1
|
Vozella V, Borgonetti V, Cruz B, Onge CMS, Bullard R, Vlkolinsky R, Ceballos DG, Ozburn AR, Roberts AJ, Ciccocioppo R, Bajo M, Roberto M. Apremilast reduces co-occurring alcohol drinking and mechanical allodynia and regulates central amygdala GABAergic transmission. JCI Insight 2025; 10:e189732. [PMID: 40260918 PMCID: PMC12016922 DOI: 10.1172/jci.insight.189732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 03/07/2025] [Indexed: 04/24/2025] Open
Abstract
The FDA-approved phosphodiesterase type 4 (PDE4) inhibitor, apremilast, has been recently investigated as a pharmacotherapy for alcohol use disorder (AUD) with promising efficacy in rodent models and humans. However, apremilast's effects on mechanical allodynia associated with AUD as well as distinct responses of this drug between males and females are understudied. The present study examined the behavioral and electrophysiological effects of apremilast in Marchigian Sardinian alcohol-preferring (msP) rats and their Wistar counterparts. We used a 2-bottle choice (2-BC) alcohol drinking procedure and tested mechanical sensitivity across our drinking regimen. Spontaneous inhibitory GABA-mediated postsynaptic currents from the central nucleus of the amygdala (CeA) following apremilast application were tested in a subset of rats using ex vivo electrophysiology. Transcript levels for Pde4a or -4b subtypes were assessed for their modulation by alcohol. Apremilast reduced alcohol drinking in both strains of rats. Apremilast reduced mechanical allodynia immediately after drinking, persisting into early and late abstinence. Apremilast increased GABAergic transmission in CeA slices of alcohol-exposed Wistars but not msP rats, suggesting neuroadaptations in msPs by excessive drinking and mechanical allodynia. Pde4 subtype transcript levels were increased in CeA by alcohol. These results suggest that apremilast alleviates co-occurring excessive drinking and pain sensitivity, and they further confirm PDE4's role in pain-associated AUD.
Collapse
Affiliation(s)
- Valentina Vozella
- Department of Translational Medicine, Scripps Research Institute, La Jolla, California, USA
| | - Vittoria Borgonetti
- Department of Translational Medicine, Scripps Research Institute, La Jolla, California, USA
| | - Bryan Cruz
- Department of Translational Medicine, Scripps Research Institute, La Jolla, California, USA
| | - Celsey M. St. Onge
- Department of Translational Medicine, Scripps Research Institute, La Jolla, California, USA
| | - Ryan Bullard
- Department of Translational Medicine, Scripps Research Institute, La Jolla, California, USA
| | - Roman Vlkolinsky
- Department of Translational Medicine, Scripps Research Institute, La Jolla, California, USA
| | - Diego Gomez Ceballos
- Department of Translational Medicine, Scripps Research Institute, La Jolla, California, USA
| | - Angela R. Ozburn
- Department of Behavioral Neuroscience at Oregon Health & Science University and VA Portland Health Care System, Portland, Oregon, USA
| | - Amanda J. Roberts
- Animal Models Core Facility, Scripps Research, La Jolla, California, USA
| | - Roberto Ciccocioppo
- Pharmacology Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | - Michal Bajo
- Department of Translational Medicine, Scripps Research Institute, La Jolla, California, USA
| | - Marisa Roberto
- Department of Translational Medicine, Scripps Research Institute, La Jolla, California, USA
| |
Collapse
|
2
|
Mateu-Mollá J, Pérez-Gálvez B, Villanueva-Blasco VJ. Pharmacological treatment for substance use disorder: A systematic review. Addict Behav 2025; 163:108242. [PMID: 39793188 DOI: 10.1016/j.addbeh.2024.108242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 11/23/2024] [Accepted: 12/31/2024] [Indexed: 01/13/2025]
Abstract
INTRODUCTION There is strong evidence of the substance dependence has a negative impact on key dimensions of health. The scientific evidence suggests that pharmacological treatment could play a fundamental role in its clinical management. OBJECTIVE The aim of this systematic review is to explore the existing pharmacological options for the treatment of substance use disorders. METHODS Following PRISMA recommendations, a review of recent scientific literature was conducted using three sources of information: Pubmed /Medline, Web of Science, and Scopus. Only randomized controlled trials published in the last five years, in both English and Spanish, and subjected to peer review were selected. The quality of the methodology used was defined through the application of the PEDro scale for quantitative studies. It was registered in the PROSPERO database. RESULTS After filtering through the respective advanced search engines, a total of 49 articles suitable for qualitative analysis were retrieved. Literature related to pharmacotherapy for alcohol use, as well as for cocaine, tobacco, cannabis, and opioid dependence, was identified among them. CONCLUSIONS There is a wide variety of drugs that can be used for the clinical management of dependence on these substances. Reflections are offered on the evidence of their effectiveness and the methodological rigor of the literature.
