Abstract
Alcohol Use Disorder (AUD) remains a challenging condition with limited effective treatment options; however new technology in drug delivery and advancements in pharmacology have paved the way for discovery of novel therapeutic targets. This review explores emerging pharmacological targets that offer new options for the management of AUD, focusing on the potential of somatostatin (SST), vasoactive intestinal peptide (VIP), glucagon-like peptide-1 (GLP-1), nociceptin (NOP), and neuropeptide S (NPS). These targets have been selected based on recent advancements in preclinical and clinical research, which suggest their significant roles in modulating alcohol consumption and related behaviors. SST dampens cortical circuits, and targeting both the SST neurons and the SST peptide itself presents promise for treating AUD and various related comorbidities. VIP neurons are modulated by alcohol and targeting the VIP system presents an unexplored avenue for addressing alcohol exposure at various stages of development. GLP-1 interacts with the dopaminergic reward system and reduces alcohol intake. Nociceptin modulates mesolimbic circuitry and agonism and antagonism of nociceptin receptor offers a complex but promising approach to reducing alcohol consumption. NPS stands out for its anxiolytic-like effects, particularly relevant for the anxiety associated with AUD. This review aims to synthesize the current understanding of these targets, highlighting their potential in developing more effective and personalized AUD therapies, and underscores the importance of continued research in identifying and validating novel targets for treatment of AUD and comorbid conditions.
Graphical Abstract
This review explores emerging pharmacological targets that offer new options for the management of AUD, focusing on the potential of somatostatin (SST), glucagon-like peptide-1 (GLP-1), nociceptin, neuropeptide S (NPS), and vasoactive intestinal peptide (VIP).

INTRODUCTION
Despite extensive efforts in prevention and intervention, Alcohol Use Disorder (AUD) remains a pervasive and challenging condition to treat1,2. AUD is a major public health issue, affecting over one-tenth of Americans aged 12 and older in 2020, with a notable increase following the Covid-19 pandemic3–6. AUD, as defined by the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition (DSM-5), is characterized by a pattern of alcohol use leading to significant impairment or distress7. In the United States, 78% of individuals over the age of 12 have consumed alcohol, with similar consumption rates among both men and women7. Alarmingly, about 21% engage in problematic forms of alcohol consumption, such as binge drinking, and nearly 6% report heavy drinking. The economic impact of alcohol misuse is staggering, estimated at $249 billion in 2010, a figure that has likely grown in the wake of increased alcohol consumption during the pandemic6,7. Furthermore, the National Institute on Alcohol Abuse and Alcoholism (NIAAA) has identified an increasing trend of high intensity drinking7,8. The lack of reliable biomarkers for AUD impedes the development and testing of new treatments9, and despite significant research investments by the National Institutes of Health (NIH) and others in understanding the brain mechanisms of AUD, the last decade has seen a scarcity of new FDA-approved treatments for this disorder10. Importantly, binge drinking among women is a rapidly growing public health crisis, with both binge and heavy alcohol use among women surging over the past decade, posing a substantial public health threat11–16
Advancements in understanding the neurocircuitry and neuromodulators involved in alcohol misuse and the transition to dependence have provided valuable insights17–24, yet there remains a critical need for further exploration of novel pharmacological targets. This exploration is essential to address the complex nature of AUD and to develop new treatment options that can effectively mitigate the multifaceted impact of alcohol consumption25,26. The current pharmacotherapies for AUD, while moderately effective, are primarily focused on specific stages of the disorder, such as reducing consumption or aiding in maintaining abstinence27. However, these treatments often fall short in addressing the broader spectrum of AUD-related behaviors and symptoms – for example, cognitive decline28, inflammation29,30, mitochondrial damage31–34, and liver dysfunction35. Targets such as corticotrophin releasing factor36–41, neuropeptide y42–44, and dynorphin45 have been reviewed at length46 – therefore here we speculate on interesting emerging novel targets that we believe warrant further investigation. While many other emerging targets exist, we have focused on somatostatin (SST), vasoactive intestinal peptide (VIP), glucagon-like peptide 1 (GLP-1), nociceptin, and neuropeptide S (NPS).
Neuropeptides, a diverse class of signaling molecules, are increasingly recognized for their critical roles in modulating brain function and behavior, particularly in responses to stress, reward, and addiction47. Unlike classical neurotransmitters, neuropeptides act on g protein coupled receptors (GPCRs), which offers unique opportunities to fine-tune neural circuitry rather than simply turning it on or off. This makes neuropeptides attractive targets for therapeutic intervention. SST, VIP, GLP-1, nociceptin, and NPS are all neuropeptides which modulate neural circuits implicated in AUD. These neuropeptides influence a wide range of alcohol-related behaviors, including anxiety, craving, and reinforcement, which are critical aspects of AUD. Furthermore, the potential of these neuropeptides to serve as biomarkers for AUD is significant; their measurable presence in peripheral systems, such as blood or cerebrospinal fluid, could provide crucial indicators for diagnosis and treatment efficacy. Given the current lack of reliable biomarkers for AUD, investigating these neuropeptides could pave the way for more targeted and personalized therapeutic approaches. Emphasizing these targets highlights their dual role in advancing our understanding of AUD’s neurobiological underpinnings and addressing the urgent need for innovative and effective treatment strategies. Importantly, preclinical work investigating the therapeutic efficacy of pharmacological targets of alcohol use often ranges from free-choice consumption drinking models, to those incorporating forced exposure such as vaporized ethanol and intragastric delivery, with varying degrees of social isolation and other stressors48. Differences in ethanol exposure and consumption models must be considered when accounting for differences in behavioral efficacy.
SOMATOSTATIN
Somatostatin (SST; also abbreviated as SRIF) neurons play a critical role in the central nervous system, functioning as key modulators of neurotransmission and neuroendocrine signaling49–53. These neurons, which produce the inhibitory neuropeptide somatostatin, are widely distributed across various brain regions, including the cortex, hippocampus, and amygdala54–56. SST exists in two primary isoforms, SST-14 and SST-28, and exerts its effects through activation of 5 G-protein-coupled somatostatin receptors (SSTRs) largely thought to be Gi/o coupled52,57–59 and further reviewed in Brockway & Crowley (2020) Known for their inhibitory action on the release of hormones and neurotransmitters, SST neurons are integral to regulating neuronal excitability and synaptic plasticity and have gained attention for their role in substance use disorders49–52,60,61. SST is released from SST neurons62 and acts to dampen cortical signaling in a long-lasting manner through activation of SST receptors63. The involvement of SST neurons and SST in a range of physiological processes including neurological conditions highlights its importance53 and potential in modulating other states involving the cortex.
Both SST neurons and their namesake neuropeptide are promising therapeutic pursuits for AUD. SST neurons within the prefrontal cortex (PFC)61,64,65 and regions of the amygdala65–67 display robust alterations from multiple animal models of alcohol exposure and consumption. Glutamatergic drive onto PFC SST neurons is reduced in early abstinence from alcohol65,68. Consistent with this effect, the intrinsic excitability of SST neurons is reduced in abstinence from alcohol (24 hours after 4 weeks of the DID procedure)64. The specific effects of alcohol on SST neurons in the PFC may be dose69 and receptor dependent70, and adolescent-exposure specific changes may emerge due to the unique circuitry of the developmental brain61,71,72. Further investigation into alcohol modulation of SST neurons throughout development in the cortex and elsewhere is necessary. Alcohol consumption reduces SST immunoreactivity70,73 further supporting the role for these neurons, and potentially the peptide itself, in sensitivity to alcohol. GPCRs uniquely expressed on SST neurons such as mGluRs74 may provide a promising route for pharmacological restoration of SST neuron function following alcohol75. For instance, mGlu1 metabotropic glutamate receptors are enriched in SST-INs and potentiate excitatory transmission on SST neurons but not neighboring pyramidal cells75. Therefore, mGlu1 agonists may serve to counteract the reduced glutamatergic drive and intrinsic excitability found in SST neurons in early abstinence from alcohol exposure. Chemogenetic (GPCR based) manipulations of PFC SST neurons have also been shown to decrease binge drinking (DID), further supporting a role for SST GPCR based interventions for AUD64. Exploring whether neuropeptide-expressing neuronal populations can be uniquely pharmacologically targeted due to their unique GPCR expression would allow for restoration of circuit-deficits with minimal off-target effects75.
In addition, emerging evidence suggests SST may be a therapeutic target itself. Modulating SST receptor activation in the PFC can alter exploratory and anxiety-like behaviors related to alcohol consumption64. Modulation of SST through site-specific injection aimed at the amygdala (of both SST and SSTR4 agonists, which are of particular note due to their expression in addiction-related brain regions such as the prefrontal cortex and limbic system) can reduce binge drinking70. Further work on the peptidergic promise of SST should focus on sex differences64 as well as SST’s potential additional actions as a vasoconstrictor in the central nervous system76. Studies have shown that the response of SST neurons to alcohol and the impact of SST on behavior can vary significantly between males and females63,64. This variation highlights the importance of considering sex as a critical factor in developing SST- and SSTR-targeted therapies for AUD53. Additionally, the broader physiological role of SST, including its potential to modulate non neuronal cell types77 within the central nervous system warrants further exploration.
New translational clinical work has further enthused SST’s relevance in AUD. Cortical SST gene expression strongly correlates with alcohol-induced changes in resting-state activity in a protective-fashion in healthy men78. SST gene expression is also reduced in the hippocampus of individuals with broader substance use disorders79. While SST has a strong correlation with multiple psychiatric disorders53, this emerging clinical literature suggests its potential unique role in substance misuse and alcohol misuse – further enthusing it as a potential target. Multiple FDA-approved analogues of SST exist (e.g., Octreotide, Sandostatin) and have modest risk profiles80–82. The SST analogue Octreotide has long been used to mitigate alcohol-induced hypotension83 in patients, and in animal models, may reduce alcohol-mediated neuropathic pain84, and alcohol induced gastric damage85 suggesting further therapeutic relevance for treating AUD simultaneously with comorbid conditions.
VASOACTIVE INTESTINAL PEPTIDE
Vasoactive intestinal peptide (VIP) is a 28 amino acid86 neuropeptide in the neocortex, it is predominantly expressed in GABAergic neurons and modulates cortical circuits through its G-protein coupled receptors VPAC1R and VPAC2R87–90. VPAC1R and VPAC2R are expressed on neurons and immune cells91 and VIP acts directly on both92. VIP reduces inflammation93,94, acts as a vasodilator95, and modulates insulin and somatostatin release96. It is known in the central nervous system for its involvement in the circadian cycle governed by the suprachiasmatic nucleus97. High levels of VIP are expressed in emotion and addiction related regions such as the frontal cortex, amygdala, hypothalamus and hippocampus98. VIP is thought to depolarize neurons and activate a hyperpolarization-activated cation current through hyperpolarization-activated cyclic-nucleotide-gated channels99. While pituitary adenylate-cyclase-activating polypeptide (PACAP), also activates VPAC1R and VPAC2R, VIP and PCAP are pharmacologically distinct100. Targeting PACAP has shown promise in modulating alcohol drinking behaviors101–105 further supporting a role for VIP and VPAC1R and VPAC2R as emerging targets for AUD164. Further, VPAC1R null mice show elevated GLP-1, suggesting an interaction between VIP/VPAC1R and GLP-1 (reviewed in the following section)106. In addition to their role in the suprachiasmatic nucleus, VIP neurons are a major subset of 5HT3 serotonin receptor expressing neurons and are located throughout the superficial layers of the neocortex107–110. They are known to release both GABA and VIP111. VIP neurons primarily target somatostatin (SST) neurons, playing a vital role in modulating cortical excitability and output through a disinhibitory mechanism55,112,113.
The influence of VIP extends to a significant interaction with alcohol. Alcohol is known to increase plasma levels of VIP114, suggesting alcohol may stimulate VIP release. VIP-stimulated release of β-endorphin is affected by alcohol115. Genetic studies have identified a link between VIP gene polymorphisms and increased alcohol consumption116. Further alcohol exposure has been shown to decrease expression of VIP. Long-term exposure to alcohol and withdrawal significantly reduces the synthesis and expression VIP in rats, with a notable decrease in VIP mRNA levels in the suprachiasmatic nucleus117. Neonatal exposure to alcohol can lead to a reduction in the density of VIP neurons in the rat suprachiasmatic nucleus118, indicating potential circadian disruptions and a increased vulnerable to early alcohol exposure. This relationship between VIP and alcohol has been extended to animal models, where VIP has been shown to affect the development of alcohol tolerance119 however, further behavioral testing on the effects of VIP on alcohol consumption in both males and females are warranted.
