Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Aug 11.
Published in final edited form as: Int Rev Neurobiol. 2020 Oct 6;157:1–29. doi: 10.1016/bs.irn.2020.09.001

Neurobiological aspects of pain in the context of alcohol use disorder

Jessica A Cucinello-Ragland 1, Scott Edwards 1,*
PMCID: PMC8356551  NIHMSID: NIHMS1730639  PMID: 33648668

Abstract

Alcohol is an effective and widely utilized analgesic. However, the chronic use of alcohol can actually facilitate nociceptive sensitivity over time, a condition known as hyperalgesia. Excessive and uncontrollable alcohol drinking is also a hallmark feature of alcohol use disorder (AUD). Both AUD and chronic pain are typically accompanied by negative affective states that may underlie reinforcement mechanisms contributing to AUD maintenance or progression. Frequent utilization of alcohol to relieve pain in individuals suffering from AUD or other chronic pain conditions may thus represent a powerful negative reinforcement construct. This chapter will describe ties between alcohol-mediated pain relief and potential exacerbation of AUD. We describe neurobiological systems engaged in alcohol analgesia as well as systems recruited in the development and maintenance of AUD and hyperalgesia. Although few effective therapies exist for either chronic pain or AUD, the common interaction of these conditions will likely lead the way for promising new discoveries of more effective and even simultaneous treatment of AUD and co-morbid hyperalgesia. An abundance of neurobiological findings from multiple laboratories has implicated a potentiation of central amygdala (CeA) signaling in both pain and AUD, and these data also suggest that attenuation of stress-related systems (including corticotropin-releasing factor, vasopressin, and glucocorticoid receptor activity) would be particularly effective and comprehensive therapeutic strategies targeting the critical intersection of somatic and motivational mechanisms driving AUD, including alcohol-induced hyperalgesia.

1. Alcohol use disorder and chronic pain

1.1. Pain as a central symptom of AUD

Alcohol use disorder (AUD, ranging from mild to severe) is characterized by an escalation of alcohol drinking and gradual emergence of negative affective symptoms (e.g., depression, anxiety) over time (Edwards & Koob, 2010). Such negative emotional states may promote escalated drinking as well as relapse during attempted abstinence periods. Consequently, AUD typically represents a chronic psychiatric condition requiring close management over the patient’s lifespan (Koob & Volkow, 2010). AUD affects roughly 10–13% of the population in the United States at any given time, and up to one-third of adults in the United States will meet criteria for AUD in their lifetime (Grant et al., 2017). In addition to the negative affective symptoms described above, both clinical and preclinical evidence demonstrates that nociceptive hypersensitivity emerges alongside chronic or excessive alcohol exposure (Dina et al., 2000; Edwards, Guerrero, et al., 2012; Edwards, Vendruscolo, et al., 2012; Edwards, Vendruscolo, Gilpin, Wojnar, & Witkiewitz, 2020; Fu et al., 2015; Gatch & Lal, 1999; Jochum, Boettger, Burkhardt, Juckel, & Bar, 2010). Facilitation of pain by alcohol is evident in that the nocifensive response to painful (noxious) stimuli is enhanced (termed hyperalgesia) and also in that innocuous stimuli (normally non-painful) become noxious (termed allodynia). Consequently, pain represents a negative subjective experience that can have a powerful influence on reward and reinforcement mechanisms, possibly facilitating the transition to substance use disorders in vulnerable individuals (McGinn & Edwards, 2016).

1.2. The negative affective dimension of chronic pain

Chronic pain affects approximately 20% of adults worldwide (Goldberg & McGee, 2011), a number that will likely increase over the next several decades given the aging global population. In the United States alone, over 50 million individuals report chronic pain at a cost of over $600 billion in healthcare expenses and lost productivity (Dahlhamer et al., 2018). While these numbers appear to eclipse the impact of other chronic diseases such as heart disease and cancer, the devastating impact of chronic pain is related more to its effects on morbidity (compared to mortality), especially with regard to the potentiation of negative affect in association with unrelieved pain. Negative affective mood states (anxiety, sadness, anger, and irritability) are considered highly aversive psychodynamic constructs that are distinguishing features of anxiety and depression (Watson & Tellegen, 1985). In general, negative affective disorders represents an umbrella term encompassing a number of mood disorders, including neuroticism, major depressive disorder, general anxiety disorder, and panic disorder.

Chronic pain and negative affective disorders have an alarming rate of comorbidity, likely because of the bidirectional relationship between the two (Elman, Borsook, & Volkow, 2013). Indeed, patients with chronic pain and high negative affect report higher pain severity and pain interference compared to patients with only one of the two disorders (Arnow et al., 2006; Naliboff, Chang, Munakata, & Mayer, 2000). In both children and adults, multiple types of pain disorders are associated with increased likelihood of a positive diagnosis for negative affective disorders, with higher pain ratings elevating the likelihood of affective disorder diagnoses (Campo et al., 2004; Means-Christensen, Roy-Byrne, Sherbourne, Craske, & Stein, 2008). It is important to note that the prevalence of comorbid chronic pain and negative affect varies depending on the type of negative affective disorder. For example, 35–85% of chronic pain patients experience comorbid depression, while 16–60% experience comorbid anxiety disorders (Bair, Robinson, Katon, & Kroenke, 2003; Fishbain, Cutler, Rosomoff, & Rosomoff, 1998; Fishbain, Goldberg, Meagher, Steele, & Rosomoff, 1986; Lee et al., 2018; McWilliams, Cox, & Enns, 2003; Williams et al., 2003).

1.3. Chronic pain and depression

Because chronic pain has a higher incidence of comorbidity with depression than anxiety, and because depression is a major global health burden (Liu et al., 2019), much of the research into the relationship between chronic pain and negative affect has revolved around depressive disorders. Clinical studies have investigated the development of depression across a number of different chronic pain conditions (Bair et al., 2003), including low back pain (France, Houpt, Skott, Krishnan, & Varia, 1986), orofacial pain (Cox et al., 2016; Feinmann, 1983), and pelvic pain (Magni, Salmi, de Leo, & Ceola, 1984; Siqueira-Campos, Da Luz, de Deus, Martinez, & Conde, 2019). Additionally, both clinical (Aguera-Ortiz, Failde, Mico, Cervilla, & Lopez-Ibor, 2011; Lee et al., 2009; von Knorring, Perris, Eisemann, Eriksson, & Perris, 1983) and preclinical (Goncalves et al., 2008; Kontinen, Kauppila, Paananen, Pertovaara, & Kalso, 1999; Schwartz et al., 2014) studies have found that chronic pain precedes and induces symptoms of depression in a variety of chronic pain conditions and models.

1.4. Chronic pain and anxiety

Although most of the research on chronic pain and negative affect has traditionally revolved around depression, there has been a rapid growth in research regarding comorbid chronic pain and anxiety disorders (Asmundson & Katz, 2009). While clinical reports of comorbid chronic pain and anxiety are highly variable across populations, chronic pain-induced anxiety-like behavior is readily observed in both neuropathic and non-neuropathic animal models of pain (Bahaaddini, Khatamsaz, Esmaeili-Mahani, Abbasnejad, & Raoof, 2016; Dimitrov, Tsuda, Cameron, & Usdin, 2014; Ji, Fu, Ruppert, & Neugebauer, 2007; Matsuzawa-Yanagida et al., 2008; Narita et al., 2006). Additionally, chronic pain can generate pain-related anxiety in humans, in which the patient experiences fear and anxiety in the anticipation of experiencing future pain (McCracken & Dhingra, 2002; McCracken, Zayfert, & Gross, 1992).

1.5. Does hyperalgesia and associated negative affect facilitate AUD severity?

An emerging question is whether and how the negative experience of chronic, unrelieved pain contributes to the maintenance of AUD. Hyperalgesia may drive both continuous alcohol drinking and relapse propensity as a powerful negative reinforcement mechanism based on the strong affective valence associated with persistent and unrelieved nociceptive hypersensitivity (Egli, Koob, & Edwards, 2012; Zale, Maisto, & Ditre, 2015). Within this context, negative reinforcement would occur whenever the perceived or real effects of alcohol drinking reduce hyperalgesia symptoms. At the basic science level, evidence for this relationship comes from a recent study where hyperalgesia symptoms in alcohol-dependent animals were attenuated by alcohol self-administration (Roltsch Hellard, Impastato, & Gilpin, 2017). At the clinical level, pain prospectively predicts AUD development (McDermott, Joyner, Hakes, Okey, & Cougle, 2018) and relapse propensity (Witkiewitz et al., 2015). A host of neurobiological and psychosocial mechanisms are shared between chronic pain and alcohol reinforcement (Ditre, Zale, & LaRowe, 2019; LeBlanc, McGinn, Itoga, & Edwards, 2015). As described within this chapter, the dysregulation of such systems may become central in the progression of AUD, while targeting these aberrant processes may represent valuable therapeutic strategies for the treatment of pain in the context of AUD.

2. Analgesic actions of alcohol

2.1. History of alcohol as an analgesic

For centuries, people have consumed alcohol for its analgesic properties, dating as far back as ancient Egypt and Greece (Crocq, 2007; McGovern, Mirzoian, & Hall, 2009). The medicinal properties of alcohol were so numerous that 14th and 15th century European physicians referred to alcohol as “aqua vitae,” or “water of life,” reporting that alcohol “eases the pain in the teeth,…[and] in the breasts when swollen” (Roueche, 1963). Many 19th and 20th century physicians prescribed and administered oral or intravenous alcohol prior to medical procedures due to its analgesic and anesthetic properties (Brown & Cutter, 1977; Chapman & William, 1951; Crocq, 2007; Hanson, 1995). In the 1930s and 1940s, scientists began investigating the analgesic effects of alcohol in the laboratory setting. Indeed, various studies have found that alcohol dampens pain in a variety of pain tests in both humans and animals. Orally consumed alcohol decreases both mechanical and thermal pain sensitivity in both humans and rodents (Gatch, 2009; Gatch & Lal, 1999; Mullin & Luckhardt, 1934; Wolff, Hardy, & Goodell, 1942; Woodrow & Eltherington, 1988). Although there is conflicting evidence regarding the analgesic efficacy of intravenous (i.v.) ethanol in rodents (Bukusoglu, Thalhammer, & Krieger, 1993), in humans i.v. alcohol has been shown to produce analgesia comparable to i.v. morphine (Saddler, James, & Harington, 1985) as well as anti-nociceptive effects against pain due to electrical stimulation (Perrino et al., 2008; Ralevski et al., 2010) and capsaicin-induced hyperalgesia (Arout et al., 2016).

2.2. Alcohol analgesia in the context of AUD

Seventy-nine percent of high-risk drinkers and 61% of individuals suffering from alcohol dependence consume alcohol to treat pain, compared to only 38% of heavy drinkers and 35% of unhealthy drinkers (Alford et al., 2016). There is a strong mutual relationship between pain and alcohol in which alcohol affects nociceptive sensitivity and pain motivates alcohol consumption (Beasley, Macfarlane, & Macfarlane, 2016; Zale et al., 2015). This shared symptomatology can often make co-morbid directionalities difficult to assess (Macfarlane & Beasley, 2015). We will next discuss these interactions, beginning with a discussion of the analgesic effects of alcohol based on drinking history and on chronic pain status.