Collapse
Affiliation(s)
- Joaquín Mateu-Mollá
- Faculty of Health Sciences. Valencian International University. Valencia, Spain.
| | - Bartolomé Pérez-Gálvez
- General Directorate of Mental Health and Addictions, Generalitat Valenciana, Valencia, Spain.
| | | |
Collapse
|
3
|
Guo S, Zhao Y, Yuan Y, Liao Y, Jiang X, Wang L, Lu W, Shi J. Progress in the development of macrophage migration inhibitory factor small-molecule inhibitors. Eur J Med Chem 2025; 286:117280. [PMID: 39854942 DOI: 10.1016/j.ejmech.2025.117280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/08/2025] [Accepted: 01/11/2025] [Indexed: 01/27/2025]
Abstract
Macrophage migration inhibitory factor (MIF) functions as a critical cytokine regulating inflammatory and immune responses. Extensive research has demonstrated its involvement in the progression of various cancers, autoimmune diseases, and inflammatory disorders, establishing it as a pivotal target for anti-inflammatory and anticancer interventions. Therapeutic strategies aimed at MIF primarily focus on suppressing its activity through small molecule inhibitors and natural compounds. This review synthesizes current knowledge on MIF, encompassing its structural characteristics, enzymatic functions, signaling pathways, and roles in disease pathogenesis. Additionally, it provides an in-depth analysis of recent advancements in MIF inhibitor development, including design methodologies, structure-activity relationships, advanced eutectic analysis techniques, and key experimental findings. The discussion aims to support the development of safer, more effective, and highly selective small molecule inhibitors targeting MIF.
Collapse
Affiliation(s)
- Shujin Guo
- Department of Health Management Center, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yingying Zhao
- Department of Geriatric Medicine, School of Medicine and Life Science, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yan Yuan
- College of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 611756, China
| | - Yang Liao
- Department of Geriatric Medicine, School of Medicine and Life Science, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xuepan Jiang
- Department of Geriatric Medicine, School of Medicine and Life Science, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lin Wang
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu, 610041, China.
| | - Wei Lu
- Department of Dermatology and Venereology, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| | - Jianyou Shi
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China; The State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
4
|
Castro EM, Lotfipour S, Leslie FM. Neuroglia in substance use disorders. HANDBOOK OF CLINICAL NEUROLOGY 2025; 210:347-369. [PMID: 40148055 DOI: 10.1016/b978-0-443-19102-2.00014-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Substance use disorders (SUD) remain a major public health concern in which individuals are unable to control their use of substances despite significant harm and negative consequences. Drugs of abuse dysregulate major brain and behavioral functions. Glial cells, primarily microglia and astrocytes, play a crucial role in these drug-induced molecular and behavioral changes. This review explores preclinical and clinical studies of how neuroglia and their associated neuroinflammatory responses contribute to SUD and reward-related properties. We evaluate preclinical and clinical evidence for targeting neuroglia as therapeutic interventions. In addition, we evaluate the literature on the gut microbiome and its role in SUD. Clinical treatments are most effective for reducing drug cravings, and some have yielded promising results in other measures of drug use. N-Acetylcysteine, through modulation of cysteine-glutamate antiporter of glial cells, shows encouraging results across a variety of drug classes. Neuroglia and gut microbiome interactions are important factors to consider with regard to SUD and could lead to novel therapeutic avenues. Age- and sex-dependent properties of neuroglia, gut microbiome, and drug use behaviors are important areas in need of further investigation.