Alcohol has also been shown to modulate VIP neurons. A recent study found that bath application of alcohol in cortical slices activates cortical VIP neurons, while chronic alcohol consumption in mice can lead to a decrease in the excitability of VIP neurons, a potential compensatory mechanism120. Further, VIP neurons in the cortex of male mice were found to remain hypoactive in withdrawal while females return to normal levels, suggesting sex differences in recovery of VIP signaling during withdrawal120. Increased VIP neuron activity following exposure to acute alcohol may correspond to increased VIP releaese. Modulation of VIP neuron activity by alcohol highlights the intricate relationship between VIP and alcohol exposure and underscores the importance of future investigations into the effect of alcohol and withdrawal on VIP neurons throughout development and across sexes.
The relationship between VIP and alcohol consumption suggests potential therapeutic applications. VIP may cross the blood brain barrier121, and delivery to the brain can be enhanced using intranasal administration of VIP incorporated into nanoparticles122. Like SST, targeting VIP neurons also presents promise in reducing alcohol consumption. Since VIP-INs express 5-HT3 receptors, and their antagonism reduces alcohol consumption123, targeting VIP neurons through other receptors for instance GPCRs expressed on VIP neurons offers a promising alternative to 5-HT3 antagonists. Moreover, targeting VIP presents promise as an unexplored target for AUD and comorbid conditions such as diabetes and inflammatory diseases, as well as protective against the effects of early alcohol exposure. Multifaceted approaches targeting VIP neurons, VIP release, and VPAC1 and 2 could lead to novel and effective treatments for alcohol-related disorders. The largely unexplored role of the VIP system in alcohol use, circuit modulation, and behavior, underscores the importance of future studies into VIP needed to further establish its role as a therapeutic target for Alcohol Use Disorder.
GLUCAGON-LIKE PEPTIDE 1
Glucagon-like peptide-1 (GLP-1)- and its g-protein coupled receptor (GLP-1R)- targeting drugs, widely recognized for their benefits in diabetes and weight management, have also sparked significant scientific interest due to their potential impact on the neurocircuitry of food intake and drug reward124,125. Secreted by the gut, GLP-1 is a 30 amino acid peptide126 predominantly known for its role in stimulating insulin secretion and inhibiting glucose secretion127. GLP-1 produced in the central nervous system itself has been implicated in inducing satiety128, stress responses129, and playing a role in the regulation of glucose levels130. GLP-1Rs are found throughout the brain with high levels in the hypothalamus and brainstem131. However, its overall pharmacological profile both in the periphery and the brain are not fully known.
Emerging research has revealed that peripheral injections of GLP-1 or an analogue reduced alcohol intake in male rats, an effect replicated by direct administration of these drugs to the ventral tegmental area (VTA)132 – broadly suggesting an interaction with dopaminergic reward circuitry. These findings have been extended to mice in thematically complementary studies133, though notably, more work is needed to establish the role of GLP-1 in females. It is important to consider off target effects of GLP-1 especially in patients who do not have diabetes and are using it for weight loss. The co-reduction in food intake along with alcohol intake134 may limit or enhance its treatment efficacy in various clinical populations and should be specifically and thoroughly characterized.
Similar to SST, the human clinical literature has provided corresponding support for GLP-1 as a target for AUD. Variability in GLP-1 receptor gene variants is correlated with functional connectivity measurements in individuals with high or low risk for AUD135. Further comprehensive work by the same group has shown not only an interaction between GLP-1R expression levels and individuals with AUD (with greater receptor mRNA expression in the hippocampus of postmortem brains of individuals with AUD), but that intravenous alcohol administration reduced GLP-1 levels136. Importantly, as GLP-1R targeting drugs are now rapidly available in the global market137–139, correlational studies of alcohol use in these populations will likely emerge with time and provide further important insight. GLP-1’s multifaceted role in metabolic regulation and its emergent impact on reward pathways underscore its potential as a therapeutic target for disorders extending beyond diabetes, including AUD, warranting further research to fully harness its clinical applications.
NOCICEPTIN
Nociceptin (also known as orphanin FQ) is a unique member of the opioid peptide family, binding to the nociceptin opioid peptide (NOP) receptor (originally named opioid receptor-like 1)140–142. It is far less investigated as compared to mu- delta- and kappa- opioid systems. Nociceptin is 17 amino acids long143, and is involved in various neural processes, ranging from pain modulation144 to behavioral responses to stress145 and reward mechanisms146. Although it shares structural features with classical opioids, nociceptin’s effects are often distinct, indicating a complex role in both normal physiology and the pathophysiology of addiction147. Nociceptin mediates consumption of other natural liquid rewards148, and likely alters the rewarding properties for multiple drugs149–154.
Increasing evidence suggests nociceptin agonism and antagonism can reduce alcohol consumption. While intracerebroventricular administration of nociceptin increases alcohol intake, indicating a facilitative effect on alcohol’s rewarding properties, repeated exposure to nociceptin leads to a decrease in alcohol consumption155. This dichotomy suggests a sophisticated interaction between nociceptin and the reinforcing attributes of alcohol, pointing to potential receptor specific effects. Later work by the same group showed nociceptin analogues reduce alcohol intake in males153. Another study found that nociceptin agonists reduce alcohol consumption and prevented reinstatement and withdrawal symptoms in alcohol preferring rats156. A separate study corroborated these results indicating nociceptin reduces withdrawal symptoms and anxiety-like behaviors in rats with acute and chronic alcohol intoxication157. Similar results were obtained in the rostromedial tegmental nucleus where nociceptin signaling reduced alcohol consumption and attenuated anxiety- and depressive- like behaviors in alcohol withdrawn rats158. Dysregulation of nociceptin receptor signaling in the central amygdala (CeA) contributes to excessive alcohol intake and is rescued by local micro-infusion of nociceptin159.
Interestingly, peripheral injections of NOP receptor antagonists can also reduce alcohol consumption, stress-induced alcohol seeking, and alcohol-induced stimulation of brain reward pathways in alcohol preferring rats160. The effects of NOP receptor antagonism in the brain may depend on the precise brain region compounds are targeted to, with reductions in consumption seen following micro-injections to the CeA and VTA, but not nucleus accumbens (NAc), of male and female alcohol preferring rats 161. A similar separate study from this same group found that antagonism of NOP receptors in the VTA and the CeA, but not in the NAc reduced alcohol seeking162. These studies highlight the complex bidirectional role of nociceptin in alcohol use and point to its role in particular key addiction related regions such as the CeA and VTA. Future work should address the role of NOP receptor agonism and antagonism in the same study. Overall this literature suggests that alcohol intake might downregulate the endogenous nociceptin system, disrupting stress regulation and enhancing alcohol consumption147. The role of both NOP receptor agonists and antagonists in decreasing alcohol consumption could be due to receptor desensitization or differential modulation of brain regions associated with reward and addiction, indicating a need for further region-specific and mechanistic research to precisely deliver treatments for AUD.
Recently, clinical studies have emerged in the literature for nociception antagonists and AUD, with antagonists advancing to clinical trials with great promise163. NOP specific agonists and partial agonists have shown promise in primates164. Buprenorphine, a semi-synthetic derivative that functions as a partial agonist at μ-opioid and NOP receptors, reduces alcohol consumption in monkeys, is effective in treating opioid use disorder165,166, and has been used off-label for depression167, suggesting it may be beneficial in AUD with comorbid depression. Further research is warranted to investigate the potential of NOP receptors in AUD treatment, potential for mitigating alcohol abuse, and sex specific effects. These and related compounds may be particularly useful in individuals with comorbid depression and AUD168.
NEUROPEPTIDE S
Neuropeptide S (NPS), a recently identified peptide, is notable for its distinctive behavioral effects, including anxiolytic-like actions and the promotion of wakefulness169–171 (for review on NPS see172,173). NPS is a 20 amino acid neuropeptide174 which binds to and activates the NPS receptor (NPSR), a previously orphan G-protein coupled receptor known as GPR154 or GPRA175,176. NPS is found in brain areas including the parabrachial region and the locus coeruleus 172, and the NPSR is unique in that it is an excitatory GPCR (Gαs and Gαq) leading to elevated intracellular levels of cAMP and calcium175,176. NPS is colocalized with excitatory neurotransmitters, further supporting the view that NPS is part of an excitatory signaling system.
In the context of AUD, NPS and its receptor, NPSR, are integral to the brain’s response to alcohol and associated anxiety. Variations in the NPSR1 gene are linked to the risk and patterns of AUD, showing notable differences based on sex and age177,178. Several preclinical studies have indicated that delivery of NPS (both intraperitoneal and intracerebroventricular) can decrease alcohol seeking in mice; however, some have shown conflicting results172 likely having to do with comorbid variables such as stress and anxiety. The deletion of the NPSR gene in mice reduces sensitivity to the effects of alcohol, while intracerebroventricularly administered NPS can diminish alcohol effects in mice179. NPS may also be affected by alcohol with marked increases in expression during withdrawal 180. Rats subjected to a week of alcohol intoxication exhibit increased NPSR gene expression in various brain areas, with more pronounced changes observed after 7 days of withdrawal180. Functionally, this upregulation correlates with enhanced anxiolytic effects of NPS180. An increase in NPSR during withdrawal from alcohol suggests a possible role in alleviating associated anxiety, a theory supported by genetic studies linking NPSR variations to anxiety traits in AUD178. Chronic alcohol exposure enhances the anxiolytic and anti-depressive effects of NPS181 further supporting the potential of NPSR to reduce anxiety experienced in AUD patients. These studies indicate that NPS not only modulates anxiety but also is involved with and potentiated by alcohol, future studies are needed to offer insights into behavioral effect of NPS both during alcohol consumption and during withdrawal.
Intriguingly, NPS’s impact on alcohol consumption varies across genetic lines172 (known to have varying basal anxiety levels and alcohol preference) as best exemplified in the differential responses seen between alcohol-preferring (msP) rats and Wistar rats182,183. While NPS reduces alcohol intake in msP rats, known for their excessive alcohol consumption and anxiety-like behaviors, it promotes alcohol-seeking behavior in Wistar rats. NPS likely reduces alcohol consumption through anxiolytic-like effects, suggesting NPS may be most useful in treating AUD populations with comorbid anxiety. These studies underscore the complexity of NPS action and highlights the necessity of considering genetics, anxiety levels, and behavioral context in its potential therapeutic application for AUD.
Targeting the NPS/NPSR system holds significant promise for AUD treatment. The first NPSR specific agonist was only recently developed184, which will hopefully spur greater clinical interests. Future developments in this area could help researchers elucidate a role for the NPSR in alcohol consumption and withdrawal symptoms. This approach could effectively tackle the challenges such as anxiety experienced during withdrawal and relapse, major obstacles in AUD therapy. NPS’s capacity to regulate stress and anxiety responses, along with its enhanced efficacy in alcohol-withdrawn subjects181 indicates that targeting this system could be highly beneficial in addressing AUD’s mood-related complexities. Continued research is vital to enhance our understanding of NPS’s impact on AUD and to create specialized treatments that leverage this neuropeptide system’s therapeutic potential.
DISCUSSION AND FUTURE DIRECTIONS
The complexity of AUD, frequently presenting alongside other psychiatric disorders, highlights the urgent need for novel ligand and receptor targets to mitigate the diverse impacts of this disorder. Ideally, emerging treatments will focus not only on addiction management but also address the underlying physiological damage and co-occurring symptoms of AUD. It is crucial to also assess the addiction potential of new therapies themselves, and to consider sex-specific responses and genetic diversity. Additional priorities should include evaluating long-term health effects, managing side effects, and specialized treatments for vulnerable populations. Researchers must overcome significant challenges in working with these neuropeptide targets, including their long-lasting effects, which can complicate the immediate recording of electrophysiological or behavioral responses. Additionally, their complex interactions within neural circuits require sophisticated and precise methodologies to accurately delineate their specific roles and effects in the context of AUD.
The field of neuropharmacology is rapidly evolving, leveraging innovative animal models and cutting-edge research techniques. Rapid and high-throughput animal models have become crucial in swiftly identifying and validating new potential targets for AUD. These models expedite the translation of preclinical findings into clinical applications, bridging the gap between laboratory research and real-world treatment scenarios. Technological advancements in neuropharmacology are significantly enhancing the potential of emerging targets in the treatment of AUD. Innovations in drug delivery systems, ultrasound, small molecule analogues, nanoparticles, nanodroplets, and nanoemulsions, are enabling more effective therapeutics and localized penetration of the blood-brain barrier185–193. This is crucial for the greater efficacy of new pharmacological targets, ensuring that therapeutic agents effectively reach specific brain areas involved in AUD. Additionally, advancements in gene therapy194 and novel neuromodulation techniques195,196 are providing novel ways to directly influence neural systems associated with AUD. These technologies are making previously challenging targets more accessible and open new avenues for exploring and validating emerging targets in AUD treatment.