Early studies investigating the analgesic effects of alcohol in people with alcoholism found that whiskey dose-dependently reduces and abolishes reported pain in alcoholics but has no analgesic effects in non-alcoholics, regardless of dose (Cutter, Maloof, Kurtz, & Jones, 1976). Later studies found that alcohol reduces pain ratings in heavy social drinkers but increases pain ratings in people who regularly consume alcohol in a home setting, suggesting that the analgesic effects of alcohol may vary based on drinking history and pattern of drinking (Brown & Cutter, 1977). From a motivational perspective, alcohol is frequently self-administered for both its stress-reducing and analgesic effects (Egli et al., 2012; Thompson, Oram, Correll, Tsermentseli, & Stubbs, 2017). Self-reports of alcohol use specifically for pain management are common (e.g., Riley & King, 2009). Problem drinkers at risk for AUD not only report more severe pain symptoms compared to non-drinkers, but also report a higher incidence of using alcohol to manage their pain (Brennan, Schutte, & Moos, 2005).

As people with AUD experience a higher incidence of pain (Brennan, Schutte, SooHoo, & Moos, 2011; Zale et al., 2015) and appear to be more sensitive to the analgesic effects of alcohol, it is not surprising that pain severity and experience affects AUD treatment outcomes. Increases in both pain interference and intensity significantly increase the risk for relapse both during and after AUD treatment, and, more specifically, pain significantly predicts heavy drinking lapses both during and after AUD treatment (Witkiewitz et al., 2015). Likewise, decreases in physical pain from the start of AUD treatment are correlated with a decreased likelihood of relapse (Jakubczyk et al., 2016). Additionally, persistent pain is associated with increased heavy alcohol use 24 months following detoxification (Larson et al., 2007).

Chronic pain also affects the analgesic efficacy of alcohol. Adults with chronic pain, regardless of age, drink alcohol more heavily than the general population (Ditre et al., 2019), and 25% of people with chronic pain report using alcohol to manage their pain symptoms (Riley & King, 2009). Although one study found that a higher percentage of younger participants self-medicated their chronic pain with alcohol compared to older participants (Riley & King, 2009), alcohol consumption is self-reported as one of the most effective pain-relief methods in elderly persons who require assistance with activities of daily living (Jakobsson, Rahm Hallberg, & Westergren, 2004). Both human and animal studies have found that chronic pain is associated with and may drive increased alcohol consumption (Butler et al., 2017; Lawton & Simpson, 2009; Yu, Hwa, Makhijani, Besheer, & Kash, 2019).

Genetic vulnerability for high alcohol drinking may also play a role in the analgesic effects of alcohol. One study found that men with a high familial-genetic risk for alcoholism report higher pain ratings and experience greater reduction of pain in response to alcohol compared to men with no familial-genetic risk for alcoholism (Stewart, Finn, & Pihl, 1995). However, more recent studies have found that the analgesic effects of alcohol do not differ based on family history (Perrino et al., 2008; Ralevski et al., 2010). Rodents selectively bred for alcohol preference display decreased thermal pain thresholds in examinations of either spinal or supraspinal nociceptive behaviors (Kampov-Polevoy et al., 1996; Kimpel, Brown, & Froehlich, 2003). Several genes have been proposed to link alcohol dependence and pain, including the catechol-O-methyl-transferase (COMT) and mu opioid receptor 1 (OPRM1) genes (Egli et al., 2012).

2.3. Neurobiological mechanisms of alcohol analgesia

Despite the surplus of evidence that alcohol produces analgesia, the spinal and supraspinal neurobiological mechanisms behind this phenomenon are drastically understudied (Thompson et al., 2017). Three current hypotheses for this mechanism of action are discussed here, but it is important to note that alcohol likely produces analgesia via interactions between all of the mechanisms described below, as well as through other undiscovered mechanisms.

2.3.1. Opioid receptor systems

As the endogenous opioid neuropeptide system is integral to the production of analgesia, many scientists have examined the hypothesis that alcohol produces its analgesic effects by interacting with opioid receptor pharmacology. Interestingly, mice with high activity (HA) opioid systems display significantly greater ethanol-induced analgesia compared to controls, while their low activity (LA) counterparts display significantly less ethanol-induced analgesia (Mogil et al., 1993). However, other studies have produced mixed results. Depending on the dose of alcohol, pain model, pain test, and experimental species, the non-selective opioid receptor antagonist naloxone has been shown to both prevent and not affect alcohol-induced analgesia (Boada, Feria, & Sanz, 1981; Campbell, Taylor, & Tizabi, 2006, 2007; Cutter & O’Farrell, 1987; Jorgensen & Hole, 1981; Saddler et al., 1985). The specific opioid receptors responsible for the analgesic effects of alcohol are yet to be definitively determined. Genetic and pharmacological manipulation has implicated the delta-opioid receptor in the differences seen between the aforementioned HA and LA mice (Poznanski et al., 2017; Sacharczuk et al., 2014). However, a recent study found that the selective delta-opioid receptor antagonist naltrindole did not affect ethanol-induced anti-nociception (Neddenriep et al., 2019). This same study found that both high doses of naltrexone and the selective kappa-opioid receptor antagonist nor-BNI prevented ethanol-induced anti-nociception in a mouse model of chronic neuropathic pain, suggesting that the analgesic effects of alcohol are also mediated through both mu- and kappa-opioid receptors (Neddenriep et al., 2019). Complementary to their role in analgesia, it is important to note that all three opioid receptor systems represent targets for treating hyperalgesia in the context of substance use disorders (Delery & Edwards, 2020).

2.3.2. GABAergic and glutamatergic signaling

Alcohol is a positive allosteric modulator of the GABAA receptor, and GABAergic signaling likely plays a major role in the analgesic effects of drinking (Davies, 2003). The benzodiazepine antagonist flumazenil abolishes the anti-nociceptive effects of alcohol in thermal pain tests in rodents (Gatch, 1999), suggesting that GABA receptors are involved in mediating alcohol-induced analgesia. Additional studies are warranted to determine the contributions of regional (both spinal and supraspinal) activation of GABAergic neurons and interneurons in regulating pain sensitivity. Interestingly, infusion of ethanol directly into the medial prefrontal cortex of rats increases mechanical pain sensitivity by activating GABAA receptors there (Geng et al., 2016). In addition to the GABAA receptor, alcohol-induced analgesia may also be mediated via interactions with the N-methyl-d-aspartate (NMDA) glutamate receptor. The NMDA receptor antagonist MK-801 decreases anti-nociception produced by alcohol, and systemic administration of both MK-801 and naloxone completely abolishes alcohol-induced anti-nociception in mice, suggesting that the analgesic effects of alcohol are mediated by both NMDA and opioid receptors (Mogil et al., 1993).

2.3.3. GIRK2 potassium channels

The G protein-coupled inwardly rectifying potassium 2 (GIRK2) channels play a major role in substance-induced analgesia and have also been implicated in alcohol-induced analgesia (Bodhinathan & Slesinger, 2014; Ikeda et al., 2002; Lewohl et al., 1999). Ethanol directly interacts with and opens GIRK2 channels (Kobayashi et al., 1999). Mice with a mutated GIRK2 channel display reduced ethanol analgesia (Kobayashi et al., 1999), while the analgesic effects of ethanol are fully abolished in GIRK2 knockout mice (Blednov, Stoffel, Alva, & Harris, 2003).

2.3.4. Additional systems and considerations

Additional research is needed to examine the contribution of other neurobiological systems to alcohol analgesia. In addition to opioid signaling, it is likely that another endogenous analgesic system, the endocannabinoid system, may be intimately involved. Endocannabinoids regulate not only pain sensitivity (Woodhams, Chapman, Finn, Hohmann, & Neugebauer, 2017), but also act as a buffer of stress and negative affect (Hillard, 2014; Morena, Patel, Bains, & Hill, 2016; Tasker et al., 2015). Extensive research has also implicated endocannabinoid signaling in alcohol reward and reinforcement (Kirson, Oleata, Parsons, Ciccocioppo, & Roberto, 2018; Serrano et al., 2018; Varodayan et al., 2016).

Another consideration for future research is whether and how tolerance to alcohol analgesia develops over time across a range of disease contexts. Damaj and colleagues (Neddenriep et al., 2019) recently demonstrated that the analgesic efficacy of repeated alcohol diminished over time in an animal model of chronic neuropathic pain. Such findings presage a possible rationale for escalation of drinking in individuals using alcohol to manage pain, possibly also driving AUD. Further research is needed to determine the neuroanatomical and molecular sites of action for alcohol analgesia. It will also be important to understand which analgesic systems or mechanisms become compromised with chronic alcohol use, as supporting these systems may represent a beneficial pharmacological strategy for targeting alcohol-associated hyperalgesia. Such treatments may also support additional strategies described in the next section, which correspond to known neuroadaptations shared between excessive alcohol use and chronic pain conditions that underlie hyperalgesia.

3. Medication strategies for treating pain in the context of AUD

3.1. Shared neuroadaptations between chronic pain, negative affect, and AUD

AUD has been hypothesized to stem from functional alterations in the neurobiological substrates of supraspinal pain processing (Egli et al., 2012), including a key contribution from the nociceptive central amygdala (CeA; Neugebauer, 2015). Importantly, the sensitization of pain-driven negative affect (Ji et al., 2007) is also thought to be mediated by the CeA and to closely interact with similar neuronal mechanisms related to the development of AUD (Edwards et al., 2020). Both alcohol dependence and persistent pain produce multiple neurophysiological adaptations within the CeA (e.g., Carrasquillo & Gereau, 2008; Gilpin, Herman, & Roberto, 2015; Ji & Neugebauer, 2009; Roberto et al., 2010; Zhang et al., 2014). The CeA receives functionally distinct projections from the pontine parabrachial area (PB, nociceptive coding) and basolateral amygdala (BLA, sensory-affective coding) that are sensitized in the context of chronic pain (Ikeda, Takahashi, Inoue, & Kato, 2007; Neugebauer, Li, Bird, Bhave, & Gereau, 2003). Chronic pain-induced activation of the CeA is accompanied by dysregulation of amygdala-driven prefrontal cortex function and resultant cognitive deficits (Ji & Neugebauer, 2011; Ji et al., 2010; Sun & Neugebauer, 2011). Importantly, executive system dysfunction is thought to play a central role in the heavily compromised decision-making that accompanies the transition from recreational drug use to substance use disorder (George & Koob, 2010), which may contribute to alcohol misuse in individuals suffering from chronic pain. Alcohol withdrawal produces hyperalgesia that is mediated by altered activity of CeA projections to the periaqueductal gray (PAG), a region that is critically involved in descending pain modulation (Avegno et al., 2018).

Elucidation of chronic alcohol-induced neuroadaptations within brain stress and nociceptive systems may provide valuable insights into potential mechanisms underlying the transition to AUD in vulnerable individuals. In these regions, elements of brain stress signaling, such as the corticotropin-releasing factor (CRF), vasopressin, and glucocorticoid receptor (GR) systems, are recruited (Edwards, Little, Richardson, & Vendruscolo, 2015; Koob & Le Moal, 2008) during the transition to dependence, as described below.