Collapse
Affiliation(s)
- Emily M Castro
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, United States
| | - Shahrdad Lotfipour
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, United States; Department of Emergency Medicine, School of Medicine, University of California, Irvine, Irvine, CA, United States; Department of Pathology and Laboratory Medicine, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Frances M Leslie
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, United States.
| |
Collapse
|
5
|
Zhang Y, Yu Z, Ye N, Zhen X. Macrophage migration inhibitory factor (MIF) in CNS diseases: Functional regulation and potential therapeutic indication. FUNDAMENTAL RESEARCH 2024; 4:1375-1388. [PMID: 39734533 PMCID: PMC11670708 DOI: 10.1016/j.fmre.2023.05.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/10/2023] [Accepted: 05/08/2023] [Indexed: 12/31/2024] Open
Abstract
Macrophage migration inhibitory factor (MIF) is a multifunctional protein that possesses cytokine, enzyme, and endocrine activities and acts as a chaperone-like molecule. Owing to its immune-inflammatory regulatory properties, the role of MIF has long been an attractive target in research on various autoimmune and inflammatory disorders. MIF is also widely expressed in the central nervous system (CNS), and its potential roles in CNS disorders have become a focus to elucidate the physiological and pathological effects of MIF and to explore its potential significance in the treatment of CNS diseases. Previously, the majority of work on MIF functional regulation was focused on MIF tautomerase inhibitors. However, mounting information has indicated that the functions of MIF extend far beyond its tautomerase activity. Here, we review the recent advances in understanding the complex roles of MIF in the pathogenesis of CNS disorders as well as the discovery and design of small molecules targeted to tautomerase and nuclease of MIF.
Collapse
Affiliation(s)
- Yu Zhang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
- Department of Pharmacy, Hubei Provincial Clinical Research Center for Parkinson's Disease, Xiangyang Key Laboratory of Movement Disorders, Xiangyang No.1 People′ Hospital, Hubei University of Medicine, Xiangyang 441000, China
| | - Zhexiang Yu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Na Ye
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Xuechu Zhen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin 2 D02, Ireland
| |
Collapse
|
6
|
Köhne S, Hillemacher T, Glahn A, Bach P. Emerging drugs in phase II and III clinical development for the treatment of alcohol use disorder. Expert Opin Emerg Drugs 2024; 29:219-232. [PMID: 38606899 DOI: 10.1080/14728214.2024.2342951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 04/10/2024] [Indexed: 04/13/2024]
Abstract
INTRODUCTION Alcohol Use Disorder (AUD) poses an ongoing significant global health burden. AUD is highly prevalent and affects not only the individuals with AUD, but also their communities and society at large. Even though pharmacotherapy is an integral part of AUD treatment, the few available substances show limited efficacy and limited clinical impact. Thus, there is a need for new innovative pharmacotherapeutic approaches. AREAS COVERED This paper provides a comprehensive review of drugs approved for the treatment of AUD as well as those currently in phase II and III development. Data from recent clinical trials has been reviewed and supplemented by additional literature based on a systematic search of the PubMed database and clinical trials registries. Compounds discussed include disulfiram, naltrexone, nalmefene, acamprosat, baclofen, sodium oxybate, doxazosin, varenicline, zonisamide, gabapentin, apremilast, ibudilast, ivermectin, tolcapone, mifepristone, suvorexant, ketamine, psilocybin, semaglutide, oxytocin and cannabidiol. EXPERT OPINION Even though the majority of the discussed compounds lack sufficient evidence to support their efficacy, multiple promising new treatment options are currently under investigation. Future research has to consider specific phenotypes and subgroups of AUD as well as a possible enhancement of the effects of psychotherapy through combination with pharmacotherapy. Practitioners should be encouraged to use available compounds to support existing therapeutic regimens.
