotine and alcohol co-administration. Sci. Rep 6, 26594. 10.1038/srep26594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Clarke JG, Stein MD, McGarry KA, Gogineni A, 2001. Interest in smoking cessation among injection drug users. Am. J. Addict 10, 159–166. 10.1080/105504901750227804. [DOI] [PubMed] [Google Scholar]
- Cooperman NA, Lu SE, Richter KP, Bernstein SL, Williams JM, 2018. Pilot Study of a Tailored Smoking Cessation Intervention for Individuals in Treatment for Opioid Dependence. Nicotine Tob. Res 20, 1152–1156. 10.1093/ntr/ntx189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Corrigall WA, Coen KM, 1991. Opiate antagonists reduce cocaine but not nicotine self-administration. Psychopharmacology 104 (2), 167–170. 10.1007/BF02244173. [DOI] [PubMed] [Google Scholar]
- Creamer MR, et al. , 2019. Tobacco Product Use and Cessation Indicators Among Adults - United States, 2018. MMWR Morb. Mortal. Wkly. Rep 68, 1013–1019. 10.15585/mmwr.mm6845a2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Creekmore FM, Lugo RA, Weiland KJ, 2004. Postoperative opiate analgesia requirements of smokers and nonsmokers. Ann. Pharmacother 38, 949–953. 10.1345/aph.1D580. [DOI] [PubMed] [Google Scholar]
- Curtis K., et al. , 2017. Increased habenular connectivity in opioid users is associated with an alphas subunit nicotinic receptor genetic variant. Am. J. Addict 26, 751–759. 10.1111/ajad.12607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dandrea KE, Cotten JF, 2021. A comparison of breathing stimulants for reversal of synthetic opioid-induced respiratory depression in conscious rats. J. Pharmacol. Exp. Ther 378 (2), 146–156. 10.1124/jpet.121.000675.JPET-AR-2021-000675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- DeNoble VJ, Mele PC, 2006. Intravenous nicotine self-administration in rats: effects of mecamylamine, hexamethonium and naloxone. Psychopharmacology 184 (3), 266–272. 10.1007/s00213-005-0054-z. [DOI] [PubMed] [Google Scholar]
- Di Chiara G, Imperato A, 1988. Drugs abused by humans preferentially increase synaptic dopamine concentrations in the mesolimbic system of freely moving rats. Proc. Nat.l Acad. Sci. U.S.A 85, 5274–5278. 10.1073/pnas.85.14.5274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ditre JW, Heckman BW, Zale EL, Kosiba JD, Maisto SA, 2016. Acute analgesic effects of nicotine and tobacco in humans: a meta-analysis. Pain 157, 1373–1381. 10.1097/j.pain.0000000000000572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Domino EF, Hirasawa-Fujita M, 2019. Tobacco Smoking and Brain Endogenous Opioid Release: More Than Nicotine Alone. Nicotine Tob. Res 21, 772–777. 10.1093/ntr/nty039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Domino EF, Hirasawa-Fujita M, Ni L, Guthrie SK, Zubieta JK, 2015. Regional brain [(11)C]carfentanil binding following tobacco smoking. Prog. Neuropsychopharmacol. Biol. Psych 59, 100–104. 10.1016/j.pnpbp.2015.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Erlich PM, et al. , 2010. Nicotinic acetylcholine receptor genes on chromosome 15q25.1 are associated with nicotine and opioid dependence severity. Hum. Genet 128, 491–499. 10.1007/s00439-010-0876-6. [DOI] [PubMed] [Google Scholar]
- Exley R, Clements MA, Hartung H, McIntosh JM, Cragg SJ, 2008. α6-containing nicotinic acetylcholine receptors dominate the nicotine control of dopamine neurotransmission in nucleus accumbens. Neuropsychopharmacology 33, 2158–2166. 10.1038/sj.npp.1301617. [DOI] [PubMed] [Google Scholar]
- Faure P, Tolu S, Valverde S, Naudé J, 2014. Role of nicotinic acetylcholine receptors in regulating dopamine neuron activity. Neuroscience 282, 86–100. 10.1016/j.neuroscience.2014.05.040. [DOI] [PubMed] [Google Scholar]
- Feng B., et al. , 2011. Blocking α4β2 and α7 nicotinic acetylcholine receptors inhibits the reinstatement of morphine-induced CPP by drug priming in mice. Behav. Brain Res 220, 100–105. 10.1016/j.bbr.2011.01.040. [DOI] [PubMed] [Google Scholar]