3.2. Corticotropin-releasing factor receptor systems

As a key mediator of the heightened negative affective response that manifests during alcohol withdrawal, the functional potentiation of CRF signaling is evident in the establishment and progression of AUD-related behaviors (including escalated alcohol drinking) observed in preclinical animal models (Edwards & Koob, 2010; Funk, Zorrilla, Lee, Rice, & Koob, 2007; Roberto et al., 2010; Schreiber & Gilpin, 2018). CRF is a neuropeptide synthesized by the paraventricular nucleus (PVN) of the hypothalamus where it initiates the hypothalamic-pituitary-adrenal (HPA) axis response to stress. CRF stimulates synthesis of adrenocorticotropic hormone (ACTH) in the anterior pituitary, which then facilitates the secretion of glucocorticoids from the adrenal gland to mediate adaptive stress responses and restore homeostasis. High concentrations of CRF are also present in basal forebrain and brainstem areas (Swanson, Sawchenko, Rivier, & Vale, 1983), where the neuropeptide regulates autonomic and behavioral responses to stress. Importantly, extra-hypothalamic CRF1 receptors (CRF1Rs) are widely expressed throughout the brain, and activation of these receptors potentiates central stress responsiveness. However, activation of complementary CRF2 receptors mediates a contrasting (but not necessarily opposing) effect on anxiety-like behavior (Zhao et al., 2007) and CeA plasticity (Varodayan et al., 2017).

Chronic pain produces a number of physiological changes throughout the body, including increases in CRF levels in the CeA (Rouwette et al., 2012). Complementary to this neuroadaptation, the anti-hyperalgesic effects of CRF1R antagonists (administered either systemically or in some cases intra-CeA) have been demonstrated in both mice and rats across a variety of pain models, including neuropathic pain, inflammatory pain, and visceral pain (e.g., Hummel et al., 2010; Ji & Neugebauer, 2007; Nijsen, Ongenae, Meulemans, & Coulie, 2005; Schwetz et al., 2004). CRF would indeed appear to play a key role in processing an array of pain modalities (e.g., mechanical, thermal, visceral), suggesting that its role is not modality-specific but may generalize to all classes of pain. Importantly, CRF1R antagonists do not alter baseline measures of various nociception-related indices (e.g., mechanical or thermal paw withdrawal thresholds, audible or ultrasonic vocalizations) in non-injured animals, indicating the absence of a direct analgesic effect (e.g., Baiamonte et al., 2014; Cohen et al., 2015; Fu & Neugebauer, 2008; Roltsch et al., 2014). In comparison, CRF infusion into the CeA produces hyperalgesia in otherwise naïve animals (Itoga et al., 2016). CRF also directly alters the electrophysiological properties of CeA neurons to drive pain sensitization. For example, rodents in a state of arthritic inflammatory pain (Neugebauer, Han, Adwanikar, Fu, & Ji, 2007) exhibit enhanced CeA excitability that is attenuated by CRF1R antagonism (Ji et al., 2007). The authors also found that the normally inhibitory role of CRF2R signaling in this region was lost in the arthritis model, further demonstrating the complementary roles of CRF1 vs CRF2 receptors in neurophysiology, including pain signaling (Ji & Neugebauer, 2008). Interestingly, arthritis increases anxiety-like behavior and this effect is attenuated by either systemic or intra-CeA CRF1R antagonism (Ji et al., 2007), suggesting that CRF signaling drives the intersection of pain and negative affect within the CeA (Egli et al., 2012; Neugebauer, Li, Bird, & Han, 2004). Stress- and alcohol-related factors other than pain also produce an enhancement of CRF activity in the CeA (Gilpin, 2012; Roberto et al., 2010), and it was also demonstrated that non-pain-related activation of CRF1R signaling in the CeA augments nociceptive sensitivity (Ji, Fu, Adwanikar, & Neugebauer, 2013).

3.3. Vasopressin receptor systems

Several emerging lines of evidence have implicated the neuropeptide vasopressin in the pathophysiology of stress- and emotion-related behaviors, often in a sexually dimorphic fashion (Bisagno & Cadet, 2014). In addition to the well-characterized role of hypothalamic vasopressin acting in conjunction with CRF to stimulate the HPA axis (Lolait, Stewart, Jessop, Young, & O’Carroll, 2007), vasopressin-synthesizing neurons are also localized in the bed nucleus of the stria terminalis (BNST) and medial amygdala (De Vries & Buijs, 1983), project extensively throughout the limbic system (Veinante & Freund-Mercier, 1997), and play a significant role in regulating various complex behaviors, including aggression and social affiliation. Vasopressin signaling within the brain is thought to underlie various aspects of emotional processing (Caldwell, Lee, Macbeth, & Young, 2008), and early studies established a specific role for central vasopressin in aversive learning and memory mechanisms (Koob & Bloom, 1982). More recently, the generation of small molecule, receptor subtype-selective antagonists has greatly advanced the characterization of the complex behavior regulated by V1b receptors (V1bR), which are thought to play a role in generalized negative affective-like behaviors, as demonstrated by the anxiolytic-like, antidepressant-like, and anti-hyperalgesic-like effects of the small molecule V1bR antagonist SSR149415 (Bradesi, Martinez, Lao, Larsson, & Mayer, 2009; Griebel et al., 2002). Studies utilizing regional microinjections of SSR149415 have established a particular role for negative affective-like V1bR signaling in the amygdala, as SSR149415 reduces both anxiety-and depression-like behavior (Salome, Stemmelin, Cohen, & Griebel, 2006). Vasopressin gene (Avp) expression levels are higher in the CeA of male Sardinian alcohol-preferring (sP) relative to alcohol-non-preferring (sNP) rats. Interestingly, the excessive drinking of sP rats reduces elevated Avp levels, while systemic SSR149415 administration is effective in reducing drinking in this population (Zhou et al., 2011). Independent lines of preclinical evidence strongly point to a role for vasopressin/V1bR signaling in the transition to alcohol dependence (Edwards, Guerrero, et al., 2012; Zhou & Kreek, 2014).

Given the literature suggesting that V1bR antagonists have substantial efficacy in reducing negative affective-like profiles and the fact that vasopressin and its central receptors (V1bR and V1aR) are highly expressed in extended amygdala and cortical areas (Stemmelin, Lukovic, Salome, & Griebel, 2005; Tribollet, Raufaste, Maffrand, & Serradeil-Le Gal, 1999), vasopressin systems could play a central role in the increased alcohol intake and hyperalgesia associated with dependence. These effects may occur via the ability of vasopressin to activate the HPA axis (Volpi, Rabadan-Diehl, & Aguilera, 2004) and/or via direct central (i.e., extra-hypothalamic) actions. Interestingly, the effects of V1bR blockade on excessive alcohol self-administration (Edwards, Guerrero, et al., 2012) closely resemble inhibition of the CRF1 receptor system on this behavior (Richardson et al., 2008), and CRF1R antagonism has already been demonstrated to alleviate symptoms of mechanical hyperalgesia in alcohol-dependent rats (Edwards, Vendruscolo, et al., 2012). Accordingly, either in addition to or in cooperation with CRF, vasopressin V1bR activity may represent another mechanism whereby negative reinforcement mechanisms regulate alcohol dependence-related symptomatology. Perhaps most importantly, recent evidence has emerged for the safety and clinical efficacy of V1bR antagonists to reduce HPA axis activation and increase abstinence in alcohol-dependent patients (Katz, Liu, Locke, Dutta, & Tracy, 2016; Ryan et al., 2017; ClinicalTrials.gov Identifier NCT01613014).

In comparison to V1bRs, V1aRs are expressed at even higher levels in the brain (Tribollet et al., 1999), and are important for mediating social behavior. Systemic and brain administration of the V1aR antagonist SR49059 improves neurological and neurobehavioral outcomes in animal models of traumatic brain injury (TBI; Krieg, Sonanini, Plesnila, & Trabold, 2015; Manaenko et al., 2011; Marmarou et al., 2014). Interestingly, pain-related affective responses are enhanced via vasopressin signaling through V1a receptors in the CeA (Cragg, Ji, & Neugebauer, 2016), an effect that is blocked via intra-CeA administration of SR49059. These findings make V1aR antagonism a promising therapeutic strategy for both TBI and pain (which are themselves highly comorbid), although the contribution of this receptor system to excessive drinking or hyperalgesia in the context of alcohol dependence has remained unexplored.

3.4. Glucocorticoid receptor system

Both alcohol and alcohol withdrawal elevate systemic glucocorticoid levels due to activation of the endocrine HPA axis (Rivier, 2014). In addition to profound dysregulation of HPA axis activity (Richardson, Lee, O’Dell, Koob, & Rivier, 2008), alcohol-dependent animals exhibit a functional increase in central brain GR signaling, a key mediator of stress responsiveness (Edwards et al., 2015). Importantly, GR antagonism via mifepristone reduces escalated drinking in both preclinical animal models and early clinical trials (Vendruscolo et al., 2015; ClinicalTrials.gov Identifier NCT01548417). As a potential biochemical correlate of sensitized central GR signaling that may underlie these promising clinical directions, previous work describes increases in GR phosphorylation in the CeA of alcohol-dependent animals (Vendruscolo et al., 2015). At the molecular level, the transcriptional activity of GR is directly controlled via phosphorylation at serine 232 (Adzic et al., 2009), and two key downstream targets of GR transcriptional activity are Crh and Crhr1 (Yao & Denver, 2007). Importantly, systemic administration of either the GR antagonist mifepristone (Dina et al., 2008) or a CRF1R antagonist (Edwards, Vendruscolo, et al., 2012) alleviates hyperalgesia in binge drinking and alcohol-dependent animals (respectively). It is also important to note that in contrast to the classic GR-mediated negative feedback regulation of CRF at the level of the HPA axis, GR activity actually facilitates CRF gene expression in certain central brain regions (Edwards et al., 2015) including the CeA (Makino, Gold, & Schulkin, 1994). Differential expression and activity of steroid receptor coactivator-1 (SRC-1) isoforms between the PVN and CeA may account for this dichotomy (Lachize et al., 2009). As described above, intra-CeA administration of CRF1R antagonists is effective in alleviating hyperalgesia in both pain and drug dependence models (McGinn & Edwards, 2016). Unfortunately, evidence for the clinical efficacy of CRF1 receptor antagonists to reduce excessive drinking has been slow to develop, with some early failures in clinical trials (e.g., Kwako et al., 2015). However, development of novel antagonists with different brain pharmacokinetics and/or a more precise selection of specific treatment populations (e.g., those suffering from co-morbid hyperalgesia) may unmask the true utility of this medication class (Spierling & Zorrilla, 2017). In addition to the CRF system, alternative stress-regulated targets either independent from or even transcriptionally regulated by GR, as in the case of V1aR (Watters, Swank, Wilkinson, & Dorsa, 1996) and V1bR (Aguilera & Rabadan-Diehl, 2000), would appear to offer a very promising path forward for treating severe AUD and associated pain conditions.

4. Summary

Pain represents a uniquely subjective experience that may exert a powerful influence on reinforcement processes, possibly facilitating the transition to substance use disorders (Miller & Gold, 2007; Shurman, Koob, & Gutstein, 2010), including AUD (Edwards et al., 2020). Recent insights into the mechanisms that mediate the analgesic efficacy and abuse-related properties of alcohol have provided a basis for a conceptualization of the neurobiological intersection of pain and AUD. The analgesic effects of alcohol have been known anecdotally for centuries and have been scientifically investigated for almost 100 years. Drinking history, alcohol dependence, chronic pain status, and genetic risk for AUD all affect the analgesic efficacy of alcohol (Edwards et al., 2020). The mechanisms behind alcohol-induced analgesia are understudied, but the primary mechanisms proposed include the endogenous opioid system, GABA/NMDA signaling, and GIRK2 channel activity. It is likely that alcohol interacts with each of these systems to produce its analgesic effects, while future studies are needed to understand the possible role of other endogenous analgesia systems, such as the endocannabinoid system.