Collapse
Affiliation(s)
- Sophie Köhne
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Medical School Hannover, Hannover, Germany
| | - Thomas Hillemacher
- Department of Psychiatry and Psychotherapy, Paracelsus Medical University Nuremberg, Nürnberg, Germany
| | - Alexander Glahn
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Medical School Hannover, Hannover, Germany
| | - Patrick Bach
- Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health, Medical Faculty Mannheim. Heidelberg University, Heidelberg, Germany
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
7
|
Miller AP, Bogdan R, Agrawal A, Hatoum AS. Generalized genetic liability to substance use disorders. J Clin Invest 2024; 134:e172881. [PMID: 38828723 PMCID: PMC11142744 DOI: 10.1172/jci172881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024] Open
Abstract
Lifetime and temporal co-occurrence of substance use disorders (SUDs) is common and compared with individual SUDs is characterized by greater severity, additional psychiatric comorbidities, and worse outcomes. Here, we review evidence for the role of generalized genetic liability to various SUDs. Coaggregation of SUDs has familial contributions, with twin studies suggesting a strong contribution of additive genetic influences undergirding use disorders for a variety of substances (including alcohol, nicotine, cannabis, and others). GWAS have documented similarly large genetic correlations between alcohol, cannabis, and opioid use disorders. Extending these findings, recent studies have identified multiple genomic loci that contribute to common risk for these SUDs and problematic tobacco use, implicating dopaminergic regulatory and neuronal development mechanisms in the pathophysiology of generalized SUD genetic liability, with certain signals demonstrating cross-species and translational validity. Overlap with genetic signals for other externalizing behaviors, while substantial, does not explain the entirety of the generalized genetic signal for SUD. Polygenic scores (PGS) derived from the generalized genetic liability to SUDs outperform PGS for individual SUDs in prediction of serious mental health and medical comorbidities. Going forward, it will be important to further elucidate the etiology of generalized SUD genetic liability by incorporating additional SUDs, evaluating clinical presentation across the lifespan, and increasing the granularity of investigation (e.g., specific transdiagnostic criteria) to ultimately improve the nosology, prevention, and treatment of SUDs.
Collapse
Affiliation(s)
| | - Ryan Bogdan
- Department of Psychological and Brain Sciences, Washington University in St. Louis, St. Louis, Missouri, USA
| | | | - Alexander S. Hatoum
- Department of Psychological and Brain Sciences, Washington University in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
8
|
Acuña AM, Park C, Leyrer-Jackson JM, Olive MF. Promising immunomodulators for management of substance and alcohol use disorders. Expert Opin Pharmacother 2024; 25:867-884. [PMID: 38803314 PMCID: PMC11216154 DOI: 10.1080/14656566.2024.2360653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024]
Abstract
INTRODUCTION The neuroimmune system has emerged as a novel target for the treatment of substance use disorders (SUDs), with immunomodulation producing encouraging therapeutic benefits in both preclinical and clinical settings. AREAS COVERED In this review, we describe the mechanism of action and immune response to methamphetamine, opioids, cocaine, and alcohol. We then discuss off-label use of immunomodulators as adjunctive therapeutics in the treatment of neuropsychiatric disorders, demonstrating their potential efficacy in affective and behavioral disorders. We then discuss in detail the mechanism of action and recent findings regarding the use of ibudilast, minocycline, probenecid, dexmedetomidine, pioglitazone, and cannabidiol to treat (SUDs). These immunomodulators are currently being investigated in clinical trials described herein, specifically for their potential to decrease substance use, withdrawal severity, central and peripheral inflammation, comorbid neuropsychiatric disorder symptomology, as well as their ability to improve cognitive outcomes. EXPERT OPINION We argue that although mixed, findings from recent preclinical and clinical studies underscore the potential benefit of immunomodulation in the treatment of the behavioral, cognitive, and inflammatory processes that underlie compulsive substance use.