- Fisher ML, LeMalefant RM, Zhou L, Huang G, Turner JR, 2017. Distinct roles of CREB within the ventral and dorsal Hippocampus in mediating nicotine withdrawal phenotypes. Neuropsychopharmacology 42, 1599–1609. 10.1038/npp.2016.257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fowler CD, Kenny PJ, 2014. Nicotine aversion: neurobiological mechanisms and relevance to tobacco dependence vulnerability. Neuropharmacology 76 Pt B, 533–544. 10.1016/j.neuropharm.2013.09.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fowler CD, Tuesta L, Kenny PJ, 2013. Role of alpha5* nicotinic acetylcholine receptors in the effects of acute and chronic nicotine treatment on brain reward function in mice. Psychopharmacology (Berl.). 10.1007/s00213-013-3235-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Funk D, Coen K, Lê AD, 2014. The role of kappa opioid receptors in stress-induced reinstatement of alcohol seeking in rats. Brain Behav. 4, 356–367. 10.1002/brb3.222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Galeote L, Kieffer BL, Maldonado R, Berrendero F, 2006. Mu-opioid receptors are involved in the tolerance to nicotine antinociception. J. Neurochem 97, 416–423. 10.1111/j.1471-4159.2006.03751.x. [DOI] [PubMed] [Google Scholar]
- Galeote L, Berrendero F, Andreea Bura S, Zimmer A, Maldonado R, 2008. Prodynorphin gene disruption increases the sensitivity to nicotine self-administration in mice. Int. J. Neuropsychopharmacol 12 (5), 615–625. 10.1017/S1461145708009450. [DOI] [PubMed] [Google Scholar]
- Goldman D, Oroszi G, Ducci F, 2005. The genetics of addictions: uncovering the genes. Nat. Rev. Genet. 6, 521–532. 10.1038/nrg1635. [DOI] [PubMed] [Google Scholar]
- Goto Y, Grace AA, 2008. Limbic and cortical information processing in the nucleus accumbens. Trends Neurosci. 31, 552–558. 10.1016/j.tins.2008.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grella SL, Funk D, Coen K, Li Z, Lê AD, 2014, May 15. Role of the kappa-opioid receptor system in stress-induced reinstatement of nicotine seeking in rats. Behav. Brain Res 265, 188–197. 10.1016/j.bbr.2014.02.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Haas AL, et al. , 2008. Cigarette smoking in opioid-using patients presenting for hospital-based medical services. Am. J. Addict 17, 65–69. 10.1080/10550490701756112. [DOI] [PubMed] [Google Scholar]
- Hall BJ, Pearson LS, Terry AV, Buccafusco JJ, 2011. The use-dependent, nicotinic antagonist BTMPS reduces the adverse consequences of morphine self-administration in rats in an abstinence model of drug seeking. Neuropharmacology 61, 798–806. 10.1016/j.neuropharm.2011.05.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hall SM, et al. , 2018. Cigarette smoking cessation intervention for buprenorphine treatment patients. Nicotine Tob. Res 20, 628–635. 10.1093/ntr/ntx113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Havemann U, Kuschinsky K, 1981. Further characterization of opioid receptors in the striatum mediating muscular rigidity in rats. Naunyn Schmiedebergs Arch. Pharmacol 317, 321–325. 10.1007/BF00501313. [DOI] [PubMed] [Google Scholar]
- Hjelmstad GO, Fields HL, 2003. Kappa opioid receptor activation in the nucleus accumbens inhibits glutamate and GABA release through different mechanisms. J. Neurophysiol 89, 2389–2395. 10.1152/jn.01115.2002. [DOI] [PubMed] [Google Scholar]
- Howell LL, 1995. Effects of caffeine on ventilation during acute and chronic nicotine administration in rhesus monkeys. J. Pharmacol. Exp. Ther 273, 1085–1094. [PubMed] [Google Scholar]
- Hser YI, McCarthy WJ, Anglin MD, 1994. Tobacco use as a distal predictor of mortality among long-term narcotics addicts. Prev. Med 23, 61–69. 10.1006/pmed.1994.1009. [DOI] [PubMed] [Google Scholar]
- Hulihan-Giblin BA, Lumpkin MD, Kellar KJ, 1990. Effects of chronic administration of nicotine on prolactin release in the rat: inactivation of prolactin response by repeated injections of nicotine. J. Pharmacol. Exp. Ther 252, 21–25. [PubMed] [Google Scholar]
- Hurt RD, et al. , 1996. Mortality following inpatient addictions treatment. Role of tobacco use in a community-based cohort. JAMA 275, 1097–1103. 10.1001/jama.275.14.1097. [DOI] [PubMed] [Google Scholar]
- Ide S., et al. , 2004. Gene polymorphisms of the mu opioid receptor in methamphetamine abusers. Ann. N. Y. Acad. Sci 1025, 316–324. [DOI] [PubMed] [Google Scholar]
- Imam MZ, Kuo A, Smith MT, 2020. Countering opioid-induced respiratory depression by non-opioids that are respiratory stimulants. F1000Res 9. 10.12688/f1000research.21738.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ismayilova N, Shoaib M, 2010. Alteration of intravenous nicotine self-administration by opioid receptor agonist and antagonists in rats. Psychopharmacology 209 (2), 211–220. 10.1007/s00213-010-1845-4. [DOI] [PubMed] [Google Scholar]