Neurophysiological processes that counter the anxiolytic and analgesic properties of alcohol may drive AUD progression through the generation of hyperalgesia and closely associated negative affective states. In turn, given that chronic pain itself often produces emotional distress, alcohol-related hyperalgesia may be closely associated with the transition to AUD via facilitation of negative reinforcement mechanisms. Strong evidence suggests that the neural substrates associated with addiction may overlap with substrates of emotional aspects of pain processing in brain regions where ascending nociceptive circuitry terminates (Egli et al., 2012). Specifically, the affective component of pain is strongly regulated by the CeA (Neugebauer, 2015). Numerous preclinical models have demonstrated a functional role for the CeA in regulating persistent hyperalgesia associated with a range of neurological and psychiatric conditions ranging from arthritis (Ji et al., 2007) to opioid dependence (McNally & Akil, 2002). This neurocircuitry may promote crosstalk among a variety of psychiatric disorders and account for the high degree of comorbidity between affective disorders, AUD and chronic pain (Edwards et al., 2020). Medications such as CRF1 receptor antagonists, vasopressin receptor antagonists, and GR antagonists that act on these shared systems would likely be effective in treating a range of affective pain-related conditions, including AUD (Fig. 1). Moreover, dependence on other abused drugs commonly taken with alcohol (e.g., nicotine) is associated with a CRF1R-dependent hyperalgesia (Baiamonte et al., 2014; Cohen et al., 2015; Edwards, Vendruscolo, et al., 2012). As a result, patients with a history of alcohol or other substance use disorders may be susceptible to the activation of a common set of nociceptive factors that could drive relapse and abuse of any or all of these substances. Thus, we and others have hypothesized that CeA neuroadaptations facilitate pain-driven motivated behaviors that underlie dependence-induced hyperalgesia and excessive drinking. It is our hope that the functional integration of pain- and addiction-related research objectives across the National Institutes of Health will promote further collaboration among addiction and pain neuroscientists, leading to broader and more integrative therapeutic strategies for these devastating conditions.

Fig. 1.

Fig. 1

There is an urgent need to understand the neurobiological mechanisms of pain and AUD, as well as a continuing need for preclinical medication development targeted to novel systems to treat these devastating conditions. As hyperalgesia represents a form of chronic stress and negative affect, we expect that this condition plays a critical role in negative reinforcement mechanisms underlying excessive drinking in the context of AUD. Following from this conceptualization, the interrogation of brain stress systems would likely represent a prolific avenue for medication development. Neurobiological changes across both hypothalamic and extra-hypothalamic stress systems (including changes in CRF, vasopressin, and GR signaling) have been observed in preclinical AUD animal models, with the central amygdala (CeA) playing a critical role in mediating the interaction of pain and negative affective-related behaviors expected to contribute to AUD.

Acknowledgments

Preparation of this review was supported by National Institute on Alcohol Abuse and Alcoholism grants AA028445, AA025996, AA007577, and AA009803.

Footnotes

Disclosure

S.E. is a consultant for Avanos Medical, Inc., a private company focused on the development of medical devices for pain management. J.A.C.R. has no conflicts related to this work.