Collapse
Affiliation(s)
- Amanda M. Acuña
- Department of Psychology, Behavioral Neuroscience and Comparative Psychology Area, Arizona State University, Tempe, Arizona, USA
| | - Connor Park
- Department of Biomedical Sciences, Creighton University School of Medicine – Phoenix, Phoenix, Arizona, USA
| | - Jonna M. Leyrer-Jackson
- Department of Biomedical Sciences, Creighton University School of Medicine – Phoenix, Phoenix, Arizona, USA
| | - M. Foster Olive
- Department of Psychology, Behavioral Neuroscience and Comparative Psychology Area, Arizona State University, Tempe, Arizona, USA
| |
Collapse
|
9
|
Kirsch DE, Ray LA, Wassum KM, Grodin EN. Anterior cingulate and medial prefrontal cortex alcohol cue reactivity varies as a function of drink preference in alcohol use disorder. Drug Alcohol Depend 2024; 256:111123. [PMID: 38367535 DOI: 10.1016/j.drugalcdep.2024.111123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 01/11/2024] [Accepted: 02/05/2024] [Indexed: 02/19/2024]
Abstract
BACKGROUND Functional MRI visual cue reactivity studies have not considered that brain responses to various alcohol-containing beverage types may vary as a function of an individual's drinking patterns and preferences. This study tested whether the brain's reward system responds differently to visual cues associated with an individuals' most commonly consumed ("preferred") alcohol beverage compared with less commonly consumed ("non-preferred") alcohol beverages in individuals with alcohol use disorder (AUD). METHODS Participants (N=70) with current AUD completed a standard visual alcohol cue reactivity procedure during fMRI and reported recent alcohol use through the Timeline Followback interview. Alcohol use patterns were used to infer drink preference. Repeated measure ANCOVAs were used to evaluate differences in subjective craving (alcohol urge) and neural reactivity to cues of individual's "preferred" versus "non-preferred" alcohol beverages. RESULTS Fifty-four (77%) participants were determined to have a "preferred" alcohol beverage, as defined by their pattern of alcohol use. These participants reported greater subjective alcohol urge (p=0.02) and activation in the anterior cingulate cortex (ACC) (p=0.005) and medial prefrontal cortex (mPFC) (p=0.001)) in response to visual cues associated with their "preferred" versus "non-preferred" alcohol beverage. Individuals with an alcohol preference did not differ from those with no alcohol preference on subjective or neural responses to their "preferred" and "non-preferred" alcohol cues. DISCUSSION Results suggest alcohol cue-elicited subjective and neural responses vary as a function of alcohol beverage preference in individuals with AUD and a behaviorally defined alcohol preference. Stronger ACC and mPFC activation may reflect greater subjective value of an individual's "preferred" alcohol beverage cue.
Collapse
Affiliation(s)
- Dylan E Kirsch
- Department of Psychology, University of California, Los Angeles, 1285 Franz Hall, Box 951563, Los Angeles, CA 90095-1563, USA
| | - Lara A Ray
- Department of Psychology, University of California, Los Angeles, 1285 Franz Hall, Box 951563, Los Angeles, CA 90095-1563, USA; Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA, USA.
| | - Kate M Wassum
- Department of Psychology, University of California, Los Angeles, 1285 Franz Hall, Box 951563, Los Angeles, CA 90095-1563, USA
| | - Erica N Grodin
- Department of Psychology, University of California, Los Angeles, 1285 Franz Hall, Box 951563, Los Angeles, CA 90095-1563, USA; Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
10
|
Ball RL, Bogue MA, Liang H, Srivastava A, Ashbrook DG, Lamoureux A, Gerring MW, Hatoum AS, Kim MJ, He H, Emerson J, Berger AK, Walton DO, Sheppard K, El Kassaby B, Castellanos F, Kunde-Ramamoorthy G, Lu L, Bluis J, Desai S, Sundberg BA, Peltz G, Fang Z, Churchill GA, Williams RW, Agrawal A, Bult CJ, Philip VM, Chesler EJ. GenomeMUSter mouse genetic variation service enables multitrait, multipopulation data integration and analysis. Genome Res 2024; 34:145-159. [PMID: 38290977 PMCID: PMC10903950 DOI: 10.1101/gr.278157.123] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 01/10/2024] [Indexed: 02/01/2024]
Abstract
Hundreds of inbred mouse strains and intercross populations have been used to characterize the function of genetic variants that contribute to disease. Thousands of disease-relevant traits have been characterized in mice and made publicly available. New strains and populations including consomics, the collaborative cross, expanded BXD, and inbred wild-derived strains add to existing complex disease mouse models, mapping populations, and sensitized backgrounds for engineered mutations. The genome sequences of inbred strains, along with dense genotypes from others, enable integrated analysis of trait-variant associations across populations, but these analyses are hampered by the sparsity of genotypes available. Moreover, the data are not readily interoperable with other resources. To address these limitations, we created a uniformly dense variant resource by harmonizing multiple data sets. Missing genotypes were imputed using the Viterbi algorithm with a data-driven technique that incorporates local phylogenetic information, an approach that is extendable to other model organisms. The result is a web- and programmatically accessible data service called GenomeMUSter, comprising single-nucleotide variants covering 657 strains at 106.8 million segregating sites. Interoperation with phenotype databases, analytic tools, and other resources enable a wealth of applications, including multitrait, multipopulation meta-analysis. We show this in cross-species comparisons of type 2 diabetes and substance use disorder meta-analyses, leveraging mouse data to characterize the likely role of human variant effects in disease. Other applications include refinement of mapped loci and prioritization of strain backgrounds for disease modeling to further unlock extant mouse diversity for genetic and genomic studies in health and disease.