- Jackson KJ, Carroll FI, Negus SS, Damaj MI, 2010. Effect of the selective kappa-opioid receptor antagonist JDTic on nicotine antinociception, reward, and withdrawal in the mouse. Psychopharmacology 209 (2), 285–294. 10.1007/s00213-010-1803-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jackson KJ, McLaughlin JP, Carroll FI, Damaj MI, 2013. Effects of the kappa opioid receptor antagonist, norbinaltorphimine, on stress and drug-induced reinstatement of nicotine-conditioned place preference in mice. Psychopharmacology (Berl) 226 (4), 763–768. 10.1007/s00213-012-2716-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jensen KP, et al. , 2015. A CHRNA5 smoking risk variant decreases the aversive effects of nicotine in humans. Neuropsychopharmacology 40, 2813–2821. 10.1038/npp.2015.131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jolley CJ, Bell J, Rafferty GF, Moxham J, Strang J, 2015. Understanding heroin overdose: a study of the acute respiratory depressant effects of injected pharmaceutical heroin. PLoS One 10. 10.1371/journal.pone.0140995 e0140995–e0140995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jones RA, 1987. Cigarettes, respiratory rate, and the relaxation paradox. Int. J. Addict. 22, 803–809. 10.3109/10826088709027460. [DOI] [PubMed] [Google Scholar]
- Jones JD, et al. , 2020. A randomized, double-blind, placebo-controlled study of the kappa opioid receptor antagonist, CERC-501, in a human laboratory model of smoking behavior. Addict. Biol 25, e12799 10.1111/adb.12799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kalman D, Morissette SB, George TP, 2005. Co-morbidity of smoking in patients with psychiatric and substance use disorders. Am. J. Addict 14, 106–123. 10.1080/10550490590924728. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kamerling SG, Wettstein JG, Sloan JW, Su TP, Martin WR, 1982. Interaction between nicotine and endogenous opioid mechanisms in the unanesthetized dog. Pharmacol. Biochem. Behav 17, 733–740. 10.1016/0091-3057(82)90355-0. [DOI] [PubMed] [Google Scholar]
- Kapur S, Sharad S, Singh RA, Gupta AK, 2007. A118g polymorphism in mu opioid receptor gene (oprm1): association with opiate addiction in subjects of Indian origin. J. Integr. Neurosci 6, 511–522. 10.1142/s0219635207001635. [DOI] [PubMed] [Google Scholar]
- Kathuria H., et al. , 2019. Perceived barriers to quitting cigarettes among hospitalized smokers with substance use disorders: a mixed methods study. Addict. Behav 95, 41–48. 10.1016/j.addbeh.2019.02.017. [DOI] [PubMed] [Google Scholar]
- Kenny PJ, Markou A, 2001. Neurobiology of the nicotine withdrawal syndrome. Pharmacol. Biochem. Behav 70, 531–549. 10.1016/s0091-3057(01)00651-7. [DOI] [PubMed] [Google Scholar]
- Kim DH, et al. , 2017. Smoking may increase postoperative opioid consumption in patients who underwent distal gastrectomy with gastroduodenostomy for early stomach Cancer: a retrospective analysis. Clin. J. Pain 33, 905–911. 10.1097/ajp.0000000000000472. [DOI] [PubMed] [Google Scholar]
- Knoll AT, Carlezon WA Jr., 2010. Dynorphin, stress, and depression. Brain Res. 1314, 56–73. 10.1016/j.brainres.2009.09.074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Krause D., et al. , 2018. The impact of the opioid antagonist naloxone on experimentally induced craving in nicotine-dependent individuals. Eur. Addict. Res 24, 255–265. 10.1159/000494346. [DOI] [PubMed] [Google Scholar]
- Kruyer A, Chioma VC, Kalivas PW, 2020. The opioid-addicted tetrapartite synapse. Biol. Psychiatry 87, 34–43. 10.1016/j.biopsych.2019.05.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kuwabara H, Heishman SJ, Brasic JR, Contoreggi C, et al. , 2014. Mu Opioid Receptor Binding Correlates with Nicotine Dependence and Reward in Smokers. Plos One 9 (12), e113694. 10.1371/journal.pone.0113694. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kwon MS, et al. , 2008. The effect of formalin pretreatment on nicotine-induced antinociceptive effect: the role of mu-opioid receptor in the hippocampus. Neuroscience 154, 415–423. 10.1016/j.neuroscience.2008.03.053. [DOI] [PubMed] [Google Scholar]