We have scrutinized a spectrum of emerging targets for AUD, each at various stages of development, from recent preclinical discoveries to those moving towards clinical trials. This is by no means an exhaustive list of emerging targets – others such as neuropeptide pituitary adenylate cyclase-activating polypeptide101–105, neurotensin197–204, neurokinins205, cholecystokinin206, and parvalbumin68,207,208 similarly deserve further attention. For example, ghrelin, a hormone predominantly secreted by the stomach, plays a critical role in regulating appetite, energy metabolism, and stimulating neurogenesis209. Antagonism of its receptor (growth hormone secretagogue receptor) is emerging therapeutic target for AUD and alcoholic liver disease, warranting clinical trials210–213. Likewise, orexins (also known as hypocretin), a neuronal and peptidergic population most well characterized for their role in energy homeostasis and wakefulness214, may also play a pivotal role in alcohol and substance misuse (recently reviewed; see215). Further, there are several well-studied receptor targets such as the metabotropic glutamate receptor 2 (mGlu2), which have shown promise in treating AUD213,216,217. While this review focuses largely on peptide targets, emerging targets ranging from natural products and psychedelic compounds to lipid transmitters have proven promising in reducing alcohol consumption218–222.
With the advent of transgenic mouse lines, genetically encoded optical biosensors, and optogenetics, alongside advancements in electrophysiology and in-vivo microscopy, we are on the cusp of being able to monitor the interactions of drugs and their targets in real time within behaving organisms62,223–226. This not only enables precise mapping of receptor activity but also offers a window into the complex signaling pathways and temporal patterns that govern alcohol use and dependence. Beyond receptor interactions, the focus is expanding to encompass the role of these targets as biomarkers, and alternative means for modulation, such as enzymes that synthesize or degrade these targets227, providing an innovative angle for therapeutic intervention. Furthermore, the integration of single-cell RNA sequencing into AUD research228,229 promises to dissect the heterogeneity of neuronal responses to alcohol, enhancing the specificity of treatments across the spectrum of comorbidities that accompany AUD. New targets will continue to emerge along with technological advancements that allow for greater specificity in treatment during various stages of the transition of casual substance misuse to dependence230,231 as well as for various individual comorbidities.
It’s prudent to consider these innovative targets in conjunction with advancements in drug delivery and neuromodulation, like new methods of delivering treatments to the brain193 and non-invasive techniques like focused ultrasound for neuronal modulation195,196. While targeted micro-infusion into specific brain regions is invaluable for isolating central effects and reducing peripheral side effects in studies, and a comprehensive examination of the systemic effects of these compounds is essential, as many of the targets discussed also significantly influence the gastrointestinal system and other peripheral organs232–236. Furthermore, the interplay between alcohol and various conditions such as eating disorders237, diabetes238, and cognitive decline239 demands more research. This need is especially urgent given the increasing number of elderly in the U.S.240. Genetic models of neurodegenerative diseases are helping to shed light on the interaction between alcohol and aging241–243. Notably, certain targets, like SST have a multifaceted role in both alcohol effects and aging241,244 while also exhibiting protection against alcohol induced pathophysiology in the periphery83–85 presenting a chance to address a variety of comorbid conditions. Additionally, in-vitro approaches like cerebral organoids245 could facilitate high-throughput screening for AUD treatment targets, and the emergence of wearable biosensors promises more information on AUD and its diverse effects246.
The literature suggests multiple favorable emerging targets for the treatment of AUD, some of which have FDA approved therapies already available, and all of which should be pursued with enthusiasm. The burgeoning array of potential targets and innovative technologies in our review heralds a new era of precision in treating AUD, promising to tailor therapies to individual needs and stages of addiction amidst an aging population with diverse comorbidities.
HIGHLIGHTS.
Despite significant research investments, new FDA-approved treatments for Alcohol Use Disorder (AUD) are scarce, and the lack of reliable biomarkers hampers progress.
Neuropeptides like somatostatin, vasoactive intestinal peptide, GLP-1, nociceptin, and neuropeptide S show promise as therapeutic targets and potential biomarkers for AUD.
Innovative animal models, advanced drug delivery systems, gene therapy, and new neuromodulation methods are crucial for developing new treatments for AUD.
FUNDING
R01AA029403–01A1, Penn State Department of Biology, and Penn State Huck Institutes of the Life Sciences Endowment Funds
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Enos G (2023). Inherent challenges in treating AUD temper hopes for major breakthrough. Alcoholism & Drug Abuse Weekly 35, 1–8. 10.1002/ADAW.33650. [DOI] [Google Scholar]
- 2.Flanagan JC, Jones JL, Jarnecke AM, and Back SE (2018). Behavioral Treatments for Alcohol Use Disorder and Post-Traumatic Stress Disorder. Alcohol Res 39, 181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Grant BF, Goldstein RB, Saha TD, Patricia Chou S, Jung J, Zhang H, Pickering RP, June Ruan W, Smith SM, Huang B, et al. (2015). Epidemiology of DSM-5 Alcohol Use Disorder: Results From the National Epidemiologic Survey on Alcohol and Related Conditions III. JAMA Psychiatry 72, 757–766. 10.1001/JAMAPSYCHIATRY.2015.0584. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.2020 National Survey on Drug Use and Health (NSDUH) Releases. https://www.samhsa.gov/data/release/2020-national-survey-drug-use-and-health-nsduh-releases.
- 5.Alcohol Use Disorder | National Institute on Alcohol Abuse and Alcoholism (NIAAA) https://www.niaaa.nih.gov/alcohol-health/overview-alcohol-consumption/alcohol-use-disorders. [Google Scholar]
- 6.Olaker VR, Kendall EK, Wang CX, Parran TV, Terebuh P, Kaelber DC, Xu R, and Davis PB (2023). Association of Recent SARS-CoV-2 Infection With New-Onset Alcohol Use Disorder, January 2020 Through January 2022. JAMA Netw Open 6, e2255496–e2255496. 10.1001/JAMANETWORKOPEN.2022.55496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Alcohol Use in the United States: Age Groups and Demographic Characteristics | National Institute on Alcohol Abuse and Alcoholism (NIAAA) https://www.niaaa.nih.gov/alcohols-effects-health/alcohol-topics/alcohol-facts-and-statistics/alcohol-use-united-states-age-groups-and-demographic-characteristics. [Google Scholar]
- 8.Hingson RW, Zha W, and White AM (2017). Drinking Beyond the Binge Threshold: Predictors, Consequences, and Changes in the U.S. Am J Prev Med 52, 717–727. 10.1016/j.amepre.2017.02.014. [DOI] [PubMed] [Google Scholar]
- 9.Heilig M, Sommer WH, and Spanagel R (2016). The Need for Treatment Responsive Translational Biomarkers in Alcoholism Research. Curr Top Behav Neurosci 28, 151–171. 10.1007/7854_2015_5006. [DOI] [PubMed] [Google Scholar]
- 10.Mason BJ, and Heyser CJ (2021). ALCOHOL USE DISORDER: THE ROLE OF MEDICATION IN RECOVERY. Alcohol Res 41. 10.35946/arcr.v41.1.07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Howard JT, Perrotte JK, Flores K, Leong C, Nocito JD, and Howard KJ (2022). Trends in Binge Drinking and Heavy Alcohol Consumption among Pregnant Women in the US, 2011 to 2020. JAMA Netw Open, E2224846. 10.1001/jamanetworkopen.2022.24846. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Towers EB, Williams IL, Qillawala EI, Rissman EF, and Lynch WJ (2022). Sex/Gender Differences in The Time-Course for the Development of Substance Use Disorder: A Focus on the Telescoping Effect. Pharmacol Rev 74. 10.1124/pharmrev.121.000361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Diehl A, Croissant B, Batra A, Mundle G, Nakovics H, and Mann K (2007). Alcoholism in women: Is it different in onset and outcome compared to men? Eur Arch Psychiatry Clin Neurosci 257, 344–351. 10.1007/s00406-007-0737-z. [DOI] [PubMed] [Google Scholar]
- 14.Piazza NJ, Vrbka JL, and Yeager RD (1989). Telescoping of alcoholism in women alcoholics. Subst Use Misuse 24, 19–28. 10.3109/10826088909047272. [DOI] [PubMed] [Google Scholar]
- 15.Randall CL, Roberts JS, Del Boca FK, Carroll KM, Connors GJ, and Mattson ME (1999). Telescoping of landmark events associated with drinking: a gender comparison. J Stud Alcohol 60, 252–260. 10.15288/jsa.1999.60.252. [DOI] [PubMed] [Google Scholar]
- 16.Ashley MJ, Olin JS, le Riche WH, Kornaczewski A, Schmidt W, and Rankin JG (1977). Morbidity in Alcoholics: Evidence for Accelerated Development of Physical Disease in Women. Arch Intern Med 137, 883–887. 10.1001/archinte.1977.03630190041012. [DOI] [PubMed] [Google Scholar]
- 17.Koob GF (2014). Neurocircuitry of alcohol addiction: synthesis from animal models. Handb Clin Neurol 125, 33–54. 10.1016/B978-0-444-62619-6.00003-3. [DOI] [PubMed] [Google Scholar]
- 18.Koob GF (2017). The dark side of addiction the horsley gantt to joseph brady connection. Journal of Nervous and Mental Disease 205, 270–272. 10.1097/NMD.0000000000000551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Koob GF (2013). Addiction is a reward deficit and stress surfeit disorder. Front Psychiatry 4. 10.3389/fpsyt.2013.00072. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.GF K, and ND V (2016). Neurobiology of addiction: a neurocircuitry analysis. Lancet Psychiatry 3, 760–773. 10.1016/S2215-0366(16)00104-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.George O, and Koob GF (2010). Individual differences in prefrontal cortex function and the transition from drug use to drug dependence. Preprint at Neurosci Biobehav Rev, https://doi.org/10.1016/j.neubiorev.2010.05.002 https://doi.org/10.1016/j.neubiorev.2010.05.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Wee S, and Koob GF (2010). The role of the dynorphin-κ opioid system in the reinforcing effects of drugs of abuse. Preprint at Springer, https://doi.org/10.1007/s00213-010-1825-8 https://doi.org/10.1007/s00213-010-1825-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Koob GF, and Volkow ND (2009). Neurocircuitry of Addiction. Neuropsychopharmacology 2010 35:1 35, 217–238. 10.1038/npp.2009.110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Kranzler HR, and Soyka M (2018). Diagnosis and Pharmacotherapy of Alcohol Use Disorder: A Review HHS Public Access. JAMA 320, 815–824. 10.1001/jama.2018.11406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Rawls E, Kummerfeld E, and Zilverstand A (2021). An integrated multimodal model of alcohol use disorder generated by data-driven causal discovery analysis. Commun Biol 4. 10.1038/s42003-021-01955-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Siomek-Gorecka A, Dlugosz A, and Czarnecki D (2021). The molecular basis of alcohol use disorder (Aud). genetics, epigenetics, and nutrition in aud: An amazing triangle. Preprint at MDPI AG, https://doi.org/10.3390/ijms22084262 https://doi.org/10.3390/ijms22084262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Burnette EM, Nieto SJ, Grodin EN, Lindsay, ·, Meredith R, Hurley B, Miotto K, Artha, ·, Gillis J, and Ray LA (2022). Novel Agents for the Pharmacological Treatment of Alcohol Use Disorder. 82, 251–274. 10.1007/s40265-021-01670-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Sachdeva A, Chandra M, Choudhary M, Dayal P, and Anand KS (2016). Alcohol-related dementia and neurocognitive impairment: A review study. Preprint at Kowsar Medical Institute, https://doi.org/10.5812/ijhrba.27976 https://doi.org/10.5812/ijhrba.27976. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Lowe PP, Morel C, Ambade A, Iracheta-Vellve A, Kwiatkowski E, Satishchandran A, Furi I, Cho Y, Gyongyosi B, Catalano D, et al. (2020). Chronic alcohol-induced neuroinflammation involves CCR2/5-dependent peripheral macrophage infiltration and microglia alterations. J Neuroinflammation 17. 10.1186/s12974-020-01972-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Bishehsari F, Magno E, Swanson G, Desai V, Voigt RM, Forsyth CB, and Keshavarzian A (2017). Alcohol and Gut-Derived Inflammation. Alcohol Res 38, 163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Hoek JB, Cahill A, and Pastorino JG (2002). Alcohol and Mitochondria: A Dysfunctional Relationship. Gastroenterology 122, 2049. 10.1053/GAST.2002.33613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Manzo-Avalos S, and Saavedra-Molina A (2010). Cellular and Mitochondrial Effects of Alcohol Consumption. Int J Environ Res Public Health 7, 4281. 10.3390/IJERPH7124281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.León BE, Kang S, Franca-Solomon G, Shang P, and Choi DS (2022). Alcohol-Induced Neuroinflammatory Response and Mitochondrial Dysfunction on Aging and Alzheimer’s Disease. Front Behav Neurosci 15, 778456. 