Collapse
Affiliation(s)
- Robyn L Ball
- The Jackson Laboratory, Bar Harbor, Maine 04609, USA;
| | - Molly A Bogue
- The Jackson Laboratory, Bar Harbor, Maine 04609, USA
| | | | - Anuj Srivastava
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut 06032, USA
| | - David G Ashbrook
- University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | | | | | - Alexander S Hatoum
- Psychological and Brain Sciences, Washington University in St. Louis, St. Louis, Missouri 63130, USA
- Artificial Intelligence and the Internet of Things Institute, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Matthew J Kim
- University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Hao He
- The Jackson Laboratory, Bar Harbor, Maine 04609, USA
| | - Jake Emerson
- The Jackson Laboratory, Bar Harbor, Maine 04609, USA
| | | | | | | | | | | | | | - Lu Lu
- University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | - John Bluis
- The Jackson Laboratory, Bar Harbor, Maine 04609, USA
| | - Sejal Desai
- The Jackson Laboratory, Bar Harbor, Maine 04609, USA
| | | | - Gary Peltz
- Department of Anesthesia, Pain and Perioperative Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Zhuoqing Fang
- Department of Anesthesia, Pain and Perioperative Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| | | | - Robert W Williams
- University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | - Arpana Agrawal
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Carol J Bult
- The Jackson Laboratory, Bar Harbor, Maine 04609, USA
| | | | | |
Collapse
|
11
|
Cruz B, Borgonetti V, Bajo M, Roberto M. Sex-dependent factors of alcohol and neuroimmune mechanisms. Neurobiol Stress 2023; 26:100562. [PMID: 37601537 PMCID: PMC10432974 DOI: 10.1016/j.ynstr.2023.100562] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/25/2023] [Accepted: 08/02/2023] [Indexed: 08/22/2023] Open
Abstract
Excessive alcohol use disrupts neuroimmune signaling across various cell types, including neurons, microglia, and astrocytes. The present review focuses on recent, albeit limited, evidence of sex differences in biological factors that mediate neuroimmune responses to alcohol and underlying neuroimmune systems that may influence alcohol drinking behaviors. Females are more vulnerable than males to the neurotoxic and negative consequences of chronic alcohol drinking, reflected by elevations of pro-inflammatory cytokines and inflammatory mediators. Differences in cytokine, microglial, astrocytic, genomic, and transcriptomic evidence suggest females are more reactive than males to neuroinflammatory changes after chronic alcohol exposure. The growing body of evidence supports that innate immune factors modulate synaptic transmission, providing a mechanistic framework to examine sex differences across neurocircuitry. Targeting neuroimmune signaling may be a viable strategy for treating AUD, but more research is needed to understand sex-specific differences in alcohol drinking and neuroimmune mechanisms.