- Lee LY, Morton RF, Kou YR, 1990. Acute effects of cigarette smoke on breathing in rats: vagal and nonvagal mechanisms. J. Appl. Physiol 68 (1985), 955–961. 10.1152/jappl.1990.68.3.955. [DOI] [PubMed] [Google Scholar]
- Lee JH, et al. , 2020. Dopaminergic regulation of nucleus accumbens cholinergic interneurons demarcates susceptibility to cocaine addiction. Biol. Psychiatry 88, 746–757. 10.1016/j.biopsych.2020.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lenz B, et al. , 2010. Smoking behaviour is associated with expression and phosphorylation of CREB in human buffy coat. The international journal of neuropsychopharmacology / official scientific journal of the Collegium Internationale Neuropsychopharmacologicum 13, 207–215. 10.1017/S1461145709991052. [DOI] [PubMed] [Google Scholar]
- Lenz B., et al. , 2012. ERK1/2 protein and mRNA levels in human blood are linked to smoking behavior. Addict Bio 17 (6), 1026–1035. 10.1111/j.1369-1600.2010.00264.x. [DOI] [PubMed] [Google Scholar]
- Lessov CN, et al. , 2004. Defining nicotine dependence for genetic research: evidence from Australian twins. Psychol. Med 34, 865–879. 10.1017/s0033291703001582. [DOI] [PubMed] [Google Scholar]
- Liu X, Jernigan C, 2011. Activation of the opioid μ1, but not δ or κ, receptors is required for nicotine reinforcement in a rat model of drug self-administration. Prog. Neuropsychopharmacol. Biol. Psychiatry 35 (1), 146–153. 10.1016/j.pnpbp.2010.10.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu X., et al. , 2009. Naltrexone attenuation of conditioned but not primary reinforcement of nicotine in rats. Psychopharmacology 202 (4), 589–598. 10.1007/s00213-008-1335-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Loney GC, King CP, Meyer PJ, 2021. Systemic nicotine enhances opioid self-administration and modulates the formation of opioid-associated memories partly through actions within the insular cortex. Sci. Rep 11 10.1038/s41598-021-81955-5, 3321–3321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mague SD, et al. , 2009. Mouse model of OPRM1 (A118G) polymorphism has sex-specific effects on drug-mediated behavior. Proc Natl Acad Sci U S A 106, 10847–10852. 10.1073/pnas.0901800106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mamaligas AA, Cai Y, Ford CP, 2016. Nicotinic and opioid receptor regulation of striatal dopamine D2-receptor mediated transmission. Sci. Rep 6, 37834. 10.1038/srep37834. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mannelli P, Wu LT, Peindl KS, Gorelick DA, 2013. Smoking and opioid detoxification: behavioral changes and response to treatment. Nicotine Tob. Res 15, 1705–1713. 10.1093/ntr/ntt046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mansvelder HD, McGehee DS, 2002. Cellular and synaptic mechanisms of nicotine addiction. J. Neurobiol 53, 606–617. 10.1002/neu.10148. [DOI] [PubMed] [Google Scholar]
- Marks MJ, Stitzel JA, Collins AC, 1989. Genetic influences on nicotine responses. Pharmacol. Biochem. Behav 33, 667–678. 10.1016/0091-3057(89)90406-1. [DOI] [PubMed] [Google Scholar]
- Martin RA, Rohsenow DJ, Tidey JW, 2019. Smokers with opioid use disorder may have worse drug use outcomes after varenicline than nicotine replacement. J. Subst. Abuse Treat. 104, 22–27. 10.1016/j.jsat.2019.06.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Matthes HW, et al. , 1996. Loss of morphine-induced analgesia, reward effect and withdrawal symptoms in mice lacking the mu-opioid-receptor gene. Nature 383, 819–823. 10.1038/383819a0. [DOI] [PubMed] [Google Scholar]
- McCrimmon DR, Mitchell GS, Alheid GF, 2008. Neurochemistry of respiratory control. Respir. Physiol. Neurobiol 164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McMillan DM, Tyndale RF, 2017. Inducing rat brain CYP2D with nicotine increases the rate of codeine tolerance; predicting the rate of tolerance from acute analgesic response. Biochem. Pharmacol 145, 158–168. 10.1016/j.bcp.2017.08.015. [DOI] [PubMed] [Google Scholar]
- Miller ME, Sigmon SC, 2015. Are pharmacotherapies ineffective in opioid-dependent smokers? Reflections on the scientific literature and future directions. Nicotine Tob. Res 17, 955–959. 10.1093/ntr/ntv030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mooney ME, et al. , 2008. Preliminary study of buprenorphine and bupropion for opioid-dependent smokers. Am. J. Addict 17, 287–292. 10.1080/10550490802138814. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nahvi S, Ning Y, Segal KS, Richter KP, Arnsten JH, 2014. Varenicline efficacy and safety among methadone maintained smokers: a randomized placebo-controlled trial. Addiction 109, 1554–1563. 10.1111/add.12631. [DOI] [PMC free article] [PubMed] [Google Scholar]