10.3389/FNBEH.2021.778456/BIBTEX. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Siggins RW, McTernan PM, Simon L, Souza-Smith FM, and Molina PE (2023). Mitochondrial Dysfunction: At the Nexus between Alcohol-Associated Immunometabolic Dysregulation and Tissue Injury. Int J Mol Sci 24. 10.3390/IJMS24108650. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Osna NA, Donohue TM, and Kharbanda KK (2017). Alcoholic Liver Disease: Pathogenesis and Current Management. Alcohol Res 38, 147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Pleil KE, and Skelly MJ (2018). CRF modulation of central monoaminergic function: Implications for sex differences in alcohol drinking and anxiety. Alcohol 72, 33. 10.1016/J.ALCOHOL.2018.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Roberto M, Spierling SR, Kirson D, and Zorrilla EP (2017). Corticotropin Releasing Factor (CRF) and Addictive Behaviors. Int Rev Neurobiol 136, 5. 10.1016/BS.IRN.2017.06.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Snyder AE, and Silberman Y (2021). Corticotropin Releasing Factor and Norepinephrine Related Circuitry Changes in the Bed Nucleus of the Stria Terminalis in Stress and Alcohol and Substance Use Disorders. Neuropharmacology 201, 108814. 10.1016/J.NEUROPHARM.2021.108814. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Agoglia AE, Crofton EJ, and Herman MA (2020). Biological intersection of sex, age, and environment in the corticotropin releasing factor (CRF) system and alcohol. Neuropharmacology 170, 108045. 10.1016/J.NEUROPHARM.2020.108045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Schreiber AL, and Gilpin NW (2018). Corticotropin-Releasing Factor (CRF) Neurocircuitry and Neuropharmacology in Alcohol Drinking. Handb Exp Pharmacol 248, 435. 10.1007/164_2017_86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Simpson S, Shankar K, Kimbrough A, and George O (2020). Role of Corticotropin-Releasing Factor in Alcohol and Nicotine Addiction. Brain Res 1740, 146850. 10.1016/J.BRAINRES.2020.146850. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Sparrow AM, Lowery-Gionta EG, Pleil KE, Li C, Sprow GM, Cox BR, Rinker JA, Jijon AM, Peňl J, Navarro M, et al. (2012). Central Neuropeptide Y Modulates Binge-Like Ethanol Drinking in C57BL/6J Mice via Y1 and Y2 Receptors. Neuropsychopharmacology 37, 1409. 10.1038/NPP.2011.327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Gilpin NW, Misra K, Herman MA, Cruz MT, Koob GF, and Roberto M (2011). Neuropeptide Y Opposes Alcohol Effects on GABA Release in Amygdala and Blocks the Transition to Alcohol Dependence. Biol Psychiatry 69, 1091. 10.1016/J.BIOPSYCH.2011.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Vena AA, Zandy SL, Cofresí RU, and Gonzales RA (2020). Behavioral, Neurobiological, and Neurochemical Mechanisms of Ethanol Self-Administration: A Translational Review. Pharmacol Ther 212, 107573. 10.1016/J.PHARMTHERA.2020.107573. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Karkhanis AN, and Al-Hasani R (2020). Dynorphin and its role in alcohol use disorder. Brain Res 1735. 10.1016/J.BRAINRES.2020.146742. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Abrahao KP, Salinas AG, and Lovinger DM (2017). Alcohol and the Brain: Neuronal Molecular Targets, Synapses, and Circuits. Neuron 96, 1223–1238. 10.1016/J.NEURON.2017.10.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.van den Pol AN (2012). Neuropeptide Transmission in Brain Circuits. Preprint at NIH Public Access, 10.1016/j.neuron.2012.09.014 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Crowley NA, Dao NC, Magee SN, Bourcier AJ, and Lowery-Gionta EG (2019). Animal Models of Alcohol Use Disorder and the Brain: From Casual Drinking to Dependence. 10.1037/tps0000198. [DOI]
- 49.Liguz-Lecznar M, Dobrzanski G, and Kossut M (2022). Somatostatin and Somatostatin-Containing Interneurons—From Plasticity to Pathology. Biomolecules 12. 10.3390/BIOM12020312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Urban-Ciecko J, and Barth AL (2016). Somatostatin-expressing neurons in cortical networks. Nat Rev Neurosci 17, 401–409. 10.1038/nrn.2016.53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Yavorska I, and Wehr M (2016). Somatostatin-expressing inhibitory interneurons in cortical circuits. Front Neural Circuits 10, 76. 10.3389/FNCIR.2016.00076/BIBTEX. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Liguz-Lecznar M, Urban-Ciecko J, and Kossut M (2016). Somatostatin and somatostatin-containing neurons in shaping neuronal activity and plasticity. Preprint at Frontiers Media S.A., https://doi.org/10.3389/fncir.2016.00048 https://doi.org/10.3389/fncir.2016.00048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Brockway DF, and Crowley NA (2020). Turning the ′Tides on Neuropsychiatric Diseases: The Role of Peptides in the Prefrontal Cortex. Front Behav Neurosci 14. 10.3389/fnbeh.2020.588400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Hendry SHC, Jones EG, Emsons PC, and O’leary JL (1984). MORPHOLOGY, DISTRIBUTION, AND SYNAPTIC RELATIONS OF SOMATOSTATIN-AND NEUROPEPTIDE Y-IMMUNOREACTIVE NEURONS IN RAT AND MONKEY NEOCORTEX. [DOI] [PMC free article] [PubMed]
- 55.Melchitzky DS, and Lewis DA (2008). Dendritic-targeting GABA neurons in monkey prefrontal cortex: Comparison of somatostatin- and calretinin-immunoreactive axon terminals. Synapse 62, 456–465. 10.1002/SYN.20514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Martel G, Dutar P, Epelbaum J, and Viollet C (2012). Somatostatinergic systems: An update on brain functions in normal and pathological aging. Front Endocrinol (Lausanne) 3, 1–15. 10.3389/fendo.2012.00154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Pradayrol L, Jörnvall H, Mutt V, and Ribet A (1980). N-terminally extended somatostatin: The primary structure of somatostatin-28. FEBS Lett 109, 55–58. 10.1016/0014-5793(80)81310-X. [DOI] [PubMed] [Google Scholar]
- 58.Wang HL, Bogen C, Reisine T, and Dichter M (1989). Somatostatin-14 and somatostatin-28 induce opposite effects on potassium currents in rat neocortical neurons. Proc Natl Acad Sci U S A 86, 9616–9620. 10.1073/pnas.86.23.9616. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Hadjidakis DJ, Raptis SA, Souvatzoglou A, Karaiskos C, Diamantopoulos EJ, and Moulopoulos SD (1986). Differences between somatostatin-28 and somatostatin-14 with respect to their biological effects in healthy humans and acromegalics. Clin Physiol Biochem 4, 372–383. [PubMed] [Google Scholar]
- 60.Robinson SL, and Thiele TE (2020). A role for the neuropeptide somatostatin in the neurobiology of behaviors associated with substances abuse and affective disorders. Preprint at Elsevier Ltd, 10.1016/j.neuropharm.2020.107983 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Sicher AR, Starnes WD, Griffith KR, Dao NC, Smith GC, Brockway DF, and Crowley NA (2023). Adolescent binge drinking leads to long-lasting changes in cortical microcircuits in mice. Neuropharmacology 234, 109561. 10.1016/J.NEUROPHARM.2023.109561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Dao NC, Brockway DF, and Crowley NA (2019). In Vitro Optogenetic Characterization of Neuropeptide Release from Prefrontal Cortical Somatostatin Neurons. Neuroscience 419, 1–4. 10.1016/j.neuroscience.2019.08.014. [DOI] [PubMed] [Google Scholar]
- 63.Brockway DF, Griffith KR, Aloimonos CM, Gordon JA, Kupferschmidt DA, and Crowley Correspondence NA (2023). Somatostatin peptide signaling dampens cortical circuits and promotes exploratory behavior. 10.1016/j.celrep.2023.112976. [DOI] [PMC free article] [PubMed]
- 64.Dao NC, Brockway DF, Suresh Nair M, Sicher AR, and Crowley NA (2021). Somatostatin neurons control an alcohol binge drinking prelimbic microcircuit in mice. Neuropsychopharmacology 2021, 1–12. 10.1038/s41386-021-01050-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Dao NC, Suresh Nair M, Magee SN, Moyer JB, Sendao V, Brockway DF, and Crowley NA (2020). Forced Abstinence From Alcohol Induces Sex-Specific Depression-Like Behavioral and Neural Adaptations in Somatostatin Neurons in Cortical and Amygdalar Regions. Front Behav Neurosci 14. 10.3389/fnbeh.2020.00086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Suresh Nair M, Dao NC, Lopez Melean D, Griffith KR, Starnes WD, Moyer JB, Sicher AR, Brockway DF, Meeks KD, and Crowley NA (2022). Somatostatin neurons in the bed nucleus of the stria terminalis play a sex-dependent role in binge Drinking. Brain Res Bull 186, 38–46. 10.1016/J.BRAINRESBULL.2022.05.010. [DOI] [PubMed] [Google Scholar]
- 67.Crofton EJ, O’Buckley TK, Bohnsack JP, Morrow AL, and Herman MA (2023). Divergent Population-Specific Effects of Chronic Ethanol Exposure on Excitability and Inhibitory Transmission in Male and Female Rat Central Amygdala. J Neurosci 43, 7056–7068. 10.1523/JNEUROSCI.0717-23.2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Joffe ME, Winder DG, and Conn PJ (2020). Contrasting sex-dependent adaptations to synaptic physiology and membrane properties of prefrontal cortex interneuron subtypes in a mouse model of binge drinking. 10.1016/j.neuropharm.2020.108126. [DOI] [PMC free article] [PubMed]
- 69.Li M, Cabrera-Garcia D, Salling MC, Au E, Yang G, and Harrison NL (2021). Alcohol reduces the activity of somatostatin interneurons in the mouse prefrontal cortex: A neural basis for its disinhibitory effect? Neuropharmacology 188, 108501. 10.1016/J.NEUROPHARM.2021.108501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Robinson SL, and Thiele TE (2023). Somatostatin signaling modulates binge drinking behavior via the central nucleus of the amygdala. Neuropharmacology 237, 109622. 10.1016/J.NEUROPHARM.2023.109622. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Sicher AR, Duerr A, Starnes WD, and Crowley NA (2022). Adolescent Alcohol and Stress Exposure Rewires Key Cortical Neurocircuitry. Preprint at Frontiers Media S.A., 10.3389/fnins.2022.896880 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Wilson DA, Fleming G, Williams CRO, Teixeira CM, Smiley JF, and Saito M (2023). Somatostatin neuron contributions to cortical slow wave dysfunction in adult mice exposed to developmental ethanol. Front Neurosci 17. 10.3389/FNINS.2023.1127711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Andrade JP, Fernando PM, Madeira MD, Paula-Barbosa MM, Cadete-Leite A, and Zimmer J (1992). Effects of chronic alcohol consumption and withdrawal on the somatostatin-immunoreactive neurons of the rat hippocampal dentate hilus. Hippocampus 2, 65–71. 10.1002/HIPO.450020109. [DOI] [PubMed] [Google Scholar]
- 74.Joffe ME, Winder DG, and Conn PJ (2021). Increased Synaptic Strength and mGlu2/3 Receptor Plasticity on Mouse Prefrontal Cortex Intra-telencephalic Pyramidal Cells Following Intermittent Access to Ethanol. Alcohol Clin Exp Res 45, 518. 10.1111/ACER.14546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Crowley NA, and Joffe ME (2021). Developing breakthrough psychiatric treatments by modulating G protein-coupled receptors on prefrontal cortex somatostatin interneurons. Neuropsychopharmacology. 10.1038/S41386-021-01119-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Long JB, Rigamonti DD, Dosaka K, Kraimer JM, and Martinez-Arizala A (1992). Somatostatin causes vasoconstriction, reduces blood flow and increases vascular permeability in the rat central nervous system. Journal of Pharmacology and Experimental Therapeutics 260. [PubMed] [Google Scholar]
- 77.Silwal A, House A, Sandoval K, Vijeth S, Umbaugh D, Crider A, Mobayen S, Neumann W, and Witt KA (2022). Novel Somatostatin Receptor-4 Agonist SM-I-26 Mitigates Lipopolysaccharide-Induced Inflammatory Gene Expression in Microglia. Neurochem Res 47, 768–780. 10.1007/S11064-021-03482-Z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Ochi R, Ueno F, Sakuma M, Tani H, Tsugawa S, Graff-Guerrero A, Uchida H, Mimura M, Oshima S, Matsushita S, et al. (2022). Patterns of functional connectivity alterations induced by alcohol reflect somatostatin interneuron expression in the human cerebral cortex. Scientific Reports 2022 12:1 12, 1–9. 10.1038/s41598-022-12035-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Valeri J, O’Donovan SM, Wang W, Sinclair D, Bollavarapu R, Gisabella B, Platt D, Stockmeier C, and Pantazopoulos H (2022). Altered expression of somatostatin signaling molecules and clock genes in the hippocampus of subjects with substance use disorder. Front Neurosci 16. 10.3389/FNINS.2022.903941. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Bornschein J, Drozdov I, and Malfertheiner P (2009). Octreotide LAR: safety and tolerability issues. Expert Opin Drug Saf 8, 755–768. 10.1517/14740330903379525. [DOI] [PubMed] [Google Scholar]
- 81.Sandostatin LAR Depot Important Safety Information. https://www.hcp.novartis.com/products/sandostatin-lar-depot/carcinoid-syndrome/safety-profile/?site=SDS-1232987GS100005&source=01030&utm_source=google&utm_medium=cpc&utm_campaign=all%20new%20branded%20campaigns&utm_content=sds-1232987&utm_term=safety%20profile&site_property=Google&omap_code=SDS-1232987&gclid=CjwKCAiAnL-sBhBnEiwAJRGigh0yTq78a6zCpUHSoYEZUPzXJnfaM4z5Cc_-HffwPRz69b8JBBzc-BoCHN8QAvD_BwE&gclsrc=aw.ds.