References

  1. Adzic M, Djordjevic J, Djordjevic A, Niciforovic A, Demonacos C, Radojcic M, et al. (2009). Acute or chronic stress induce cell compartment-specific phosphorylation of glucocorticoid receptor and alter its transcriptional activity in Wistar rat brain. The Journal of Endocrinology, 202(1), 87–97. 10.1677/JOE-08-0509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Aguera-Ortiz L, Failde I, Mico JA, Cervilla J, & Lopez-Ibor JJ (2011). Pain as a symptom of depression: Prevalence and clinical correlates in patients attending psychiatric clinics. Journal of Affective Disorders, 130(1–2), 106–112. 10.1016/j.jad.2010.10.022. [DOI] [PubMed] [Google Scholar]
  3. Aguilera G, & Rabadan-Diehl C (2000). Regulation of vasopressin V1b receptors in the anterior pituitary gland of the rat. Experimental Physiology, 85(s1), 19S–26S. 10.1111/j.1469-445x.2000.tb00004.x. [DOI] [PubMed] [Google Scholar]
  4. Alford DP, German JS, Samet JH, Cheng DM, Lloyd-Travaglini CA, & Saitz R (2016). Primary care patients with drug use report chronic pain and self-medicate with alcohol and other drugs. Journal of General Internal Medicine, 31(5), 486–491. 10.1007/s11606-016-3586-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Arnow BA, Hunkeler EM, Blasey CM, Lee J, Constantino MJ, Fireman B, et al. (2006). Comorbid depression, chronic pain, and disability in primary care. Psychosomatic Medicine, 68(2), 262–268. 10.1097/01.psy.0000204851.15499.fc. [DOI] [PubMed] [Google Scholar]
  6. Arout CA, Perrino AC Jr., Ralevski E, Acampora G, Koretski J, Limoncelli D, et al. (2016). Effect of intravenous ethanol on capsaicin-induced hyperalgesia in human subjects. Alcoholism, Clinical and Experimental Research, 40(7), 1425–1429. 10.1111/acer.13095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Asmundson GJ, & Katz J (2009). Understanding the co-occurrence of anxiety disorders and chronic pain: State-of-the-art. Depression and Anxiety, 26(10), 888–901. 10.1002/da.20600. [DOI] [PubMed] [Google Scholar]
  8. Avegno EM, Lobell TD, Itoga CA, Baynes BB, Whitaker AM, Weera MM, et al. (2018). Central amygdala circuits mediate hyperalgesia in alcohol-dependent rats. The Journal of Neuroscience, 38(36), 7761–7773. 10.1523/JNEUROSCI.0483-18.2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Bahaaddini M, Khatamsaz S, Esmaeili-Mahani S, Abbasnejad M, & Raoof M (2016). The role of trigeminal nucleus caudalis orexin 1 receptor in orofacial pain-induced anxiety in rat. Neuroreport, 27(15), 1107–1113. 10.1097/WNR.0000000000000660. [DOI] [PubMed] [Google Scholar]
  10. Baiamonte BA, Valenza M, Roltsch EA, Whitaker AM, Baynes BB, Sabino V, et al. (2014). Nicotine dependence produces hyperalgesia: Role of corticotropin-releasing factor-1 receptors (CRF1Rs) in the central amygdala (CeA). Neuropharmacology, 77, 217–223. 10.1016/j.neuropharm.2013.09.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Bair MJ, Robinson RL, Katon W, & Kroenke K (2003). Depression and pain comorbidity: A literature review. Archives of Internal Medicine, 163(20), 2433–2445. 10.1001/archinte.163.20.2433. [DOI] [PubMed] [Google Scholar]
  12. Beasley MJ, Macfarlane TV, & Macfarlane GJ (2016). Is alcohol consumption related to likelihood of reporting chronic widespread pain in people with stable consumption? Results from UK biobank. Pain, 157(11), 2552–2560. 10.1097/j.pain.0000000000000675. [DOI] [PubMed] [Google Scholar]
  13. Bisagno V, & Cadet JL (2014). Stress, sex, and addiction: Potential roles of corticotropin-releasing factor, oxytocin, and arginine-vasopressin. Behavioural Pharmacology, 25(5–6), 445–457. 10.1097/FBP.0000000000000049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Blednov YA, Stoffel M, Alva H, & Harris RA (2003). A pervasive mechanism for analgesia: Activation of GIRK2 channels. Proceedings of the National Academy of Sciences of the United States of America, 100(1), 277–282. 10.1073/pnas.012682399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Boada J, Feria M, & Sanz E (1981). Inhibitory effect of naloxone on the ethanol-induced antinociception in mice. Pharmacological Research Communications, 13(7), 673–678. 10.1016/s0031-6989(81)80055-0. [DOI] [PubMed] [Google Scholar]
  16. Bodhinathan K, & Slesinger PA (2014). Alcohol modulation of G-protein-gated inwardly rectifying potassium channels: From binding to therapeutics. Frontiers in Physiology, 5, 76. 10.3389/fphys.2014.00076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Bradesi S, Martinez V, Lao L, Larsson H, & Mayer EA (2009). Involvement of vasopressin 3 receptors in chronic psychological stress-induced visceral hyperalgesia in rats. American Journal of Physiology. Gastrointestinal and Liver Physiology, 296(2), G302–G309. 10.1152/ajpgi.90557.2008. [DOI] [PubMed] [Google Scholar]
  18. Brennan PL, Schutte KK, & Moos RH (2005). Pain and use of alcohol to manage pain: Prevalence and 3-year outcomes among older problem and non-problem drinkers. Addiction, 100(6), 777–786. 10.1111/j.1360-0443.2005.01074.x. [DOI] [PubMed] [Google Scholar]
  19. Brennan PL, Schutte KK, SooHoo S, & Moos RH (2011). Painful medical conditions and alcohol use: A prospective study among older adults. Pain Medicine, 12(7), 1049–1059. 10.1111/j.1526-4637.2011.01156.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Brown RA, & Cutter HSG (1977). Alcohol, customary drinking behavior, and pain. Journal of Abnormal Psychology, 86(2), 179–188. 10.1037/0021-843X.86.2.179. [DOI] [PubMed] [Google Scholar]
  21. Bukusoglu C, Thalhammer JG, & Krieger NR (1993). Analgesia with anesthetic steroids and ethanol. Analgesia and Anesthesia, 77(1), 27–31. [PubMed] [Google Scholar]
  22. Butler RK, Knapp DJ, Ulici V, Longobardi L, Loeser RF, & Breese GR (2017). A mouse model for chronic pain-induced increase in ethanol consumption. Pain, 158(3), 457–462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Caldwell HK, Lee HJ, Macbeth AH, & Young WS 3rd. (2008). Vasopressin: Behavioral roles of an “original” neuropeptide. Progress in Neurobiology, 84(1), 1–24. 10.1016/j.pneurobio.2007.10.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Campbell VC, Taylor RE, & Tizabi Y (2006). Antinociceptive effects of alcohol and nicotine: Involvement of the opioid system. Brain Research, 1097(1), 71–77. 10.1016/j.brainres.2006.04.054. [DOI] [PubMed] [Google Scholar]
  25. Campbell VC, Taylor RE, & Tizabi Y (2007). Effects of selective opioid receptor antagonists on alcohol-induced and nicotine-induced antinociception. Alcoholism, Clinical and Experimental Research, 31(8), 1435–1440. 10.1111/j.1530-0277.2007.00432.x. [DOI] [PubMed] [Google Scholar]
  26. Campo JV, Bridge J, Ehmann M, Altman S, Lucas A, Birmaher B, et al. (2004). Recurrent abdominal pain, anxiety, and depression in primary care. Pediatrics, 113(4), 817–824. 10.1542/peds.113.4.817. [DOI] [PubMed] [Google Scholar]
  27. Carrasquillo Y, & Gereau RW (2008). Hemispheric lateralization of a molecular signal for pain modulation in the amygdala. Molecular Pain, 4, 24. 10.1186/1744-8069-4-24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Chapman ER, & William PT (1951). Intravenous alcohol as an obstetrical analgesia. American Journal of Obstetrics and Gynecology, 61(3), 676–679. [DOI] [PubMed] [Google Scholar]
  29. Cohen A, Treweek J, Edwards S, Leao RM, Schulteis G, Koob GF, et al. (2015). Extended access to nicotine leads to a CRF1 receptor dependent increase in anxiety-like behavior and hyperalgesia in rats. Addiction Biology, 20(1), 56–68. 10.1111/adb.12077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Cox DR, Ashby S, DeConde AS, Mace JC, Orlandi RR, Smith TL, et al. (2016). Dyad of pain and depression in chronic rhinosinusitis. International Forum of Allergy & Rhinology, 6(3), 308–314. 10.1002/alr.21664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Cragg B, Ji G, & Neugebauer V (2016). Differential contributions of vasopressin V1A and oxytocin receptors in the amygdala to pain-related behaviors in rats. Molecular Pain, 12. 10.1177/1744806916676491. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Crocq MA (2007). Historical and cultural aspects of man’s relationship with addictive drugs. Dialogues in Clinical Neuroscience, 9(4), 355–361. Retrieved from https://www.ncbi.nlm.nih.gov/pubmed/18286796. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Cutter HS, Maloof B, Kurtz NR, & Jones WC (1976). “Feeling no pain” differential responses to pain by alcoholics and nonalcoholics before and after drinking. Journal of Studies on Alcohol, 37(3), 273–277. 10.15288/jsa.1976.37.273. [DOI] [PubMed] [Google Scholar]
  34. Cutter HS, & O’Farrell TJ (1987). Experience with alcohol and the endogenous opioid system in ethanol analgesia. Addictive Behaviors, 12(4), 331–343. 10.1016/0306-4603(87)90047-5. [DOI] [PubMed] [Google Scholar]
  35. Dahlhamer J, Lucas J, Zelaya C, Nahin R, Mackey S, DeBar L, et al. (2018). Prevalence of chronic pain and high-impact chronic pain among adults—United States, 2016. MMWR: Morbidity and Mortality Weekly Report, 67(36), 1001–1006. 10.15585/mmwr.mm6736a2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Davies M (2003). The role of GABAA receptors in mediating the effects of alcohol in the central nervous system. Journal of Psychiatry & Neuroscience, 28(4), 263–274. Retrieved from https://www.ncbi.nlm.nih.gov/pubmed/12921221. [PMC free article] [PubMed] [Google Scholar]
  37. De Vries GJ, & Buijs RM (1983). The origin of the vasopressinergic and oxytocinergic innervation of the rat brain with special reference to the lateral septum. Brain Research, 273(2), 307–317. 10.1016/0006-8993(83)90855-7. [DOI] [PubMed] [Google Scholar]
  38. Delery EC, & Edwards S (2020). Neuropeptide and cytokine regulation of pain in the context of substance use disorders. Neuropharmacology, 174, 108153. 10.1016/j.neuropharm.2020.108153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Dimitrov EL, Tsuda MC, Cameron HA, & Usdin TB (2014). Anxiety- and depression-like behavior and impaired neurogenesis evoked by peripheral neuropathy persist following resolution of prolonged tactile hypersensitivity. The Journal of Neuroscience, 34(37), 12304–12312. 10.1523/JNEUROSCI.0312-14.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Dina OA, Barletta J, Chen X, Mutero A, Martin A, Messing RO, et al. (2000). Key role for the epsilon isoform of protein kinase C in painful alcoholic neuropathy in the rat. Journal of Neuroscience, 20(22), 8614–8619. Retrieved from https://www.ncbi.nlm.nih.gov/pubmed/11069970. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Dina OA, Khasar SG, Alessandri-Haber N, Green PG, Messing RO, & Levine JD (2008). Alcohol-induced stress in painful alcoholic neuropathy. The European Journal of Neuroscience, 27(1), 83–92. 10.1111/j.1460-9568.2007.05987.x. [DOI] [PubMed] [Google Scholar]
  42. Ditre JW, Zale EL, & LaRowe LR (2019). A reciprocal model of pain and substance use: Transdiagnostic considerations, clinical implications, and future directions. Annual Review of Clinical Psychology, 15, 503–528. 10.1146/annurev-clinpsy-050718-095440. [DOI] [PubMed] [Google Scholar]