- National Academies of Sciences, E, et al. , 2017. In: Phillips JK, Ford MA, Bonnie RJ (Eds.), Pain Management and the Opioid Epidemic: Balancing Societal and Individual Benefits and Risks of Prescription Opioid Use. National Academies Press, US. Copyright 2017 by the National Academy of Sciences. All rights reserved. [PubMed] [Google Scholar]
- Nestler EJ, Kenny PJ, Russo SJ, Shaefer A, 2020. Molecular Neuropharmacology: a Foundation for Clinical Neuroscience, 4th edn. McGraw Hill. [Google Scholar]
- Nisell M, Nomikos GG, Svensson TH, 1994. Infusion of nicotine in the ventral tegmental area or the nucleus accumbens of the rat differentially affects accumbal dopamine release. Pharmacol. Toxicol. 75, 348–352. 10.1111/j.1600-0773.1994.tb00373.x. [DOI] [PubMed] [Google Scholar]
- Nuechterlein EB, Ni L, Domino EF, Zubieta JK, 2016. Nicotine-specific and non-specific effects of cigarette smoking on endogenous opioid mechanisms. Prog Neuropsychopharmacol Biol Psychiatry 69, 69–77. 10.1016/j.pnpbp.2016.04.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nygard SK, Hourguettes NJ, Sobczak GG, Carlezon WA, Bruchas MR, 2016. Stress-induced reinstatement of nicotine preference requires Dynorphin/Kappa opioid activity in the basolateral amygdala. J. Neurosci 36 (38), 9937–9948. 10.1523/JNEUROSCI.0953-16.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- O’Donnell J, Gladden RM, Mattson CL, Hunter CT, Davis NL, 2020. Vital Signs: Characteristics of Drug Overdose Deaths Involving Opioids and Stimulants - 24 States and the District of Columbia, January-June 2019. MMWR Morb. Mortal. Wkly. Rep 69, 1189–1197. 10.15585/mmwr.mm6935a1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ossipov MH, Dussor GO, Porreca F, 2010. Central modulation of pain. J. Clin. Invest 120, 3779–3787. 10.1172/jci43766. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Parker MA, Weinberger AH, 2020. Opioid use disorder trends from 2002 to 2017 by cigarette smoking status in the United States. Nicotine Tob. Res 10.1093/ntr/ntaa189. [DOI] [PubMed] [Google Scholar]
- Parker MA, Streck JM, Sigmon SC, 2018. Associations between opioid and nicotine dependence in nationally representative samples of United States adult daily smokers. Drug Alcohol Depend. 186, 167–170. 10.1016/j.drugalcdep.2018.01.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Perkins KA, et al. , 2008. Dopamine and opioid gene variants are associated with increased smoking reward and reinforcement owing to negative mood. Behav. Pharmacol 19, 641–649. 10.1097/FBP.0b013e32830c367c. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pistillo F, Clementi F, Zoli M, Gotti C, 2015. Nicotinic, glutamatergic and dopaminergic synaptic transmission and plasticity in the mesocorticolimbic system: focus on nicotine effects. Prog. Neurobiol 124, 1–27. 10.1016/j.pneurobio.2014.10.002. [DOI] [PubMed] [Google Scholar]
- Plesner K, Jensen HI, Hojsted J, 2016. Smoking history, nicotine dependence and opioid use in patients with chronic non-malignant pain. Acta Anaesthesiol. Scand 60, 988–994. 10.1111/aas.12741. [DOI] [PubMed] [Google Scholar]
- Pomerleau OF, Fertig JB, Seyler LE, Jaffe J, 1983. Neuroendocrine reactivity to nicotine in smokers. Psychopharmacology (Berl.) 81, 61–67. 10.1007/BF00439275. [DOI] [PubMed] [Google Scholar]
- Portugal GS, Wilkinson DS, Turner JR, Blendy JA, Gould TJ, 2012. Developmental effects of acute, chronic, and withdrawal from chronic nicotine on fear conditioning. Neurobiol. Learn. Mem 97, 482–494. 10.1016/j.nlm.2012.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Quach BC, et al. , 2020. Expanding the genetic architecture of nicotine dependence and its shared genetics with multiple traits. Nat. Commun 11, 5562. 10.1038/s41467-020-19265-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ray R., et al. , 2007. Genetic variation in mu-opioid-receptor-interacting proteins and smoking cessation in a nicotine replacement therapy trial. Nicotine Tob. Res. 9, 1237–1241. [DOI] [PubMed] [Google Scholar]