- 82.SANDOSTATIN® LAR® Safety Information, Warnings & Precautions. https://www.sandostatin.com/en/neuroendocrine-tumors/sandostatin-and-net/sandostatin-safety/.
- 83.Chaudhuri KR, Thomaides T, Watson L, and Mathias CJ (1995). Octreotide reduces alcohol-induced hypotension and orthostatic symptoms in primary autonomic failure. QJM: An International Journal of Medicine 88, 719–725. 10.1093/OXFORDJOURNALS.QJMED.A068996. [DOI] [PubMed] [Google Scholar]
- 84.Jiang R, and Wei H (2021). Beneficial effects of octreotide in alcohol-induced neuropathic pain. Role of H2S, BDNF, TNF-α and Nrf2. Acta Cir Bras 36, 360408. 10.1590/ACB360408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Xu WF, Wang Y, Huang H, Wu JW, Che Y, Ding CJ, Zhang Q, Cao WL, and Cao LJ (2022). Octreotide-based therapies effectively protect mice from acute and chronic gastritis. Eur J Pharmacol 928. 10.1016/J.EJPHAR.2022.174976. [DOI] [PubMed] [Google Scholar]
- 86.Tsukada T, Horovitch SJ, Montminy MR, Mandel G, and Goodman RH (1985). Structure of the human vasoactive intestinal polypeptide gene. DNA 4, 293–300. 10.1089/DNA.1985.4.293. [DOI] [PubMed] [Google Scholar]
- 87.Hu E, Demmou L, Cauli B, Gallopin T, Ne Geoffroy H, Harris-Warrick RM, Le Paupardin-Tritsch D, Lambolez B, Vincent P, and Hepp RR (2011). VIP, CRF, and PACAP Act at Distinct Receptors to Elicit Different cAMP/PKA Dynamics in the Neocortex. Cerebral Cortex March 21, 708–718. 10.1093/cercor/bhq143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Ciranna L, Topolnik L, Caleo M, Cunha-Reis D, and Caulino-Rocha A (2020). VIP Modulation of Hippocampal Synaptic Plasticity: A Role for VIP Receptors as Therapeutic Targets in Cognitive Decline and Mesial Temporal Lobe Epilepsy. 10.3389/fncel.2020.00153. [DOI] [PMC free article] [PubMed]
- 89.Cherubini E, Topolnik L, Feldmeyer D, Junior Apicella A, and Marchionni I (2022). Citation: Apicella Aj and Marchionni I (2022) VIP-Expressing GABAergic Neurons: Disinhibitory vs. Inhibitory Motif and Its Role in Communication Across Neocortical Areas. Front. Cell. Neurosci 16, 811484. 10.3389/fncel.2022.811484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Couvineau A, and Laburthe M (2012). VPAC receptors: Structure, molecular pharmacology and interaction with accessory proteins. Br J Pharmacol 166, 42–50. 10.1111/J.1476-5381.2011.01676.X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Delgado M, and Ganea D (2011). Vasoactive intestinal peptide: a neuropeptide with pleiotropic immune functions. Amino Acids 2011 45:1 45, 25–39. 10.1007/S00726-011-1184-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Ganea D, Hooper KM, and Kong W (2015). THE NEUROPEPTIDE VIP: DIRECT EFFECTS ON IMMUNE CELLS AND INVOLVEMENT IN INFLAMMATORY AND AUTOIMMUNE DISEASES. Acta Physiol (Oxf) 213, 442. 10.1111/APHA.12427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Eger M, Liron T, Hiram-Bab S, Awida Z, Giladi E, Dangoor D, Fridkin M, Kohavi D, Gozes I, and Gabet Y (2021). Therapeutic Potential of Vasoactive Intestinal Peptide and its Derivative Stearyl-Norleucine-VIP in Inflammation-Induced Osteolysis. Front Pharmacol 12, 638128. 10.3389/FPHAR.2021.638128/BIBTEX. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Martínez C, Juarranz Y, Gutiérrez-Cañas I, Carrión M, Pérez-García S, Villanueva-Romero R, Castro D, Lamana A, Mellado M, González-álvaro I, et al. (2020). A Clinical Approach for the Use of VIP Axis in Inflammatory and Autoimmune Diseases. Int J Mol Sci 21. 10.3390/IJMS21010065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Iwasaki M, Akiba Y, and Kaunitz JD (2019). Recent advances in vasoactive intestinal peptide physiology and pathophysiology: focus on the gastrointestinal system. F1000Res 8. 10.12688/F1000RESEARCH.18039.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Szecowka J, Sandberg E, and Efendic S (1980). The Interaction of Vasoactive Intestinal Polypeptide (VIP), Glucose and Arginine on the Secretion of Insulin, Glucagon and Somatostatin in the Perfused Rat Pancreas. Diabetologia 19, 137–142. [DOI] [PubMed] [Google Scholar]
- 97.Achilly NP (2016). Properties of VIP+ synapses in the suprachiasmatic nucleus highlight their role in circadian rhythm. J Neurophysiol 115, 2701. 10.1152/JN.00393.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Emson PC, Fahrenkrug J, and Spokes EGS (1979). Vasoactive intestinal polypeptide (VIP): distribution in normal human brain and in Huntington’s disease. Brain Res 173, 174–178. 10.1016/0006-8993(79)91109-0. [DOI] [PubMed] [Google Scholar]
- 99.Sun QQ, Prince DA, and Huguenard JR (2003). Vasoactive intestinal polypeptide and pituitary adenylate cyclase-activating polypeptide activate hyperpolarization-activated cationic current and depolarize thalamocortical neurons in vitro. J Neurosci 23, 2751–2758. 10.1523/JNEUROSCI.23-07-02751.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Lu J, Piper SJ, Zhao P, Miller LJ, Wootten D, and Sexton PM (2022). Targeting VIP and PACAP Receptor Signaling: New Insights into Designing Drugs for the PACAP Subfamily of Receptors. Preprint at MDPI, https://doi.org/10.3390/ijms23158069 https://doi.org/10.3390/ijms23158069. [DOI] [PMC free article] [PubMed]
- 101.Ferragud A, Velazquez-Sanchez C, Minnig MA, Sabino V, and Cottone P (2021). Pituitary adenylate cyclase-activating polypeptide (PACAP) modulates dependence-induced alcohol drinking and anxiety-like behavior in male rats. Neuropsychopharmacology 46, 509–518. 10.1038/s41386-020-00904-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Minnig MA, Blasio A, Ferragud A, Sami YN, Erhard EE, Clark RH, DiLeo A, Giuliano C, Everitt BJ, Cottone P, et al. (2022). Pituitary adenylate cyclase-activating polypeptide type 1 receptor within the nucleus accumbens core mediates excessive alcohol drinking in alcohol-preferring rats. Neuropharmacology 212. 10.1016/j.neuropharm.2022.109063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Gargiulo AT, Curtis GR, and Barson JR (2020). Pleiotropic pituitary adenylate cyclase-activating polypeptide (PACAP): Novel insights into the role of PACAP in eating and drug intake. Preprint at Elsevier B.V., 10.1016/j.brainres.2019.146626 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Gargiulo AT, Pirino BE, Curtis GR, and Barson JR (2021). Effects of pituitary adenylate cyclase-activating polypeptide isoforms in nucleus accumbens subregions on ethanol drinking. Addiction Biology 26. 10.1111/adb.12972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Gupta A, Gargiulo AT, Curtis GR, Badve PS, Pandey S, and Barson JR (2018). Pituitary adenylate cyclase-activating polypeptide-27 (PACAP-27) in the thalamic paraventricular nucleus is stimulated by ethanol drinking. Alcohol Clin Exp Res 42, 1650. 10.1111/ACER.13826. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Sanford D, Luong L, Vu JP, Oh S, Gabalski A, Lewis M, Pisegna JR, and Germano P (2022). The VIP/VPAC1R Pathway Regulates Energy and Glucose Homeostasis by Modulating GLP-1, Glucagon, Leptin and PYY Levels in Mice. Biology (Basel) 11. 10.3390/biology11030431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Hensch TK, Prinz AA, Cauli B, Letzkus JJ, Tao HW, Mesik L, Ma W-P, Li L-Y, Ibrahim LA, Huang ZJ, et al. (2015). Functional response properties of VIP-expressing inhibitory neurons in mouse visual and auditory cortex. 10.3389/fncir.2015.00022. [DOI] [PMC free article] [PubMed]
- 108.Lee SH, Hjerling-Leffler J, Zagha E, Fishell G, and Rudy B (2010). The Largest Group of Superficial Neocortical GABAergic Interneurons Expresses Ionotropic Serotonin Receptors. Journal of Neuroscience 30, 16796–16808. 10.1523/JNEUROSCI.1869-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Georgiou C, Kehayas V, Lee KS, Brandalise F, Sahlender DA, Blanc J, Knott G, and Holtmaat A (2022). A subpopulation of cortical VIP-expressing interneurons with highly dynamic spines. Commun Biol 5. 10.1038/S42003-022-03278-Z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Engel M, Smidt MP, van Hooft JA, Homberg J, and Feldmeyer D (2013). The serotonin 5-HT 3 receptor: a novel neurodevelopmental target. 10.3389/fncel.2013.00076. [DOI] [PMC free article] [PubMed]
- 111.Magistretti PJ (1990). VIP neurons in the cerebral cortex. Trends Pharmacol Sci 11, 250–254. 10.1016/0165-6147(90)90253-5. [DOI] [PubMed] [Google Scholar]
- 112.Nigro MJ, Staiger JF, Topolnik L, Brandon MP, Badrinarayanan S, Manseau F, and Williams S (2021). A Characterization of the Electrophysiological and Morphological Properties of Vasoactive Intestinal Peptide (VIP) Interneurons in the Medial Entorhinal Cortex (MEC). 10.3389/fncir.2021.653116. [DOI] [PMC free article] [PubMed]
- 113.Williams LE, and Holtmaat A (2019). Higher-Order Thalamocortical Inputs Gate Synaptic Long-Term Potentiation via Disinhibition. Neuron 101, 91–102.e4. 10.1016/J.NEURON.2018.10.049. [DOI] [PubMed] [Google Scholar]
- 114.Staub JL, Sarles H, Chayvialle JA, Descos F, Lassmann V, and Vague P (1981). Relationship between intravenous ethanol, alcohol-induced inhibition of pancreatic secretion and plasma concentration of immunoreactive pancreatic polypeptide, vasoactive intestinal peptide, and somatostatin in man. Regul Pept 2, 61–68. 10.1016/0167-0115(81)90066-5. [DOI] [PubMed] [Google Scholar]
- 115.Poplawski MM, Boyadjieva N, and Sarkar DK (2005). Vasoactive Intestinal Peptide and Corticotropin-Releasing Hormone Increase β-Endorphin Release and Proopiomelanocortin Messenger RNA Levels in Primary Cultures of Hypothalamic Cells: Effects of Acute and Chronic Ethanol Treatment. Alcohol Clin Exp Res 29, 648–655. 10.1097/01.ALC.0000158834.11252.2E. [DOI] [PubMed] [Google Scholar]
- 116.Kovanen L, Saarikoski ST, Haukka J, Pirkola S, Aromaa A, Lönnqvist J, and Partonen T (2010). GENETICS Circadian Clock Gene Polymorphisms in Alcohol Use Disorders and Alcohol Consumption. 10.1093/alcalc/agq035. [DOI] [PubMed]
- 117.Madeira MD, Andrade JP, Lieberman AR, Sousa N, Almeida OFX, and Paula-Barbosa MM (1997). Chronic Alcohol Consumption and Withdrawal Do Not Induce Cell Death in the Suprachiasmatic Nucleus, But Lead to Irreversible Depression of Peptide Immunoreactivity and mRNA Levels. [DOI] [PMC free article] [PubMed]
- 118.Farnell YZ, Allen GC, Neuendorff N, West JR, Chen Wei-Jung A, and Earnest DJ (2009). Effects of neonatal alcohol exposure on vasoactive intestinal polypeptide neurons in the rat suprachiasmatic nucleus. Alcohol 43, 387–396. 10.1016/j.alcohol.2009.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Szabó G, Mácsai M, Schek É, and Telegdy G (1998). The Effect of Vasoactive Intestinal Polypeptide and Pituitary Adenylate Cyclase Activating Polypeptide on Tolerance to Morphine and Alcohol in Micea. Ann N Y Acad Sci 865, 566–569. 10.1111/J.1749-6632.1998.TB11238.X. [DOI] [PubMed] [Google Scholar]
- 120.Thompson SM, Fabian CB, Ferranti AS, and Joffe ME (2023). Acute alcohol and chronic drinking bidirectionally regulate the excitability of prefrontal cortex vasoactive intestinal peptide interneurons. Neuropharmacology 238, 109638. 10.1016/J.NEUROPHARM.2023.109638. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Lee MR, and Jayant RD (2019). Penetration of the Blood-Brain-Barrier by Peripheral Neuropeptides: New Approaches to Enhancing Transport and Endogenous Expression. Cell Tissue Res 375, 287. 10.1007/S00441-018-2959-Y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Gao X, Wu B, Zhang Q, Chen J, Zhu J, Zhang W, Rong Z, Chen H, and Jiang X (2007). Brain delivery of vasoactive intestinal peptide enhanced with the nanoparticles conjugated with wheat germ agglutinin following intranasal administration. J Control Release 121, 156–167. 10.1016/J.JCONREL.2007.05.026. [DOI] [PubMed] [Google Scholar]