  43. Edwards S, Guerrero M, Ghoneim OM, Roberts E, & Koob GF (2012). Evidence that vasopressin V1b receptors mediate the transition to excessive drinking in ethanol-dependent rats. Addiction Biology, 17(1), 76–85. 10.1111/j.1369-1600.2010.00291.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Edwards S, & Koob GF (2010). Neurobiology of dysregulated motivational systems in drug addiction. Future Neurology, 5(3), 393–401. 10.2217/fnl.10.14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Edwards S, Little HJ, Richardson HN, & Vendruscolo LF (2015). Divergent regulation of distinct glucocorticoid systems in alcohol dependence. Alcohol, 49(8), 811–816. 10.1016/j.alcohol.2015.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Edwards S, Vendruscolo LF, Gilpin NW, Wojnar M, & Witkiewitz K (2020). Alcohol and pain: A translational review of preclinical and clinical findings to inform future treatment strategies. Alcoholism, Clinical and Experimental Research, 44(2), 368–383. 10.1111/acer.14260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Edwards S, Vendruscolo LF, Schlosburg JE, Misra KK, Wee S, Park PE, et al. (2012). Development of mechanical hypersensitivity in rats during heroin and ethanol dependence: Alleviation by CRF(1) receptor antagonism. Neuropharmacology, 62(2), 1142–1151. 10.1016/j.neuropharm.2011.11.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Egli M, Koob GF, & Edwards S (2012). Alcohol dependence as a chronic pain disorder. Neuroscience and Biobehavioral Reviews, 36(10), 2179–2192. 10.1016/j.neubiorev.2012.07.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Elman I, Borsook D, & Volkow ND (2013). Pain and suicidality: Insights from reward and addiction neuroscience. Progress in Neurobiology, 109, 1–27. 10.1016/j.pneurobio.2013.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Feinmann C (1983). Psychogenic facial pain: Presentation and treatment. Journal of Psychosomatic Research, 27(5), 403–410. 10.1016/0022-3999(83)90076-4. [DOI] [PubMed] [Google Scholar]
  51. Fishbain DA, Cutler RB, Rosomoff HL, & Rosomoff RS (1998). Do antidepressants have an analgesic effect in psychogenic pain and somatoform pain disorder? A meta-analysis. Psychosomatic Medicine, 60(4), 503–509. 10.1097/00006842-199807000-00019. [DOI] [PubMed] [Google Scholar]
  52. Fishbain DA, Goldberg M, Meagher BR, Steele R, & Rosomoff H (1986). Male and female chronic pain patients categorized by DSM-III psychiatric diagnostic criteria. Pain, 26(2), 181–197. 10.1016/0304-3959(86)90074-6. [DOI] [PubMed] [Google Scholar]
  53. France RD, Houpt JL, Skott A, Krishnan KR, & Varia IM (1986). Depression as a psychopathological disorder in chronic low back pain patients. Journal of Psychosomatic Research, 30(2), 127–133. 10.1016/0022-3999(86)90041-3. [DOI] [PubMed] [Google Scholar]
  54. Fu R, Gregor D, Peng Z, Li J, Bekker A, & Ye J (2015). Chronic intermittent voluntary alcohol drinking induces hyperalgesia in Sprague-Dawley rats. International Journal of Physiology, Pathophysiology and Pharmacology, 7(3), 136–144. Retrieved from https://www.ncbi.nlm.nih.gov/pubmed/26823962. [PMC free article] [PubMed] [Google Scholar]
  55. Fu Y, & Neugebauer V (2008). Differential mechanisms of CRF1 and CRF2 receptor functions in the amygdala in pain-related synaptic facilitation and behavior. The Journal of Neuroscience, 28(15), 3861–3876. 10.1523/JNEUROSCI.0227-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Funk CK, Zorrilla EP, Lee MJ, Rice KC, & Koob GF (2007). Corticotropin-releasing factor 1 antagonists selectively reduce ethanol self-administration in ethanol-dependent rats. Biological Psychiatry, 61(1), 78–86. 10.1016/j.biopsych.2006.03.063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Gatch MB (1999). Effects of benzodiazepines on acute and chronic ethanol-induced nociception in rats. Alcoholism, Clinical and Experimental Research, 23(11), 1736–1743. Retrieved from https://www.ncbi.nlm.nih.gov/pubmed/10591589. [PubMed] [Google Scholar]
  58. Gatch MB (2009). Ethanol withdrawal and hyperalgesia. Current Drug Abuse Reviews, 2(1),41–50. 10.2174/1874473710902010041. [DOI] [PubMed] [Google Scholar]
  59. Gatch MB, & Lal H (1999). Effects of ethanol and ethanol withdrawal on nociception in rats. Alcoholism, Clinical and Experimental Research, 23(2), 328–333. Retrieved from https://www.ncbi.nlm.nih.gov/pubmed/10069564. [PubMed] [Google Scholar]
  60. Geng KW, He T, Wang RR, Li CL, Luo WJ, Wu FF, et al. (2016). Ethanol increases mechanical pain sensitivity in rats via activation of GABAA receptors in medial prefrontal cortex. Neuroscience Bulletin, 32(5), 433–444. 10.1007/s12264-016-0063-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. George O, & Koob GF (2010). Individual differences in prefrontal cortex function and the transition from drug use to drug dependence. Neuroscience and Biobehavioral Reviews, 35(2), 232–247. 10.1016/j.neubiorev.2010.05.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Gilpin NW (2012). Corticotropin-releasing factor (CRF) and neuropeptide Y (NPY): Effects on inhibitory transmission in central amygdala, and anxiety- & alcohol-related behaviors. Alcohol, 46(4), 329–337. 10.1016/j.alcohol.2011.11.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Gilpin NW, Herman MA, & Roberto M (2015). The central amygdala as an integrative hub for anxiety and alcohol use disorders. Biological Psychiatry, 77, 859–869. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Goldberg DS, & McGee SJ (2011). Pain as a global public health priority. BMC Public Health, 11, 770. 10.1186/1471-2458-11-770. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Goncalves L, Silva R, Pinto-Ribeiro F, Pego JM, Bessa JM, Pertovaara A, et al. (2008). Neuropathic pain is associated with depressive behaviour and induces neuro-plasticity in the amygdala of the rat. Experimental Neurology, 213(1), 48–56. 10.1016/j.expneurol.2008.04.043. [DOI] [PubMed] [Google Scholar]
  66. Grant BF, Chou SP, Saha TD, Pickering RP, Kerridge BT, Ruan WJ, et al. (2017). Prevalence of 12-month alcohol use, high-risk drinking, and DSM-IV alcohol use disorder in the United States, 2001–2002 to 2012–2013: Results from the national epidemiologic survey on alcohol and related conditions. JAMA Psychiatry, 74(9), 911–923. 10.1001/jamapsychiatry.2017.2161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Griebel G, Simiand J, Serradeil-Le Gal C, Wagnon J, Pascal M, Scatton B, et al. (2002). Anxiolytic- and antidepressant-like effects of the non-peptide vasopressin V1b receptor antagonist, SSR149415, suggest an innovative approach for the treatment of stress-related disorders. Proceedings of the National Academy of Sciences of the United States of America, 99(9), 6370–6375. 10.1073/pnas.092012099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Hanson DJ (1995). Preventing alcohol abuse: Alcohol, culture, and control. Praeger. [Google Scholar]
  69. Hillard CJ (2014). Stress regulates endocannabinoid-CB1 receptor signaling. Seminars in Immunology, 26(5), 380–388. 10.1016/j.smim.2014.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Hummel M, Cummons T, Lu P, Mark L, Harrison JE, Kennedy JD, et al. (2010). Pain is a salient “stressor” that is mediated by corticotropin-releasing factor-1 receptors. Neuropharmacology, 59(3), 160–166. 10.1016/j.neuropharm.2010.05.001. [DOI] [PubMed] [Google Scholar]
  71. Ikeda K, Kobayashi T, Kumanishi T, Yano R, Sora I, & Niki H (2002). Molecular mechanisms of analgesia induced by opioids and ethanol: Is the GIRK channel one of the keys? Neuroscience Research, 44(2), 121–131. 10.1016/s0168-0102(02)00094-9. [DOI] [PubMed] [Google Scholar]
  72. Ikeda R, Takahashi Y, Inoue K, & Kato F (2007). NMDA receptor-independent synaptic plasticity in the central amygdala in the rat model of neuropathic pain. Pain, 127(1–2), 161–172. 10.1016/j.pain.2006.09.003. [DOI] [PubMed] [Google Scholar]
  73. Itoga CA, Roltsch Hellard EA, Whitaker AM, Lu YL, Schreiber AL, Baynes BB, et al. (2016). Traumatic stress promotes hyperalgesia via corticotropin-releasing factor-1 receptor (CRFR1) signaling in central amygdala. Neuropsychopharmacology, 41, 2463–2472. 10.1038/npp.2016.44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Jakobsson U, Rahm Hallberg I, & Westergren A (2004). Pain management in elderly persons who require assistance with activities of daily living: A comparison of those living at home with those in special accommodations. European Journal of Pain, 8(4), 335–344. 10.1016/j.ejpain.2003.10.007. [DOI] [PubMed] [Google Scholar]
  75. Jakubczyk A, Ilgen MA, Kopera M, Krasowska A, Klimkiewicz A, Bohnert A, et al. (2016). Reductions in physical pain predict lower risk of relapse following alcohol treatment. Drug and Alcohol Dependence, 158, 167–171. 10.1016/j.drugalcdep.2015.11.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Ji G, Fu Y, Adwanikar H, & Neugebauer V (2013). Non-pain-related CRF1 activation in the amygdala facilitates synaptic transmission and pain responses. Molecular Pain, 9, 2. 10.1186/1744-8069-9-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Ji G, Fu Y, Ruppert KA, & Neugebauer V (2007). Pain-related anxiety-like behavior requires CRF1 receptors in the amygdala. Molecular Pain, 3, 13. 10.1186/1744-8069-3-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Ji G, & Neugebauer V (2007). Differential effects of CRF1 and CRF2 receptor antagonists on pain-related sensitization of neurons in the central nucleus of the amygdala. Journal of Neurophysiology, 97(6), 3893–3904. 10.1152/jn.00135.2007. [DOI] [PubMed] [Google Scholar]
  79. Ji G, & Neugebauer V (2008). Pro- and anti-nociceptive effects of corticotropin-releasing factor (CRF) in central amygdala neurons are mediated through different receptors. Journal of Neurophysiology, 99(3), 1201–1212. 10.1152/jn.01148.2007. [DOI] [PubMed] [Google Scholar]
  80. Ji G, & Neugebauer V (2009). Hemispheric lateralization of pain processing by amygdala neurons. Journal of Neurophysiology, 102(4), 2253–2264. 10.1152/jn.00166.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Ji G, & Neugebauer V (2011). Pain-related deactivation of medial prefrontal cortical neurons involves mGluR1 and GABA(A) receptors. Journal of Neurophysiology, 106(5), 2642–2652. 10.1152/jn.00461.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Ji G, Sun H, Fu Y, Li Z, Pais-Vieira M, Galhardo V, et al. (2010). Cognitive impairment in pain through amygdala-driven prefrontal cortical deactivation. The Journal of Neuroscience, 30(15), 5451–5464. 10.1523/JNEUROSCI.0225-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Jochum T, Boettger MK, Burkhardt C, Juckel G, & Bar KJ (2010). Increased pain sensitivity in alcohol withdrawal syndrome. European Journal of Pain, 14(7), 713–718. 10.1016/j.ejpain.2009.11.008. [DOI] [PubMed] [Google Scholar]
  84. Jorgensen HA, & Hole K (1981). Does ethanol stimulate brain opiate receptors? Studies on receptor binding and naloxone inhibition of ethanol-induced effects. European Journal of Pharmacology, 75(4), 223–229. 10.1016/0014-2999(81)90548-3. [DOI] [PubMed] [Google Scholar]
  85. Kampov-Polevoy AB, Kasheffskaya OP, Overstreet DH, Rezvani AH, Viglinskaya IV, Badistov BA, et al. (1996). Pain sensitivity and saccharin intake in alcohol-preferring and -nonpreferring rat strains. Physiology & Behavior, 59(4–5), 683–688. 10.1016/0031-9384(95)02110-8. [DOI] [PubMed] [Google Scholar]
  86. Katz DA, Liu W, Locke C, Dutta S, & Tracy KA (2016). Clinical safety and hypothalamic-pituitary-adrenal axis effects of the arginine vasopressin type 1B receptor antagonist ABT-436. Psychopharmacology, 233(1), 71–81. 10.1007/s00213-015-4089-5. [DOI] [PubMed] [Google Scholar]
  87. Kimpel MW, Brown MM, & Froehlich JC (2003). Pain thresholds in alcohol preferring and non-preferring rats: Diurnal and repeated trial line differences. Alcoholism, Clinical and Experimental Research, 27(12), 1921–1928. [DOI] [PubMed] [Google Scholar]
  88. Kirson D, Oleata CS, Parsons LH, Ciccocioppo R, & Roberto M (2018). CB1 and ethanol effects on glutamatergic transmission in the central amygdala of male and female msP and Wistar rats. Addiction Biology, 23(2), 676–688. 10.1111/adb.12525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Kobayashi T, Ikeda K, Kojima H, Niki H, Yano R, Yoshioka T, et al. (1999). Ethanol opens G-protein-activated inwardly rectifying K+ channels. Nature Neuroscience, 2(12), 1091–1097. 10.1038/16019. [DOI] [PubMed] [Google Scholar]
  90. Kontinen VK, Kauppila T, Paananen S, Pertovaara A, & Kalso E (1999). Behavioural measures of depression and anxiety in rats with spinal nerve ligation-induced neuropathy. Pain, 80(1–2), 341–346. 10.1016/s0304-3959(98)00230-9. [DOI] [PubMed] [Google Scholar]
  91. Koob GF, & Bloom FE (1982). Behavioral effects of neuropeptides: Endorphins and vasopressin. Annual Review of Physiology, 44, 571–582. 10.1146/annurev.ph.44.030182.003035. [DOI] [PubMed] [Google Scholar]