- Ray R., et al. , 2011. Human Mu Opioid Receptor (OPRM1 A118G) polymorphism is associated with brain mu-opioid receptor binding potential in smokers. Proc. Natl. Acad. Sci. U. S.A 108, 9268–9273. 10.1073/pnas.1018699108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Redila VA, Chavkin C, 2008. Stress-induced reinstatement of cocaine seeking is mediated by the kappa opioid system. Psychopharmacology 200, 59–70. 10.1007/s00213-008-1122-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reid MS, et al. , 2008. Smoking cessation treatment in community-based substance abuse rehabilitation programs. J. Subst. Abuse Treat 35, 68–77. 10.1016/j.jsat.2007.08.010. [DOI] [PubMed] [Google Scholar]
- Ren J, Ding X, Greer JJ, 2019. Activating α4β2 nicotinic acetylcholine receptors alleviates fentanyl-induced respiratory depression in rats. Anesthesiology 130, 1017–1031. 10.1097/aln.0000000000002676. [DOI] [PubMed] [Google Scholar]
- Ren J, Ding X, Greer JJ, 2020. Countering opioid-induced respiratory depression in male rats with nicotinic acetylcholine receptor partial agonists varenicline and ABT 594. Anesthesiology (Philadelphia) 132, 1197–1211. 10.1097/ALN.0000000000003128. [DOI] [PubMed] [Google Scholar]
- Rezayof A, Zatali H, Haeri-Rohani A, Zarrindast MR, 2006. Dorsal hippocampal muscarinic and nicotinic receptors are involved in mediating morphine reward. Behav. Brain Res 166, 281–290. 10.1016/j.bbr.2005.08.010. [DOI] [PubMed] [Google Scholar]
- Rezayof A, Nazari-Serenjeh F, Zarrindast M-R, Sepehri H, Delphi L, 2007. Morphine-induced place preference: involvement of cholinergic receptors of the ventral tegmental area. Eur. J. Pharmacol 562, 92–102. 10.1016/j.ejphar.2007.01.081. [DOI] [PubMed] [Google Scholar]
- Richter KP, Choi WS, McCool RM, Harris KJ, Ahluwalia JS, 2004. Smoking cessation services in U.S. Methadone maintenance facilities. Psychiatr. Serv 55, 1258–1264. 10.1176/appi.ps.55.11.1258. [DOI] [PubMed] [Google Scholar]
- Robles CF, et al. , 2014. Effects of kappa opioid receptors on conditioned place aversion and social interaction in males and females. Behav. Brain Res 262, 84–93. 10.1016/j.bbr.2014.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rohsenow DJ, et al. , 2017. Varenicline versus nicotine patch with brief advice for smokers with substance use disorders with or without depression: effects on smoking, substance use and depressive symptoms. Addiction 112, 1808–1820. 10.1111/add.13861. [DOI] [PubMed] [Google Scholar]
- Sakoori K, Murphy NP, 2009. Enhanced nicotine sensitivity in nociceptin/orphanin FQ receptor knockout mice. Neuropharmacology 56 (5), 896–904. 10.1016/j.neuropharm.2009.01.016. [DOI] [PubMed] [Google Scholar]
- Sarginson JE, et al. , 2011. Markers in the 15q24 nicotinic receptor subunit gene cluster (CHRNA5-A3-B4) predict severity of nicotine addiction and response to smoking cessation therapy. Am. J. Med. Genet. B Neuropsychiatr. Genet 156B, 275–284. 10.1002/ajmg.b.31155. [DOI] [PubMed] [Google Scholar]
- Schmitz JM, Grabowski J, Rhoades H, 1994. The effects of high and low doses of methadone on cigarette smoking. Drug Alcohol Depend. 34, 237–242. 10.1016/0376-8716(94)90162-7. [DOI] [PubMed] [Google Scholar]
- Scofield MD, et al. , 2016. The nucleus accumbens: mechanisms of addiction across drug classes reflect the importance of glutamate homeostasis. Pharmacol. Rev 68, 816–871. 10.1124/pr.116.012484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Scott DJ, Domino EF, Heitzeg MM, Koeppe RA, Ni L, Zubieta JK, 2007. Smoking modulation of mu-opioid and dopamine D2 receptor-mediated neurotransmission in humans. Neuropsychopharmacology 32 (2), 450–457. 10.1038/sj.npp.1301238. [DOI] [PubMed] [Google Scholar]
- Shams J., et al. , 2006. Effects of ultra-low doses of nicotine on the expression of morphine-induced conditioned place preference in mice. Behav. Pharmacol 17, 629–635. 10.1097/FBP.0b013e3280102d68. [DOI] [PubMed] [Google Scholar]
- Sherva R., et al. , 2010. Variation in nicotinic acetylcholine receptor genes is associated with multiple substance dependence phenotypes. Neuropsychopharmacology 35, 1921–1931. 10.1038/npp.2010.64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Skrbic R., et al. , 2017. Naloxone antagonizes soman-induced central respiratory depression in rats. Basic Clin. Pharmacol. Toxicol 120, 615–620. 10.1111/bcpt.12745. [DOI] [PubMed] [Google Scholar]