- 123.Engleman EA, Rodd ZA, Bell RL, and Murphy JM (2008). The Role of 5-HT3 Receptors in Drug Abuse and as a Target for Pharmacotherapy. CNS Neurol Disord Drug Targets 7, 454. 10.2174/187152708786927886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Kruse Klausen M, Thomsen, | Morgane Gitta Wortwein, |, and Fink-Jensen A (2021). The role of glucagon-like peptide 1 (GLP-1) in addictive disorders. Mental Health Services. 10.1111/bph.v179.4/issuetoc. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Skibicka KP (2013). The central GLP-1: implications for food and drug reward. Front Neurosci 7. 10.3389/FNINS.2013.00181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Gupta V (2013). Glucagon-like peptide-1 analogues: An overview. Indian J Endocrinol Metab 17, 413. 10.4103/2230-8210.111625. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Müller TD, Finan B, Bloom SR, D’Alessio D, Drucker DJ, Flatt PR, Fritsche A, Gribble F, Grill HJ, Habener JF, et al. (2019). Glucagon-like peptide 1 (GLP-1). Mol Metab 30, 72. 10.1016/J.MOLMET.2019.09.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Maniscalco JW, Kreisler AD, and Rinaman L (2012). Satiation and stress-induced hypophagia: Examining the role of hindbrain neurons expressing prolactin-releasing peptide or glucagon-like peptide 1. Front Neurosci 6, 39238. 10.3389/FNINS.2012.00199/BIBTEX. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Holt MK, and Trapp S (2016). The physiological role of the brain GLP-1 system in stress. Cogent Biol 2, 1229086. 10.1080/23312025.2016.1229086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Jessen L, Smith EP, Ulrich-Lai Y, Herman JP, Seeley RJ, Sandoval D, and D’Alessio D (2017). Central nervous system GLP-1 receptors regulate islet hormone secretion and glucose homeostasis in male rats. Endocrinology 158, 2124–2133. 10.1210/en.2016-1826. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Cabou C, and Burcelin R (2011). GLP-1, the Gut-Brain, and Brain-Periphery Axes. Rev Diabet Stud 8, 418. 10.1900/RDS.2011.8.418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Shirazi RH, Dickson SL, and Skibicka KP (2013). Gut Peptide GLP-1 and Its Analogue, Exendin-4, Decrease Alcohol Intake and Reward. PLoS One 8, 61965. 10.1371/journal.pone.0061965. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Egecioglu E, Steensland P, Fredriksson I, Feltmann K, Engel JA, and Jerlhag E (2013). The glucagon-like peptide 1 analogue Exendin-4 attenuates alcohol mediated behaviors in rodents. Psychoneuroendocrinology 38, 1259–1270. 10.1016/J.PSYNEUEN.2012.11.009. [DOI] [PubMed] [Google Scholar]
- 134.Sirohi S, Schurdak JD, Seeley RJ, Benoit SC, and Davis JF (2016). Central & peripheral glucagon-like peptide-1 receptor signaling differentially regulate addictive behaviors. 10.1016/j.physbeh.2016.04.013. [DOI] [PubMed]
- 135.Farokhnia M, Fede SJ, Grodin EN, Browning BD, Crozier ME, Schwandt ML, Hodgkinson CA, Momenan R, and Leggio L (2022). Differential association between the GLP1R gene variants and brain functional connectivity according to the severity of alcohol use. Scientific Reports 2022 12:1 12, 1–10. 10.1038/s41598-022-17190-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Farokhnia M, Browning BD, Crozier ME, Sun H, Akhlaghi F, and Leggio L (2022). The glucagon-like peptide-1 system is modulated by acute and chronic alcohol exposure: Findings from human laboratory experiments and a post-mortem brain study. Addiction Biology 27, e13211. 10.1111/ADB.13211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Dong M, Wen S, and Zhou L (2022). The Relationship Between the Blood-Brain-Barrier and the Central Effects of Glucagon-Like Peptide-1 Receptor Agonists and Sodium-Glucose Cotransporter-2 Inhibitors. Diabetes, Metabolic Syndrome and Obesity 15, 2583–2597. 10.2147/DMSO.S375559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Wang JY, Wang QW, Yang XY, Yang W, Li DR, Jin JY, Zhang HC, and Zhang XF (2023). GLP−1 receptor agonists for the treatment of obesity: Role as a promising approach. Preprint at Frontiers Media S.A, 10.3389/fendo.2023.1085799 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Nauck MA, Quast DR, Wefers J, and Meier JJ (2021). GLP-1 receptor agonists in the treatment of type 2 diabetes – state-of-the-art. Mol Metab 46, 101102. 10.1016/J.MOLMET.2020.101102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Corder G, Castro DC, Bruchas MR, and Scherrer G (2018). Endogenous and Exogenous Opioids in Pain. Annu Rev Neurosci 41, 453–473. 10.1146/ANNUREV-NEURO-080317-061522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Wtorek K, and Janecka A (2021). Potential of Nociceptin/Orphanin FQ Peptide Analogs for Drug Development. Chem Biodivers 18. 10.1002/CBDV.202000871. [DOI] [PubMed] [Google Scholar]
- 142.D’Oliveira da Silva F, Robert C, Lardant E, Pizzano C, Bruchas MR, Guiard BP, Chauveau F, and Moulédous L (2023). Targeting Nociceptin/Orphanin FQ receptor to rescue cognitive symptoms in a mouse neuroendocrine model of chronic stress. Mol Psychiatry. 10.1038/S41380-023-02363-X. [DOI] [PubMed] [Google Scholar]
- 143.Armstead WM (2011). Nociceptin/Orphanin Phenylalanine Glutamine (FQ) Receptor and Cardiovascular Disease. Cardiovasc Ther 29, 23–28. 10.1111/J.1755-5922.2010.00241.X. [DOI] [PubMed] [Google Scholar]
- 144.El Daibani A, and Che T (2022). Spotlight on Nociceptin/Orphanin FQ Receptor in the Treatment of Pain. Molecules 27. 10.3390/MOLECULES27030595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Ubaldi M, Cannella N, Borruto AM, Petrella M, Micioni Di Bonaventura MV, Soverchia L, Stopponi S, Weiss F, Cifani C, and Ciccocioppo R (2021). Role of Nociceptin/Orphanin FQ-NOP Receptor System in the Regulation of Stress-Related Disorders. Int J Mol Sci 22. 10.3390/IJMS222312956. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Zaveri NT (2011). The Nociceptin/Orphanin FQ Receptor (NOP) as a Target for Drug Abuse Medications. Curr Top Med Chem 11, 1151. 10.2174/156802611795371341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Koob GF (2021). Drug addiction: Hyperkatifeia/negative reinforcement as a framework for medications development. Pharmacol Rev 73, 163–201. 10.1124/PHARMREV.120.000083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Parker KE, Pedersen CE, Gomez AM, Spangler SM, Walicki MC, Feng SY, Stewart SL, Otis JM, Al-Hasani R, McCall JG, et al. (2019). A Paranigral VTA Nociceptin Circuit that Constrains Motivation for Reward. Cell 178, 653–671.e19. 10.1016/J.CELL.2019.06.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Ciccocioppo R, Panocka I, Rino, ·, Quitadamo FE, and Massi M (1999). Ethanol induces conditioned place preference in genetically selected alcohol-preferring rats. [DOI] [PubMed]
- 150.Murphy NP, Lee Y, and Maidment NT (1999). Orphanin FQrnociceptin blocks acquisition of morphine place preference. [DOI] [PubMed]
- 151.Ciccocioppo R, Angeletti S, Panocka I, and Massi M (2000). Nociceptin/orphanin FQ and drugs of abuse. [DOI] [PubMed]
- 152.Panocka RCI, Domenico CP, and Massi RM (1999). RAPID COMMUNICATION Effect of nociceptin on alcohol intake in alcohol-preferring rats. [DOI] [PubMed]
- 153.Ciccocioppo R, Polidori C, Antonelli L, Salvadori S, Guerrini R, and Massi M (2002). Pharmacological characterization of the nociceptin receptor which mediates reduction of alcohol drinking in rats. Peptides (N.Y.) 23, 117–125. 10.1016/S0196-9781(01)00587-3. [DOI] [PubMed] [Google Scholar]
- 154.Ciccocioppo R, Angeletti S, Sanna PP, Weiss F, and Massi M (2000). Effect of nociceptinrorphanin FQ on the rewarding properties of morphine. [DOI] [PubMed]
- 155.Panocka RCI, Domenico CP, and Massi RM (1999). Effect of nociceptin on alcohol intake in alcohol-preferring rats. [DOI] [PubMed]
- 156.Ciccocioppo R, Stopponi S, Economidou D, Kuriyama M, Kinoshita H, Heilig M, Roberto M, Weiss F, and Teshima K (2014). Chronic Treatment with Novel Brain-Penetrating Selective NOP Receptor Agonist MT-7716 Reduces Alcohol Drinking and Seeking in the Rat. Neuropsychopharmacology 2014 39:11 39, 2601–2610. 10.1038/npp.2014.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Economidou D, Cippitelli A, Stopponi S, Braconi S, Clementi S, Ubaldi M, Martin-Fardon R, Weiss F, Massi M, and Ciccocioppo R (2011). Activation of Brain NOP Receptors Attenuates Acute and Protracted Alcohol Withdrawal Symptoms in the Rat. Alcohol Clin Exp Res 35, 747–755. 10.1111/j.1530-0277.2010.01392.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Li W, Ren Z, Tang Y, Fu Y, Sun S, Ding R, Hou J, Mai Y, Zhan B, Zhu Y, et al. (2023). Rostromedial tegmental nucleus nociceptin/orphanin FQ (N/OFQ) signaling regulates anxiety- and depression-like behaviors in alcohol withdrawn rats. Neuropsychopharmacology 48, 908–919. 10.1038/S41386-022-01482-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Economidou D, Hansson AC, Weiss F, Terasmaa A, Sommer WH, Cippitelli A, Fedeli A, Martin-Fardon R, Massi M, Ciccocioppo R, et al. (2008). Dysregulation of Nociceptin/Orphanin FQ Activity in the Amygdala Is Linked to Excessive Alcohol Drinking in the Rat. Biol Psychiatry 64, 211–218. 10.1016/j.biopsych.2008.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Rorick-Kehn LM, Ciccocioppo R, Wong CJ, Witkin JM, Martinez-Grau MA, Stopponi S, Adams BL, Katner JS, Perry KW, Toledo MA, et al. (2016). A novel, orally-bioavailable nociceptin receptor antagonist, LY2940094, reduces ethanol self-administration and ethanol-seeking in animal models. Alcohol Clin Exp Res 40, 945. 10.1111/ACER.13052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Borruto AM, Fotio Y, Stopponi S, Brunori G, Petrella M, Caputi FF, Romualdi P, Candeletti S, Narendran R, Rorick-Kehn LM, et al. (2020). NOP receptor antagonism reduces alcohol drinking in male and female rats through mechanisms involving the central amygdala and ventral tegmental area. Br J Pharmacol 177, 1525–1537. 10.1111/bph.14915. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Borruto AM, Fotio Y, Stopponi S, Petrella M, De Carlo S, Domi A, Ubaldi M, Weiss F, and Ciccocioppo R (2021). NOP receptor antagonism attenuates reinstatement of alcohol-seeking through modulation of the mesolimbic circuitry in male and female alcohol-preferring rats. Neuropsychopharmacology 46, 2121–2131. 10.1038/S41386-021-01096-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Post A, Smart TS, Jackson K, Mann J, Mohs R, Rorick-Kehn L, Statnick M, Anton R, O’Malley SS, and Wong CJ (2016). Proof-of-Concept Study to Assess the Nociceptin Receptor Antagonist LY2940094 as a New Treatment for Alcohol Dependence. Alcohol Clin Exp Res 40, 1935–1944. 10.1111/ACER.13147. [DOI] [PubMed] [Google Scholar]