  92. Koob GF, & Le Moal M (2008). Addiction and the brain antireward system. Annual Review of Psychology, 59, 29–53. 10.1146/annurev.psych.59.103006.093548. [DOI] [PubMed] [Google Scholar]
  93. Koob GF, & Volkow ND (2010). Neurocircuitry of addiction. Neuropsychopharmacology, 35(1), 217–238. 10.1038/npp.2009.110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Krieg SM, Sonanini S, Plesnila N, & Trabold R (2015). Effect of small molecule vasopressin V1a and V2 receptor antagonists on brain edema formation and secondary brain damage following traumatic brain injury in mice. Journal of Neurotrauma, 32(4), 221–227. 10.1089/neu.2013.3274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Kwako LE, Spagnolo PA, Schwandt ML, Thorsell A, George DT, Momenan R, et al. (2015). The corticotropin releasing hormone-1 (CRH1) receptor antagonist pexacerfont in alcohol dependence: A randomized controlled experimental medicine study. Neuropsychopharmacology, 40(5), 1053–1063. 10.1038/npp.2014.306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Lachize S, Apostolakis EM, van der Laan S, Tijssen AM, Xu J, de Kloet ER, et al. (2009). Steroid receptor coactivator-1 is necessary for regulation of corticotropin-releasing hormone by chronic stress and glucocorticoids. Proceedings of the National Academy of Sciences of the United States of America, 106(19), 8038–8042. 10.1073/pnas.0812062106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Larson MJ, Paasche-Orlow M, Cheng DM, Lloyd-Travaglini C, Saitz R, & Samet JH (2007). Persistent pain is associated with substance use after detoxification: A prospective cohort analysis. Addiction, 102(5), 752–760. 10.1111/j.1360-0443.2007.01759.x. [DOI] [PubMed] [Google Scholar]
  98. Lawton J, & Simpson J (2009). Predictors of alcohol use among people experiencing chronic pain. Psychology, Health & Medicine, 14(4), 487–501. 10.1080/13548500902923177. [DOI] [PubMed] [Google Scholar]
  99. LeBlanc DM, McGinn MA, Itoga CA, & Edwards S (2015). The affective dimension of pain as a risk factor for drug and alcohol addiction. Alcohol, 49(8), 803–809. 10.1016/j.alcohol.2015.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Lee HS, Lee S, Park S, Baek Y, Youn JH, Cho DB, et al. (2018). The association between somatic and psychological discomfort and health-related quality of life according to the elderly and non-elderly. Quality of Life Research, 27(3), 673–681. 10.1007/s11136-017-1715-1. [DOI] [PubMed] [Google Scholar]
  101. Lee P, Zhang M, Hong JP, Chua HC, Chen KP, Tang SW, et al. (2009). Frequency of painful physical symptoms with major depressive disorder in Asia: Relationship with disease severity and quality of life. The Journal of Clinical Psychiatry, 70(1), 83–91. 10.4088/jcp.08m04114. [DOI] [PubMed] [Google Scholar]
  102. Lewohl JM, Wilson WR, Mayfield RD, Brozowski SJ, Morrisett RA, & Harris RA (1999). G-protein-coupled inwardly rectifying potassium channels are targets of alcohol action. Nature Neuroscience, 2(12), 1084–1090. 10.1038/16012. [DOI] [PubMed] [Google Scholar]
  103. Liu CH, Zhang GZ, Li B, Li M, Woelfer M, Walter M, et al. (2019). Role of inflammation in depression relapse. Journal of Neuroinflammation, 16, 90. 10.1186/s12974-019-1475-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Lolait SJ, Stewart LQ, Jessop DS, Young WS 3rd, & O’Carroll AM (2007). The hypothalamic-pituitary-adrenal axis response to stress in mice lacking functional vasopressin V1b receptors. Endocrinology, 148(2), 849–856. 10.1210/en.2006-1309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Macfarlane GJ, & Beasley M (2015). Alcohol consumption in relation to risk and severity of chronic widespread pain: Results from a UK population-based study. Arthritis Care & Research, 67(9), 1297–1303. 10.1002/acr.22604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Magni G, Salmi A, de Leo D, & Ceola A (1984). Chronic pelvic pain and depression. Psychopathology, 17(3), 132–136. 10.1159/000284030. [DOI] [PubMed] [Google Scholar]
  107. Makino S, Gold PW, & Schulkin J (1994). Corticosterone effects on corticotropin-releasing hormone mRNA in the central nucleus of the amygdala and the parvocellular region of the paraventricular nucleus of the hypothalamus. Brain Research, 640(1–2), 105–112. 10.1016/0006-8993(94)91862-7. [DOI] [PubMed] [Google Scholar]
  108. Manaenko A, Fathali N, Khatibi NH, Lekic T, Hasegawa Y, Martin R, et al. (2011). Arginine-vasopressin V1a receptor inhibition improves neurologic outcomes following an intracerebral hemorrhagic brain injury. Neurochemistry International, 58(4), 542–548. 10.1016/j.neuint.2011.01.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Marmarou CR, Liang X, Abidi NH, Parveen S, Taya K, Henderson SC, et al. (2014). Selective vasopressin-1a receptor antagonist prevents brain edema, reduces astrocytic cell swelling and GFAP, V1aR and AQP4 expression after focal traumatic brain injury. Brain Research, 1581, 89–102. 10.1016/j.brainres.2014.06.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Matsuzawa-Yanagida K, Narita M, Nakajima M, Kuzumaki N, Niikura K, Nozaki H, et al. (2008). Usefulness of antidepressants for improving the neuropathic pain-like state and pain-induced anxiety through actions at different brain sites. Neuropsychopharmacology, 33(8), 1952–1965. 10.1038/sj.npp.1301590. [DOI] [PubMed] [Google Scholar]
  111. McCracken LM, & Dhingra L (2002). A short version of the Pain Anxiety Symptoms Scale (PASS-20): Preliminary development and validity. Pain Research & Management, 7(1), 45–50. 10.1155/2002/517163. [DOI] [PubMed] [Google Scholar]
  112. McCracken LM, Zayfert C, & Gross RT (1992). The Pain Anxiety Symptoms Scale: Development and validation of a scale to measure fear of pain. Pain, 50(1), 67–73. 10.1016/0304-3959(92)90113-p. [DOI] [PubMed] [Google Scholar]
  113. McDermott KA, Joyner KJ, Hakes JK, Okey SA, & Cougle JR (2018). Pain interference and alcohol, nicotine, and cannabis use disorder in a national sample of substance users. Drug and Alcohol Dependence, 186, 53–59. 10.1016/j.drugalcdep.2018.01.011. [DOI] [PubMed] [Google Scholar]
  114. McGinn MA, & Edwards S (2016). CRF1 receptor signaling at the intersection of pain and opioid addiction. In Preedy V (Ed.), Vol. 1. The neuropathology of drug addictions and substance misuse (pp. 891–896). Academic Press. [Google Scholar]
  115. McGovern PE, Mirzoian A, & Hall GR (2009). Ancient Egyptian herbal wines. Proceedings. National Academy of Sciences. United States of America, 106(18), 7361–7366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. McNally GP, & Akil H (2002). Role of corticotropin-releasing hormone in the amygdala and bed nucleus of the stria terminalis in the behavioral, pain modulatory, and endocrine consequences of opiate withdrawal. Neuroscience, 112(3), 605–617. 10.1016/s0306-4522(02)00105-7. [DOI] [PubMed] [Google Scholar]
  117. McWilliams LA, Cox BJ, & Enns MW (2003). Mood and anxiety disorders associated with chronic pain: An examination in a nationally representative sample. Pain, 106(1–2), 127–133. 10.1016/s0304-3959(03)00301-4. [DOI] [PubMed] [Google Scholar]
  118. Means-Christensen AJ, Roy-Byrne PP, Sherbourne CD, Craske MG, & Stein MB (2008). Relationships among pain, anxiety, and depression in primary care. Depression and Anxiety, 25(7), 593–600. 10.1002/da.20342. [DOI] [PubMed] [Google Scholar]
  119. Miller NS, & Gold MS (2007). Opiate prescription medication dependence and pain perceptions. Journal of Addictive Diseases, 26(Suppl. 1), 65–71. 10.1300/J069v26S01_07. [DOI] [PubMed] [Google Scholar]
  120. Mogil JS, Marek P, Yirmiya R, Balian H, Sadowski B, Taylor AN, et al. (1993). Antagonism of the non-opioid component of ethanol-induced analgesia by the NMDA receptor antagonist MK-801. Brain Research, 602(1), 126–130. 10.1016/0006-8993(93)90251-h. [DOI] [PubMed] [Google Scholar]
  121. Morena M, Patel S, Bains JS, & Hill MN (2016). Neurobiological interactions between stress and the endocannabinoid system. Neuropsychopharmacology, 41(1), 80–102. 10.1038/npp.2015.166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Mullin FJ, & Luckhardt AB (1934). The effect of alcohol on cutaneous tactile and pain sensitivity. American Journal of Physiology, 109, 77–78. [Google Scholar]
  123. Naliboff BD, Chang L, Munakata J, & Mayer EA (2000). Towards an integrative model of irritable bowel syndrome. Progress in Brain Research, 122, 413–423. 10.1016/s0079-6123(08)62154-8. [DOI] [PubMed] [Google Scholar]
  124. Narita M, Kaneko C, Miyoshi K, Nagumo Y, Kuzumaki N, Nakajima M, et al. (2006). Chronic pain induces anxiety with concomitant changes in opioidergic function in the amygdala. Neuropsychopharmacology, 31(4), 739–750. 10.1038/sj.npp.1300858. [DOI] [PubMed] [Google Scholar]
  125. Neddenriep B, Bagdas D, Contreras KM, Ditre JW, Wolstenholme JT, Miles MF, et al. (2019). Pharmacological mechanisms of alcohol analgesic-like properties in mouse models of acute and chronic pain. Neuropharmacology, 160, 107793. 10.1016/j.neuropharm.2019.107793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Neugebauer V (2015). Amygdala pain mechanisms. Handbook of Experimental Pharmacology,227, 261–284. 10.1007/978-3-662-46450-2_13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Neugebauer V, Han JS, Adwanikar H, Fu Y, & Ji G (2007). Techniques for assessing knee joint pain in arthritis. Molecular Pain, 3, 8. 10.1186/1744-8069-3-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Neugebauer V, Li W, Bird GC, Bhave G, & Gereau RWT (2003). Synaptic plasticity in the amygdala in a model of arthritic pain: Differential roles of metabotropic glutamate receptors 1 and 5. Journal of Neuroscience, 23(1), 52–63. Retrieved from https://www.ncbi.nlm.nih.gov/pubmed/12514201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Neugebauer V, Li W, Bird GC, & Han JS (2004). The amygdala and persistent pain. The Neuroscientist, 10(3), 221–234. 10.1177/1073858403261077. [DOI] [PubMed] [Google Scholar]
  130. Nijsen M, Ongenae N, Meulemans A, & Coulie B (2005). Divergent role for CRF1 and CRF2 receptors in the modulation of visceral pain. Neurogastroenterology and Motility, 17(3), 423–432. 10.1111/j.1365-2982.2005.00644.x. [DOI] [PubMed] [Google Scholar]
  131. Perrino AC Jr., Ralevski E, Acampora G, Edgecombe J, Limoncelli D, & Petrakis IL (2008). Ethanol and pain sensitivity: Effects in healthy subjects using an acute pain paradigm. Alcoholism, Clinical and Experimental Research, 32(6), 952–958. 10.1111/j.1530-0277.2008.00653.x. [DOI] [PubMed] [Google Scholar]
  132. Poznanski P, Lesniak A, Korostynski M, Szklarczyk K, Lazarczyk M, Religa P, et al. (2017). Delta-opioid receptor antagonism leads to excessive ethanol consumption in mice with enhanced activity of the endogenous opioid system. Neuropharmacology, 118, 90–101. 10.1016/j.neuropharm.2017.03.016. [DOI] [PubMed] [Google Scholar]
  133. Ralevski E, Perrino A, Acampora G, Koretski J, Limoncelli D, & Petrakis I (2010). Analgesic effects of ethanol are influenced by family history of alcoholism and neuroticism. Alcoholism, Clinical and Experimental Research, 34(8), 1433–1441. 10.1111/j.1530-0277.2010.01228.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Richardson HN, Lee SY, O’Dell LE, Koob GF, & Rivier CL (2008). Alcohol self-administration acutely stimulates the hypothalamic-pituitary-adrenal axis, but alcohol dependence leads to a dampened neuroendocrine state. The European Journal of Neuroscience, 28(8), 1641–1653. 10.1111/j.1460-9568.2008.06455.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Richardson HN, Zhao Y, Fekete EM, Funk CK, Wirsching P, Janda KD, et al. (2008). MPZP: A novel small molecule corticotropin-releasing factor type 1 receptor (CRF1) antagonist. Pharmacology, Biochemistry, and Behavior, 88(4), 497–510. 10.1016/j.pbb.2007.10.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Riley JL 3rd, & King C (2009). Self-report of alcohol use for pain in a multi-ethnic community sample. The Journal of Pain, 10(9), 944–952. 10.1016/j.jpain.2009.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Rivier C (2014). Role of hypothalamic corticotropin-releasing factor in mediating alcohol-induced activation of the rat hypothalamic-pituitary-adrenal axis. Frontiers in Neuroendocrinology, 35(2), 221–233. 10.1016/j.yfrne.2013.10.005. [DOI] [PubMed] [Google Scholar]