- Sloan JW, Martin WR, Bostwick M, 1989. Mechanisms involved in the respiratory depressant actions of nicotine in anesthetized rats. Pharmacol. Biochem. Behav 34, 559–564. 10.1016/0091-3057(89)90558-3. [DOI] [PubMed] [Google Scholar]
- Smith JS, et al. , 2012. Stress-induced activation of the dynorphin/κ-opioid receptor system in the amygdala potentiates nicotine conditioned place preference. J. Neurosci 32 (4), 1488–1495. 10.1523/JNEUROSCI.2980-11.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stein MD, et al. , 2013. Varenicline for smoking cessation among methadone-maintained smokers: a randomized clinical trial. Drug Alcohol Depend. 133, 486–493. 10.1016/j.drugalcdep.2013.07.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Story J, Stark MJ, 1991. Treating cigarette smoking in methadone maintenance clients. J. Psychoactive Drugs 23, 203–215. 10.1080/02791072.1991.10472237. [DOI] [PubMed] [Google Scholar]
- Strang J, et al. , 2016. Naloxone without the needle – systematic review of candidate routes for non-injectable naloxone for opioid overdose reversal. Drug Alcohol Depend. 163, 16–23. 10.1016/j.drugalcdep.2016.02.042. [DOI] [PubMed] [Google Scholar]
- Streck JM, et al. , 2020. Investigating tobacco withdrawal in response to reduced nicotine cigarettes among smokers with opioid use disorder and other vulnerabilities. Exp. Clin. Psychopharmacol 28, 714–723. 10.1037/pha0000350. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sullivan PF, Kendler KS, 1999. The genetic epidemiology of smoking. Nicotine Tob. Res 1 (Suppl 2), S51–57. 10.1080/14622299050011811 discussion S69-70. [DOI] [PubMed] [Google Scholar]
- Talka R, Salminen O, Whiteaker P, Lukas RJ, Tuominem RK, 2013. Nicotine-morphine interactions at alpha4beta2, alpha7 and alpha3(*) nicotinic acetylcholine receptors. Eur J Pharmacol. 701 (1–3), 57–64. 10.1016/j.ejphar.2013.01.005. [DOI] [PubMed] [Google Scholar]
- Tan EC, Tan CH, Karupathivan U, Yap EP, 2003. Mu opioid receptor gene polymorphisms and heroin dependence in Asian populations. Neuroreport 14, 569–572. 10.1097/00001756-200303240-00008. [DOI] [PubMed] [Google Scholar]
- Tanda G, Di Chiara G, 1998. A dopamine-mu1 opioid link in the rat ventral tegmentum shared by palatable food (Fonzies) and non-psychostimulant drugs of abuse. Eur. J. Neurosci 10, 1179–1187. 10.1046/j.1460-9568.1998.00135.x. [DOI] [PubMed] [Google Scholar]
- Tobaldini G., et al. , 2020. Pain and stress: functional evidence that supra-spinal mechanisms involved in pain-induced analgesia mediate stress-induced analgesia. Behav. Pharmacol 31, 159–167. 10.1097/fbp.0000000000000529. [DOI] [PubMed] [Google Scholar]
- Torralva R, Janowsky A, 2019. Noradrenergic mechanisms in fentanyl-mediated rapid death explain failure of naloxone in the opioid crisis. J. Pharmacol. Exp. Ther 371, 453–475. 10.1124/jpet.119.258566. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Torralva R., et al. , 2020. Fentanyl but not morphine interacts with nonopioid recombinant human neurotransmitter receptors and transporters. J. Pharmacol. Exp. Ther 374, 376–391. 10.1124/jpet.120.265561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Trigo JM, Zimmer A, Maldonado R, 2009. Nicotine anxiogenic and rewarding effects are decreased in mice lacking β-endorphin. Neuropharmacology 56 (8), 1147–1153. 10.1016/j.neuropharm.2009.03.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Turner JR, et al. , 2013a. Divergent functional effects of sazetidine-a and varenicline during nicotine withdrawal. Neuropsychopharmacology 38, 2035–2047. 10.1038/npp.2013.105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Turner JR, et al. , 2013b. Evidence from mouse and man for a role of neuregulin 3 in nicotine dependence. Mol. Psychiatry. 10.1038/mp.2013.104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Umana IC, et al. , 2017. Nicotinic modulation of descending pain control circuitry. Pain 158, 1938–1950. 10.1097/j.pain.0000000000000993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Varga AG, Reid BT, Kieffer BL, Levitt ES, 2019. Differential effects of morphine on a dispersed respiratory network to induce respiratory depression in awake mice. Faseb J. 33 (546), 542–546.542. 10.1096/fasebj.2019.33.1_supplement.546.2. [DOI] [Google Scholar]