- 164.Flynn SM, Epperly PM, Davenport AT, Cami-Kobeci G, Husbands SM, Ko MC, and Czoty PW (2019). Effects of stimulation of mu opioid and nociceptin/orphanin FQ peptide (NOP) receptors on alcohol drinking in rhesus monkeys. Neuropsychopharmacology 44, 1476. 10.1038/S41386-019-0390-Z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Mariani JJ, Dobbins RL, Heath A, Gray F, and Hassman H (2024). Open-label investigation of rapid initiation of extended-release buprenorphine in patients using fentanyl and fentanyl analogs. Am J Addict 33. 10.1111/AJAD.13484. [DOI] [PubMed] [Google Scholar]
- 166.Borris JB, Dowd-Green C, Bowman LA, Nesbit SA, Fingerhood M, and Stewart RW (2024). Evaluation of opioid use disorder treatment outcomes in patients receiving split daily versus once daily dosing of buprenorphine-naloxone. Journal of substance use and addiction treatment 156, 209209. 10.1016/J.JOSAT.2023.209209. [DOI] [PubMed] [Google Scholar]
- 167.Ehrich E, Turncliff R, Du Y, Leigh-Pemberton R, Fernandez E, Jones R, and Fava M (2015). Evaluation of opioid modulation in major depressive disorder. Neuropsychopharmacology 40, 1448–1455. 10.1038/NPP.2014.330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Witkin JM, Wallace TL, and Martin WJ (2019). Therapeutic Approaches for NOP Receptor Antagonists in Neurobehavioral Disorders: Clinical Studies in Major Depressive Disorder and Alcohol Use Disorder with BTRX-246040 (LY2940094). Handb Exp Pharmacol 254, 399–415. 10.1007/164_2018_186. [DOI] [PubMed] [Google Scholar]
- 169.Xu Y-L, Reinscheid RK, Huitron-Resendiz S, Clark SD, Wang Z, Lin SH, Brucher FA, Zeng J, nociceptin, or, Reinscheid, orphanin F., et al. (2004). Neuropeptide S: A Neuropeptide Promoting Arousal and Anxiolytic-like Effects tides such as hypocretin/orexin (Hcrt/Ox) (Sutcliffe and de Lecea. Neuron 43, ga-lanin. [DOI] [PubMed] [Google Scholar]
- 170.Okamura N, Garau C, Duangdao DM, Clark SD, Jü Ngling K, Pape H-C, and Reinscheid RK (2011). Neuropeptide S Enhances Memory During the Consolidation Phase and Interacts with Noradrenergic Systems in the Brain. Neuropsychopharmacology 36, 744–752. 10.1038/npp.2010.207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Rizzi A, Vergura R, Marzola G, Ruzza C, Guerrini R, Salvadori S, Regoli D, and Calo G (2008). Neuropeptide S is a stimulatory anxiolytic agent: A behavioural study in mice. Br J Pharmacol 154, 471–479. 10.1038/BJP.2008.96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Cannella N, Borruto AM, Petrella M, Micioni MV, Bonaventura D, Soverchia L, Cifani C, De Carlo S, Domi E, and Ubaldi M (2022). A Role for Neuropeptide S in Alcohol and Cocaine Seeking. 10.3390/ph15070800. [DOI] [PMC free article] [PubMed]
- 173.Tobinski A-M, Rappeneau V, Ruzza C, and Reinscheid RK (2021). pharmaceuticals Role of the Neuropeptide S System in Emotionality, Stress Responsiveness and Addiction-Like Behaviours in Rodents: Relevance to Stress-Related Disorders. 10.3390/ph14080780. [DOI] [PMC free article] [PubMed]
- 174.Botticelli L, Micioni E, Bonaventura D, Ubaldi M, Ciccocioppo R, Cifani C, Micioni MV, Reinscheid K, and Ruzza C (2021). pharmaceuticals The Neural Network of Neuropeptide S (NPS): Implications in Food Intake and Gastrointestinal Functions. Pharmaceuticals 14, 293. 10.3390/ph14040293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Reinscheid RK, and Ruzza C (2021). pharmaceuticals Pharmacology, Physiology and Genetics of the Neuropeptide S System. 10.3390/ph14050401. [DOI] [PMC free article] [PubMed]
- 176.Li Y-Q, Gundlach AL, Olucha-Bordonau FE, Adori C, Hökfelt T, Barde S, Bogdanovic N, Uhlén M, Reinscheid RR, and Kovacs GG (2015). Neuropeptide S- and Neuropeptide S receptor-expressing neuron populations in the human pons. Frontiers in Neuroanatomy | www.frontiersin.org 9, 126. 10.3389/fnana.2015.00126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Laas K, Reif A, Akkermann K, Kiive E, Domschke K, Lesch KP, Veidebaum T, and Harro J (2015). Neuropeptide S receptor gene variant and environment: contribution to alcohol use disorders and alcohol consumption. Addiction Biology 20, 605–616. 10.1111/ADB.12149. [DOI] [PubMed] [Google Scholar]
- 178.Tobinski AM, and Rappeneau V (2021). Role of the neuropeptide S system in emotionality, stress responsiveness and addiction-like behaviours in rodents: Relevance to stress-related disorders. Preprint at MDPI AG, https://doi.org/10.3390/ph14080780 https://doi.org/10.3390/ph14080780. [DOI] [PMC free article] [PubMed]
- 179.Ghazal P, Corsi M, Roth A, Faggioni F, Corti C, Merlo Pick E, Pucciarelli S, Ciccocioppo R, and Ubaldi M (2014). Paradoxical response to the sedative effects of diazepam and alcohol in C57BL/6J mice lacking the neuropeptide S receptor. Peptides (N.Y.) 61, 107–113. 10.1016/J.PEPTIDES.2014.09.010. [DOI] [PubMed] [Google Scholar]
- 180.Ruggeri B, Braconi S, Cannella N, Kallupi M, Soverchia L, Ciccocioppo R, and Ubaldi M (2010). Neuropeptide S receptor gene expression in alcohol withdrawal and protracted abstinence in postdependent rats. Alcohol Clin Exp Res 34, 90–97. 10.1111/J.1530-0277.2009.01070.X. [DOI] [PubMed] [Google Scholar]
- 181.Enquist J, Ferwerda M, Madhavan A, Hok D, and Whistler JL (2012). Chronic Ethanol Potentiates the Effect of Neuropeptide S in the Basolateral Amygdala and Shows Increased Anxiolytic and Anti-Depressive Effects. Neuropsychopharmacology 37, 2436–2445. 10.1038/npp.2012.102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Badia-Elder NE, Henderson AN, Bertholomey ML, Dodge NC, and Stewart RB (2008). The Effects of Neuropeptide S on Ethanol Drinking and Other Related Behaviors in Alcohol-Preferring and -Nonpreferring Rats. Alcohol Clin Exp Res 32, 1380–1387. 10.1111/J.1530-0277.2008.00713.X. [DOI] [PubMed] [Google Scholar]
- 183.Cannella N, Kallupi M, Li HW, Stopponi S, Cifani C, Ciccocioppo R, and Ubaldi M (2016). Neuropeptide S differently modulates alcohol-related behaviors in alcohol-preferring and non-preferring rats. Psychopharmacology (Berl) 233, 2915–2924. 10.1007/S00213-016-4333-7/FIGURES/5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Clark SD, Kenakin TP, Gertz S, Hassler C, Gay EA, Langston TL, Reinscheid RK, and Runyon SP (2017). Identification of the first biased NPS receptor agonist that retains anxiolytic and memory promoting effects with reduced levels of locomotor stimulation. Neuropharmacology 118, 69–78. 10.1016/J.NEUROPHARM.2017.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Terstappen GC, Meyer AH, Bell RD, and Zhang W (2021). Strategies for delivering therapeutics across the blood–brain barrier. Preprint at Nature Research, 10.1038/s41573-021-00139-y . [DOI] [PubMed] [Google Scholar]
- 186.Haumann R, Videira JC, Kaspers GJL, van Vuurden DG, and Hulleman E (2020). Overview of Current Drug Delivery Methods Across the Blood–Brain Barrier for the Treatment of Primary Brain Tumors. Preprint at Adis, 10.1007/s40263-020-00766-w . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Achar A, Myers R, and Ghosh C (2021). Drug delivery challenges in brain disorders across the blood–brain barrier: Novel methods and future considerations for improved therapy. Preprint at MDPI, 10.3390/biomedicines9121834 . [DOI] [PMC free article] [PubMed]
- 188.Dong X (2018). Current Strategies for Brain Drug Delivery. Theranostics 8, 1481–1493. 10.7150/thno.21254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Teleanu RI, Preda MD, Niculescu AG, Vladâcenco O, Radu CI, Grumezescu AM, and Teleanu DM (2022). Current Strategies to Enhance Delivery of Drugs across the Blood–Brain Barrier. Pharmaceutics 14. 10.3390/pharmaceutics14050987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Zhang S, Zhang S, Luo S, Tang P, Wan M, Wu D, and Gao W (2022). Ultrasound-assisted brain delivery of nanomedicines for brain tumor therapy: advance and prospect Journal of Nanobiotechnology. J Nanobiotechnology 20, 287. 10.1186/s12951-022-01464-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Bérard C, Desgranges S, Dumas N, Novell A, Larrat B, Hamimed M, Taulier N, Estève MA, Correard F, and Contino-Pépin C (2022). Perfluorocarbon Nanodroplets as Potential Nanocarriers for Brain Delivery Assisted by Focused Ultrasound-Mediated Blood–Brain Barrier Disruption. Pharmaceutics 14. 10.3390/PHARMACEUTICS14071498/S1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Zhou Y, and Han Y (2022). Engineered bacteria as drug delivery vehicles: Principles and prospects. Engineering Microbiology 2, 100034. 10.1016/J.ENGMIC.2022.100034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Crowley NA, and Medina SH (2022). Targeted and transient opening of the blood brain barrier in discrete neurocircuits and brain regions. Neuropsychopharmacology. 10.1038/S41386-022-01380-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Ford MM, George BE, Van Laar VS, Holleran KM, Naidoo J, Hadaczek P, Vanderhooft LE, Peck EG, Dawes MH, Ohno K, et al. (2023). GDNF gene therapy for alcohol use disorder in male non-human primates. Nature Medicine | 29. 10.1038/s41591-023-02463-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Chen H, Zhu N, Osman M, Biskowitz R, Liu J, Khandare S, Butler P, Wong PK, and Kothapalli SR (2021). A transparent low intensity pulsed ultrasound (LIPUS) chip for high-throughput cell stimulation. Lab Chip 21, 4734–4742. 10.1039/D1LC00667C. [DOI] [PubMed] [Google Scholar]
- 196.Murphy KR, Farrell JS, Gomez JL, Stedman QG, Li N, Leung SA, Good CH, Qiu Z, Firouzi K, Pauly KB, et al. (2022). A tool for monitoring cell type–specific focused ultrasound neuromodulation and control of chronic epilepsy. Proc Natl Acad Sci U S A 119. 10.1073/PNAS.2206828119/-/DCSUPPLEMENTAL. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Keller BN, Hajnal A, Browning KN, Arnold AC, and Silberman Y (2022). Involvement of the Dorsal Vagal Complex in Alcohol-Related Behaviors. Preprint at Frontiers Media S.A., 10.3389/fnbeh.2022.801825 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Grupp LA, Harding S, Grupp A, Harding •S, and Grupp LA (1996). Neurotensin attenuates the reduction in alcohol drinking produced by angiotensin II Key words Angiotensin II-Neurotensin • Alcohol drinking-Renin-angiotensin system. [DOI] [PubMed]
- 199.Gereau GB, Torruella-Suárez ML, Sizer SE, Zhou D, Wykoff LA, Boyt KM, Alvarez-Pamir A, and McElligott ZA (2023). GABA Release From Central Amygdala Neurotensin Neurons Differentially Modulates Reward and Consummatory Behavior in Male and Female Mice. bioRxiv. 10.1101/2023.09.14.557768. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Hartmann MC, and Pleil KE (2021). Circuit and neuropeptide mechanisms of the paraventricular thalamus across stages of alcohol and drug use. Neuropharmacology 198. 10.1016/J.NEUROPHARM.2021.108748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Gereau GB, Garrison SD, and McElligott ZA (2023). Neurotensin and energy balance. J Neurochem 166, 189–200. 10.1111/JNC.15868. [DOI] [PubMed] [Google Scholar]
- 202.Torruella-Suárez ML, and McElligott ZA (2020). Neurotensin in reward processes. Neuropharmacology 167. 10.1016/J.NEUROPHARM.2020.108005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Rodríguez FD, Sánchez ML, and Coveñas R (2023). Neurotensin and Alcohol Use Disorders: Towards a Pharmacological Treatment. Int J Mol Sci 24. 10.3390/IJMS24108656. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Pandey S, Badve PS, Curtis GR, Leibowitz SF, and Barson JR (2019). Neurotensin in the posterior thalamic paraventricular nucleus: inhibitor of pharmacologically relevant ethanol drinking. Addiction biology 24, 3–16. 10.1111/ADB.12546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Schank JR (2020). Neurokinin Receptors in Drug and Alcohol Addiction. Brain Res 1734, 146729. 10.1016/J.BRAINRES.2020.146729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Ballaz S, Espinosa N, and Bourin M (2021). Does endogenous cholecystokinin modulate alcohol intake? Neuropharmacology 193, 108539. 10.1016/J.NEUROPHARM.2021.108539. [DOI] [PubMed] [Google Scholar]