  138. Roberto M, Cruz MT, Gilpin NW, Sabino V, Schweitzer P, Bajo M, et al. (2010). Corticotropin releasing factor-induced amygdala gamma-aminobutyric acid release plays a key role in alcohol dependence. Biological Psychiatry, 67(9), 831–839. 10.1016/j.biopsych.2009.11.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Roltsch EA, Baynes BB, Mayeux JP, Whitaker AM, Baiamonte BA, & Gilpin NW (2014). Predator odor stress alters corticotropin-releasing factor-1 receptor (CRF1R)-dependent behaviors in rats. Neuropharmacology, 79, 83–89. 10.1016/j.neuropharm.2013.11.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Roltsch Hellard EA, Impastato RA, & Gilpin NW (2017). Intra-cerebral and intra-nasal melanocortin-4 receptor antagonist blocks withdrawal hyperalgesia in alcohol-dependent rats. Addiction Biology, 22(3), 692–701. 10.1111/adb.12360. [DOI] [PubMed] [Google Scholar]
  141. Roueche B (1963). Alcohol in human culture. In Salvatore LP (Ed.), Alcohol and civilization (pp. 167–182). New York: McGraw-Hill. [Google Scholar]
  142. Rouwette T, Vanelderen P, de Reus M, Loohuis NO, Giele J, van Egmond J, et al. (2012). Experimental neuropathy increases limbic forebrain CRF. European Journal of Pain, 16(1), 61–71. 10.1016/j.ejpain.2011.05.016. [DOI] [PubMed] [Google Scholar]
  143. Ryan ML, Falk DE, Fertig JB, Rendenbach-Mueller B, Katz DA, Tracy KA, et al. (2017). A phase 2, double-blind, placebo-controlled randomized trial assessing the efficacy of ABT-436, a novel V1b receptor antagonist, for alcohol dependence. Neuropsychopharmacology, 42(5), 1012–1023. 10.1038/npp.2016.214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Sacharczuk M, Lesniak A, Lipkowski AW, Korostynski M, Przewlocki R, & Sadowski B (2014). Association between the A107V substitution in the delta-opioid receptors and ethanol drinking in mice selected for high and low analgesia. Addiction Biology, 19(4), 643–651. 10.1111/adb.12030. [DOI] [PubMed] [Google Scholar]
  145. Saddler JM, James MF, & Harington AP (1985). Naloxone does not reverse ethanol analgesia in man. Clinical and Experimental Pharmacology & Physiology, 12(4), 359–364. 10.1111/j.1440-1681.1985.tb00883.x. [DOI] [PubMed] [Google Scholar]
  146. Salome N, Stemmelin J, Cohen C, & Griebel G (2006). Differential roles of amygdaloid nuclei in the anxiolytic- and antidepressant-like effects of the V1b receptor antagonist, SSR149415, in rats. Psychopharmacology, 187(2), 237–244. 10.1007/s00213-006-0424-1. [DOI] [PubMed] [Google Scholar]
  147. Schreiber AL, & Gilpin NW (2018). Corticotropin-releasing factor (CRF) neurocircuitry and neuropharmacology in alcohol drinking. Handbook of Experimental Pharmacology, 248, 435–471. 10.1007/164_2017_86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  148. Schwartz N, Temkin P, Jurado S, Lim BK, Heifets BD, Polepalli JS, et al. (2014). Chronic pain. Decreased motivation during chronic pain requires long-term depression in the nucleus accumbens. Science, 345(6196), 535–542. 10.1126/science.1253994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  149. Schwetz I, Bradesi S, McRoberts JA, Sablad M, Miller JC, Zhou H, et al. (2004). Delayed stress-induced colonic hypersensitivity in male Wistar rats: Role of neurokinin-1 and corticotropin-releasing factor-1 receptors. American Journal of Physiology. Gastrointestinal and Liver Physiology, 286(4), G683–G691. 10.1152/ajpgi.00358.2003. [DOI] [PubMed] [Google Scholar]
  150. Serrano A, Pavon FJ, Buczynski MW, Schlosburg J, Natividad LA, Polis IY, et al. (2018). Deficient endocannabinoid signaling in the central amygdala contributes to alcohol dependence-related anxiety-like behavior and excessive alcohol intake. Neuropsychopharmacology, 43(9), 1840–1850. 10.1038/s41386-018-0055-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. Shurman J, Koob GF, & Gutstein HB (2010). Opioids, pain, the brain, and hyperkatifeia: A framework for the rational use of opioids for pain. Pain Medicine, 11(7), 1092–1098. 10.1111/j.1526-4637.2010.00881.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Siqueira-Campos VME, Da Luz RA, de Deus JM, Martinez EZ, & Conde DM (2019). Anxiety and depression in women with and without chronic pelvic pain: prevalence and associated factors. Journal of Pain Research, 12, 1223–1233. 10.2147/JPR.S195317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  153. Spierling SR, & Zorrilla EP (2017). Don’t stress about CRF: Assessing the translational failures of CRF1 antagonists. Psychopharmacology, 234(9–10), 1467–1481. 10.1007/s00213-017-4556-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. Stemmelin J, Lukovic L, Salome N, & Griebel G (2005). Evidence that the lateral septum is involved in the antidepressant-like effects of the vasopressin V1b receptor antagonist, SSR149415. Neuropsychopharmacology, 30(1), 35–42. 10.1038/sj.npp.1300562. [DOI] [PubMed] [Google Scholar]
  155. Stewart SH, Finn PR, & Pihl RO (1995). A dose-response study of the effects of alcohol on the perceptions of pain and discomfort due to electric shock in men at high familial-genetic risk for alcoholism. Psychopharmacology, 119(3), 261–267. 10.1007/BF02246289. [DOI] [PubMed] [Google Scholar]
  156. Sun H, & Neugebauer V (2011). mGluR1, but not mGluR5, activates feed-forward inhibition in the medial prefrontal cortex to impair decision making. Journal of Neurophysiology, 106(2), 960–973. 10.1152/jn.00762.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  157. Swanson LW, Sawchenko PE, Rivier J, & Vale WW (1983). Organization of ovine corticotropin-releasing factor immunoreactive cells and fibers in the rat brain: An immunohistochemical study. Neuroendocrinology, 36(3), 165–186. 10.1159/000123454. [DOI] [PubMed] [Google Scholar]
  158. Tasker JG, Chen C, Fisher MO, Fu X, Rainville JR, & Weiss GL (2015). Endocannabinoid regulation of neuroendocrine systems. International Review of Neurobiology, 125, 163–201. 10.1016/bs.irn.2015.09.003. [DOI] [PubMed] [Google Scholar]
  159. Thompson T, Oram C, Correll CU, Tsermentseli S, & Stubbs B (2017). Analgesic effects of alcohol: A systematic review and meta-analysis of controlled experimental studies in healthy participants. The Journal of Pain, 18(5), 499–510. 10.1016/j.jpain.2016.11.009. [DOI] [PubMed] [Google Scholar]
  160. Tribollet E, Raufaste D, Maffrand J, & Serradeil-Le Gal C (1999). Binding of the non-peptide vasopressin V1a receptor antagonist SR-49059 in the rat brain: An in vitro and in vivo autoradiographic study. Neuroendocrinology, 69(2), 113–120. 10.1159/000054409. [DOI] [PubMed] [Google Scholar]
  161. Varodayan FP, Correia D, Kirson D, Khom S, Oleata CS, Luu G, et al. (2017). CRF modulates glutamate transmission in the central amygdala of naïve and ethanol-dependent rats. Neuropharmacology, 125, 418–428. 10.1016/j.neuropharm.2017.08.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  162. Varodayan FP, Soni N, Bajo M, Luu G, Madamba SG, Schweitzer P, et al. (2016). Chronic ethanol exposure decreases CB1 receptor function at GABAergic synapses in the rat central amygdala. Addiction Biology, 21(4), 788–801. 10.1111/adb.12256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Veinante P, & Freund-Mercier MJ (1997). Distribution of oxytocin- and vasopressin-binding sites in the rat extended amygdala: A histoautoradiographic study. The Journal of Comparative Neurology, 383(3), 305–325. Retrieved from https://www.ncbi.nlm.nih.gov/pubmed/9205043. [PubMed] [Google Scholar]
  164. Vendruscolo LF, Estey D, Goodell V, Macshane LG, Logrip ML, Schlosburg JE, et al. (2015). Glucocorticoid receptor antagonism decreases alcohol seeking in alcohol-dependent individuals. The Journal of Clinical Investigation, 125(8), 3193–3197. 10.1172/JCI79828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  165. Volpi S, Rabadan-Diehl C, & Aguilera G (2004). Vasopressinergic regulation of the hypothalamic pituitary adrenal axis and stress adaptation. Stress, 7(2), 75–83. 10.1080/10253890410001733535. [DOI] [PubMed] [Google Scholar]
  166. von Knorring L, Perris C, Eisemann M, Eriksson U, & Perris H (1983). Pain as a symptom in depressive disorders. II. Relationship to personality traits as assessed by means of KSP. Pain, 17(4), 377–384. 10.1016/0304-3959(83)90169-0. [DOI] [PubMed] [Google Scholar]
  167. Watson D, & Tellegen A (1985). Toward a consensual structure of mood. Psychological Bulletin, 98(2), 219–235. 10.1037//0033-2909.98.2.219. [DOI] [PubMed] [Google Scholar]
  168. Watters JJ, Swank MW, Wilkinson CW, & Dorsa DM (1996). Evidence for glucocorticoid regulation of the rat vasopressin V1a receptor gene. Peptides, 17(1), 67–73. 10.1016/0196-9781(95)02085-3. [DOI] [PubMed] [Google Scholar]
  169. Williams LS, Jones WJ, Shen J, Robinson RL, Weinberger M, & Kroenke K (2003). Prevalence and impact of depression and pain in neurology outpatients. Journal of Neurology, Neurosurgery, and Psychiatry, 74(11), 1587–1589. 10.1136/jnnp.74.11.1587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  170. Witkiewitz K, Vowles KE, McCallion E, Frohe T, Kirouac M, & Maisto SA (2015). Pain as a predictor of heavy drinking and any drinking lapses in the COMBINE study and the UK Alcohol Treatment Trial. Addiction, 110(8), 1262–1271. 10.1111/add.12964. [DOI] [PMC free article] [PubMed] [Google Scholar]
  171. Wolff HG, Hardy JD, & Goodell H (1942). Studies on pain: Measurement of the effect of ethyl alcohol on the pain threshold and on the “alarm” reaction. Journal de Pharmacologie, 75, 38–49. [Google Scholar]
  172. Woodhams SG, Chapman V, Finn DP, Hohmann AG, & Neugebauer V (2017). The cannabinoid system and pain. Neuropharmacology, 124, 105–120. 10.1016/j.neuropharm.2017.06.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  173. Woodrow KM, & Eltherington LG (1988). Feeling no pain: Alcohol as an analgesic. Pain, 32(2), 159–163. 10.1016/0304-3959(88)90064-4. [DOI] [PubMed] [Google Scholar]
  174. Yao M, & Denver RJ (2007). Regulation of vertebrate corticotropin-releasing factor genes. General and Comparative Endocrinology, 153(1–3), 200–216. 10.1016/j.ygcen.2007.01.046. [DOI] [PubMed] [Google Scholar]
  175. Yu W, Hwa LS, Makhijani VH, Besheer J, & Kash TL (2019). Chronic inflammatory pain drives alcohol drinking in a sex-dependent manner for C57BL/6J mice. Alcohol, 77, 135–145. 10.1016/j.alcohol.2018.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  176. Zale EL, Maisto SA, & Ditre JW (2015). Interrelations between pain and alcohol: An integrative review. Clinical Psychology Review, 37, 57–71. 10.1016/j.cpr.2015.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  177. Zhang Z, Tao W, Hou YY, Wang W, Lu YG, & Pan ZZ (2014). Persistent pain facilitates response to morphine reward by downregulation of central amygdala GABAergic function. Neuropsychopharmacology, 39(9), 2263–2271. 10.1038/npp.2014.77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  178. Zhao Y, Valdez GR, Fekete EM, Rivier JE, Vale WW, Rice KC, et al. (2007). Subtype-selective corticotropin-releasing factor receptor agonists exert contrasting, but not opposite, effects on anxiety-related behavior in rats. The Journal of Pharmacology and Experimental Therapeutics, 323(3), 846–854. 10.1124/jpet.107.123208. [DOI] [PubMed] [Google Scholar]
  179. Zhou Y, Colombo G, Carai MA, Ho A, Gessa GL, & Kreek MJ (2011). Involvement of arginine vasopressin and V1b receptor in alcohol drinking in Sardinian alcohol-preferring rats. Alcoholism, Clinical and Experimental Research, 35(10), 1876–1883. 10.1111/j.1530-0277.2011.01532.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  180. Zhou Y, & Kreek MJ (2014). Alcohol: A stimulant activating brain stress responsive systems with persistent neuroadaptation. Neuropharmacology, 87, 51–58. 10.1016/j.neuropharm.2014.05.044. [DOI] [PubMed] [Google Scholar]

RESOURCES