- Vihavainen T, Mijatovic J, Piepponen TP, Tuominen RK, Ahtee L, 2006. Effect of morphine on locomotor activity and striatal monoamine metabolism in nicotine-withdrawn mice. Behav. Brain Res 173, 85–93. 10.1016/j.bbr.2006.06.004. [DOI] [PubMed] [Google Scholar]
- Vihavainen T, Piltonen M, Tuominen RK, Korpi ER, Ahtee L, 2008a. Morphine–nicotine interaction in conditioned place preference in mice after chronic nicotine exposure. Eur. J. Pharmacol 587, 169–174. 10.1016/j.ejphar.2008.03.028. [DOI] [PubMed] [Google Scholar]
- Vihavainen T., et al. , 2008b. Chronic nicotine modifies the effects of morphine on extracellular striatal dopamine and ventral tegmental GABA. J. Neurochem 107, 844–854. 10.1111/j.1471-4159.2008.05676.x. [DOI] [PubMed] [Google Scholar]
- Volkow ND, Jones EB, Einstein EB, Wargo EM, 2019. Prevention and treatment of opioid misuse and addiction: a review. JAMA Psych. (Chicago, Ill.) 76, 208–216. 10.1001/jamapsychiatry.2018.3126. [DOI] [PubMed] [Google Scholar]
- Walker DM, Nestler EJ, 2018. Neuroepigenetics and addiction. Handb. Clin. Neurol 148, 747–765. 10.1016/B978-0-444-64076-5.00048-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Walters CL, Cleck JN, Kuo YC, Blendy JA, 2005. Mu-opioid receptor and CREB activation are required for nicotine reward. Neuron (Cambridge, Mass.) 46, 933–943. 10.1016/j.neuron.2005.05.005. [DOI] [PubMed] [Google Scholar]
- Wewers ME, Dhatt RK, Snively TA, Tejwani GA, 1999. The effect of chronic administration of nicotine on antinociception, opioid receptor binding and metenkelphalin levels in rats. Brain Res. 822 (1–2), 107–113. 10.1016/s0006-8993(99)01095-1. [DOI] [PubMed] [Google Scholar]
- Wright VL, et al. , 2019. Inhibition of alpha7 nicotinic receptors in the ventral hippocampus selectively attenuates reinstatement of morphine-conditioned place preference and associated changes in AMPA receptor binding. Addict. Biol 24, 590–603. 10.1111/adb.12624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xing J, Kornhauser JM, Xia Z, Thiele EA, Greenberg ME, 1998. Nerve growth factor activates extracellular signal-regulated kinase and p38 mitogen-activated protein kinase pathways to stimulate CREB serine 133 phosphorylation. Mol Cell Bio 18 (4), 1946–1955. 10.1128/MCB.18.4.1946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Young-Wolff KC, Klebaner D, Weisner C, Von Korff M, Campbell CI, 2017. Smoking status and opioid-related problems and concerns among men and women on chronic opioid therapy. Clin. J. Pain 33, 730–737. 10.1097/AJP.0000000000000461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zarrindast M-R, Faraji N, Rostami P, Sahraei H, Ghoshouni H, 2003. Cross-tolerance between morphine- and nicotine-induced conditioned place preference in mice. Pharmacol. Biochem. Behav 74 (2), 363–369. 10.1016/S0091-3057(02)01002-X. [DOI] [PubMed] [Google Scholar]
- Zarrindast M-R, Fattahi Z, Rostami P, Rezayof A, 2005. Role of the cholinergic system in the rat basolateral amygdala on morphine-induced conditioned place preference. Pharmacol. Biochem. Behav 82, 1–10. 10.1016/j.pbb.2005.02.018. [DOI] [PubMed] [Google Scholar]
- Zhai H, Li Y, Wang X, Lu L, 2008. Drug-induced alterations in the extracellular signal-regulated kinase (ERK) signalling pathway: implications for reinforcement and reinstatement. Cell. Mol. Neurobiol 28, 157–172. 10.1007/s10571-007-9240-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang L, Kendler KS, Chen X, 2006. The mu-opioid receptor gene and smoking initiation and nicotine dependence. Behav. Brain Funct 2, 28. 10.1186/1744-9081-2-28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou W., et al. , 2006. Role of acetylcholine transmission in nucleus accumbens and ventral tegmental area in heroin-seeking induced by conditioned cues. Neuroscience 144, 1209–1218. 10.1016/j.neuroscience.2006.11.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou H., et al. , 2020. Association of OPRM1 functional coding variant with opioid use disorder: a genome-wide association study. JAMA Psychiatry 77, 1072–1080. 10.1001/jamapsychiatry.2020.1206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zuo W., et al. , 2016. Nicotine regulates activity of lateral habenula neurons via presynaptic and postsynaptic mechanisms. Sci. Rep 6, 32937. 10.1038/srep32937. [DOI] [PMC free article] [PubMed] [Google Scholar]