- 207.Patton MS, Heckman M, Kim C, Mu C, and Mathur BN (2021). Compulsive alcohol consumption is regulated by dorsal striatum fast-spiking interneurons. Neuropsychopharmacology 46, 351–359. 10.1038/S41386-020-0766-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Ferranti AS, Johnson KA, Winder DG, Conn PJ, and Joffe ME (2022). Prefrontal cortex parvalbumin interneurons exhibit decreased excitability and potentiated synaptic strength after ethanol reward learning. Alcohol 101, 17–26. 10.1016/J.ALCOHOL.2022.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Abdalla MMI (2015). Ghrelin – Physiological Functions and Regulation. Eur Endocrinol 11, 90. 10.17925/EE.2015.11.02.90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Richardson RS, Sulima A, Rice KC, Kucharczk JA, Janda KD, Nisbett KE, Koob GF, Vendruscolo LF, and Leggio L (2023). Pharmacological GHSR (ghrelin receptor) blockade reduces alcohol binge-like drinking in male and female mice. Neuropharmacology 238. 10.1016/J.NEUROPHARM.2023.109643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Kharbanda KK, Farokhnia M, Deschaine SL, Bhargava R, Rodriguez-Flores M, Casey CA, Goldstone AP, Jerlhag E, Leggio L, and Rasineni K (2022). Role of the ghrelin system in alcohol use disorder and alcohol-associated liver disease: A narrative review. Alcohol Clin Exp Res 46, 2149–2159. 10.1111/ACER.14967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Jerlhag E (2023). Animal studies reveal that the ghrelin pathway regulates alcohol-mediated responses. Front Psychiatry 14. 10.3389/FPSYT.2023.1050973. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Farokhnia M, Faulkner ML, Piacentino D, Lee MR, and Leggio L (2019). Ghrelin: From a gut hormone to a potential therapeutic target for alcohol use disorder. Physiol Behav 204, 49–57. 10.1016/J.PHYSBEH.2019.02.008. [DOI] [PubMed] [Google Scholar]
- 214.Chieffi S, Carotenuto M, Monda V, Valenzano A, Villano I, Precenzano F, Tafuri D, Salerno M, Filippi N, Nuccio F, et al. (2017). Orexin system: The key for a healthy life. Front Neurol 8, 266332. 10.3389/FPHYS.2017.00357/BIBTEX. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Moorman DE (2018). The hypocretin/orexin system as a target for excessive motivation in alcohol use disorders. Psychopharmacology (Berl) 235, 1663. 10.1007/S00213-018-4871-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Johnson KA, Liput DJ, Homanics GE, and Lovinger DM (2020). Age-dependent impairment of metabotropic glutamate receptor 2-dependent long-term depression in the mouse striatum by chronic ethanol exposure. Alcohol 82, 11–21. 10.1016/J.ALCOHOL.2019.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Johnson KA, and Lovinger DM (2020). Allosteric modulation of metabotropic glutamate receptors in alcohol use disorder: Insights from preclinical investigations. Adv Pharmacol 88, 193–232. 10.1016/BS.APHA.2020.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.Jensen ME, Stenbæk DS, Juul TS, Fisher PMD, Ekstrøm CT, Knudsen GM, and Fink-Jensen A (2022). Protocol: Psilocybin-assisted therapy for reducing alcohol intake in patients with alcohol use disorder: protocol for a randomised, double-blinded, placebo-controlled 12-week clinical trial (The QUANTUM Trip Trial). BMJ Open 12. 10.1136/BMJOPEN-2022-066019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Wang F, Li Y, Zhang YJ, Zhou Y, Li S, and Li H Bin (2016). Natural products for the prevention and treatment of hangover and alcohol use disorder. Preprint at MDPI AG, 10.3390/molecules21010064 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Penetar DM, Toto LH, Lee DYW, and Lukas SE (2015). A single dose of kudzu extract reduces alcohol consumption in a binge drinking paradigm. Drug Alcohol Depend 153, 194–200. 10.1016/j.drugalcdep.2015.05.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Crowley NA, Magee SN, Feng M, Jefferson SJ, Morris CJ, Dao NC, Brockway DF, and Luscher B (2019). Ketamine normalizes binge drinking-induced defects in glutamatergic synaptic transmission and ethanol drinking behavior in female but not male mice. Neuropharmacology 149, 35–44. 10.1016/j.neuropharm.2019.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Tomczyk M, Zovko-Končić M, and Chrostek L (2012). Phytotherapy of Alcoholism. 10.1177/1934578X1200700243 7, 273–280. [DOI] [PubMed] [Google Scholar]
- 223.Al-Hasani R, Wong JMT, Mabrouk OS, McCall JG, Schmitz GP, Porter-Stransky KA, Aragona BJ, Kennedy RT, and Bruchas MR (2018). In vivo detection of optically-evoked opioid peptide release. Elife 7. 10.7554/eLife.36520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.Inutsuka A, Ino D, and Onaka T (2021). Detection of neuropeptides in vivo and open questions for current and upcoming fluorescent sensors for neuropeptides. Peptides (N.Y.) 136, 170456. 10.1016/J.PEPTIDES.2020.170456. [DOI] [PubMed] [Google Scholar]
- 225.Crowley NA, Bloodgood DW, Hardaway JA, Kendra AM, McCall JG, Al-Hasani R, McCall NM, Yu W, Schools ZL, Krashes MJ, et al. (2016). Dynorphin Controls the Gain of an Amygdalar Anxiety Circuit. Cell Rep 14, 2774–2783. 10.1016/j.celrep.2016.02.069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Wang H, Qian T, Zhao Y, Zhuo Y, Wu C, Osakada T, Chen P, Chen Z, Ren H, Yan Y, et al. (2023). A tool kit of highly selective and sensitive genetically encoded neuropeptide sensors. Science (1979) 382. 10.1126/SCIENCE.ABQ8173/SUPPL_FILE/SCIENCE.ABQ8173_DATA_S1_AND_S2.ZIP. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Hartman K, Mielczarek P, Smoluch M, and Silberring J (2020). Inhibitors of neuropeptide peptidases engaged in pain and drug dependence. Neuropharmacology 175, 108137. 10.1016/J.NEUROPHARM.2020.108137. [DOI] [PubMed] [Google Scholar]
- 228.Brenner E, Tiwari GR, Kapoor M, Liu Y, Brock A, and Mayfield RD (2020). Single cell transcriptome profiling of the human alcohol-dependent brain. Hum Mol Genet 29, 1144. 10.1093/HMG/DDAA038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229.Van Den Oord EJCG, Lin,;, Xie Y, Zhao M, Aberg KA, and Clark SL (2022). A single-nucleus transcriptomics study of alcohol use disorder in the nucleus accumbens. medRxiv, 2022.06.16.22272431. 10.1101/2022.06.16.22272431. [DOI] [PubMed] [Google Scholar]
- 230.Koob GF, and Le Moal M (2008). Neurobiological mechanisms for opponent motivational processes in addiction. In Philosophical Transactions of the Royal Society B: Biological Sciences (Royal Society), pp. 3113–3123. 10.1098/rstb.2008.0094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231.Koob GF, and Le Moal M (2008). Addiction and the brain antireward system. Annu Rev Psychol 59, 29–53. 10.1146/ANNUREV.PSYCH.59.103006.093548. [DOI] [PubMed] [Google Scholar]
- 232.Shamsi BH, Chatoo M, Xu XK, Xu X, and Chen XQ (2021). Versatile Functions of Somatostatin and Somatostatin Receptors in the Gastrointestinal System. Front Endocrinol (Lausanne) 12, 652363. 10.3389/FENDO.2021.652363/BIBTEX. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233.Wu X, Conlin VS, Morampudi V, Ryz NR, Nasser Y, Bhinder G, Bergstrom KS, Yu HB, Waterhouse CCM, Buchan AMJ, et al. (2015). Vasoactive Intestinal Polypeptide Promotes Intestinal Barrier Homeostasis and Protection Against Colitis in Mice. PLoS One 10, e0125225. 10.1371/JOURNAL.PONE.0125225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234.Iwasaki M, Akiba Y, and Kaunitz JD (2019). Recent advances in vasoactive intestinal peptide physiology and pathophysiology: focus on the gastrointestinal system. F1000Res 8. 10.12688/F1000RESEARCH.18039.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235.Tulassay Z (1998). Somatostatin and the gastrointestinal tract. Scand J Gastroenterol Suppl 228, 115–121. 10.1080/003655298750026642. [DOI] [PubMed] [Google Scholar]
- 236.Yan M, Man S, Sun B, Ma L, Guo L, Huang L, and Gao W (2023). Gut liver brain axis in diseases: the implications for therapeutic interventions. Signal Transduct Target Ther 8. 10.1038/S41392-023-01673-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237.Curtis GR, Coudriet JM, Sanzalone L, Mack NR, Stein LM, Hayes MR, and Barson JR (2019). Short- and long-access palatable food self-administration results in different phenotypes of binge-type eating. Physiol Behav 212. 10.1016/J.PHYSBEH.2019.112700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238.Walter KN, Wagner JA, Cengiz E, Tamborlane WV, and Petry NM (2016). The need for research addressing alcohol use disorder and diabetes. Addiction (Abingdon, England) 111, 763–765. 10.1111/ADD.13187. [DOI] [PubMed] [Google Scholar]
- 239.Han L, and Jia J (2021). Long-term effects of alcohol consumption on cognitive function in seniors: a cohort study in China. BMC Geriatr 21, 1–10. 10.1186/S12877-021-02606-Y/FIGURES/3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.2020 Census: 1 in 6 People in the United States Were 65 and Over. https://www.census.gov/library/stories/2023/05/2020-census-united-states-older-population-grew.html.
- 241.Fuhrmann G, Strosser MT, Besnard F, Kempf E, Kempf J, and Ebel A (1986). Genotypic differences in age and chronic alcohol exposure effects on somatostatin levels in hippocampus and striatum in mice. Neurochem Res 11, 625–636. 10.1007/BF00965332/METRICS. [DOI] [PubMed] [Google Scholar]
- 242.Seemiller LR, Flores-Cuadra J, Griffith KR, Smith GC, and Crowley NA (2024). Alcohol and stress exposure across the lifespan are key risk factors for Alzheimer’s Disease and cognitive decline. Neurobiol Stress 29, 100605. 10.1016/J.YNSTR.2024.100605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243.Smith GC, Griffith KR, Sicher AR, Brockway DF, Proctor EA, Crowley NA, and Lab M (2024). MODERATE ALCOHOL CONSUMPTION INDUCES LASTING IMPACTS ON PREFRONTAL CORTICAL SIGNALING IN MICE. bioRxiv, 2024.04.03.587955. 10.1101/2024.04.03.587955. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244.Rofo F, Yilmaz CU, Metzendorf N, Gustavsson T, Beretta C, Erlandsson A, Sehlin D, Syvänen S, Nilsson P, and Hultqvist G (2021). Enhanced neprilysin-mediated degradation of hippocampal Aβ42 with a somatostatin peptide that enters the brain. Theranostics 11, 789–804. 10.7150/THNO.50263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245.Arzua T, Yan Y, Jiang C, Logan S, Allison RL, Wells C, Kumar SN, Schäfer R, and Bai X (2020). Modeling alcohol-induced neurotoxicity using human induced pluripotent stem cell-derived three-dimensional cerebral organoids. Translational Psychiatry 2020 10:1 10, 1–21. 10.1038/s41398-020-01029-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246.Christie G, Gual A, Nicola M. Di, Davis-Martin RE, Alessi SM, and Boudreaux ED (2021). Alcohol Use Disorder in the Age of Technology: A Review of Wearable Biosensors in Alcohol Use Disorder Treatment. Wearable Biosensors in Alcohol Use Disorder Treatment. Front. Psychiatry 12, 642813. 10.3389/fpsyt.2021.642813. [DOI] [PMC free article] [PubMed] [Google Scholar]