Collapse
Affiliation(s)
- Bryan Cruz
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA, 92073
| | - Vittoria Borgonetti
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA, 92073
| | - Michal Bajo
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA, 92073
| | - Marisa Roberto
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA, 92073
| |
Collapse
|
12
|
Ball RL, Bogue MA, Liang H, Srivastava A, Ashbrook DG, Lamoureux A, Gerring MW, Hatoum AS, Kim M, He H, Emerson J, Berger AK, Walton DO, Sheppard K, Kassaby BE, Castellanos F, Kunde-Ramamoorthy G, Lu L, Bluis J, Desai S, Sundberg BA, Peltz G, Fang Z, Churchill GA, Williams RW, Agrawal A, Bult CJ, Philip VM, Chesler EJ. GenomeMUSter mouse genetic variation service enables multi-trait, multi-population data integration and analyses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.08.552506. [PMID: 37609331 PMCID: PMC10441370 DOI: 10.1101/2023.08.08.552506] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Hundreds of inbred laboratory mouse strains and intercross populations have been used to functionalize genetic variants that contribute to disease. Thousands of disease relevant traits have been characterized in mice and made publicly available. New strains and populations including the Collaborative Cross, expanded BXD and inbred wild-derived strains add to set of complex disease mouse models, genetic mapping resources and sensitized backgrounds against which to evaluate engineered mutations. The genome sequences of many inbred strains, along with dense genotypes from others could allow integrated analysis of trait - variant associations across populations, but these analyses are not feasible due to the sparsity of genotypes available. Moreover, the data are not readily interoperable with other resources. To address these limitations, we created a uniformly dense data resource by harmonizing multiple variant datasets. Missing genotypes were imputed using the Viterbi algorithm with a data-driven technique that incorporates local phylogenetic information, an approach that is extensible to other model organism species. The result is a web- and programmatically-accessible data service called GenomeMUSter ( https://muster.jax.org ), comprising allelic data covering 657 strains at 106.8M segregating sites. Interoperation with phenotype databases, analytic tools and other resources enable a wealth of applications including multi-trait, multi-population meta-analysis. We demonstrate this in a cross-species comparison of the meta-analysis of Type 2 Diabetes and of substance use disorders, resulting in the more specific characterization of the role of human variant effects in light of mouse phenotype data. Other applications include refinement of mapped loci and prioritization of strain backgrounds for disease modeling to further unlock extant mouse diversity for genetic and genomic studies in health and disease.
Collapse
|
13
|
Grodin EN, Meredith LR, Burnette EM, Miotto K, Irwin MR, Ray LA. Baseline C-reactive protein levels are predictive of treatment response to a neuroimmune modulator in individuals with an alcohol use disorder: a preliminary study. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2023; 49:333-344. [PMID: 36282988 PMCID: PMC10840759 DOI: 10.1080/00952990.2022.2124918] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 09/07/2022] [Accepted: 09/12/2022] [Indexed: 01/25/2023]
Abstract
Background: Inflammation is implicated in alcohol use disorder (AUD). Ibudilast, a neuroimmune modulator, shows promise for the treatment of AUD. Elevated inflammation, indicated by high levels of C-reactive protein (CRP), represents a possible subtype of AUD, which may be associated with treatment response to ibudilast.Objectives: The current study evaluated CRP as a predictor of treatment response to ibudilast; hypothesizing that ibudilast would be more effective at reducing drinking and alcohol cue-reactivity in individuals with higher CRP levels.Methods: This is a secondary analysis of a clinical trial of ibudilast for AUD, which found that ibudilast reduced heavy drinking in individuals with AUD. Fifty-one individuals were randomized to receive ibudilast (n = 24 [16 M/8F]) or placebo (n = 27 [18 M/9F]) for two weeks. Participants provided blood samples at baseline to assess CRP levels, completed daily assessments of alcohol use, and an fMRI alcohol cue-reactivity task at study mid-point. Models tested the effects of medication, CRP levels, and their interaction on drinks per drinking day and alcohol cue-reactivity.Results: There was a significant interaction between medication and CRP (F = 3.80, p = .03), such that the ibudilast high CRP group had fewer drinks per drinking day compared to the ibudilast low CRP group. CRP moderated the effect of medication on brain activation in a cluster extending from the left inferior frontal gyrus to the right-dorsal striatum (Z = 4.55, p < .001). This interaction was driven by attenuated cue-reactivity in the ibudilast high CRP group relative to the ibudilast low CRP and placebo high CRP groups.Conclusions: This study serves as an initial investigation into predictors of clinical response to ibudilast treatment and suggests that a baseline proinflammatory profile may enhance clinical efficacy.
Collapse
Affiliation(s)
- Erica N. Grodin
- Department of Psychology, University of California at Los Angeles, Los Angeles, CA
| | - Lindsay R. Meredith
- Department of Psychology, University of California at Los Angeles, Los Angeles, CA
| | - Elizabeth M. Burnette
- Department of Psychology, University of California at Los Angeles, Los Angeles, CA
- Neuroscience Interdepartmental Program, University of California at Los Angeles, Los Angeles, CA
| | - Karen Miotto
- Department of Psychiatry and Biobehavioral Sciences, University of California at Los Angeles, Los Angeles, CA
| | - Michael R. Irwin
- Department of Psychology, University of California at Los Angeles, Los Angeles, CA
- Department of Psychiatry and Biobehavioral Sciences, University of California at Los Angeles, Los Angeles, CA
- Jane & Terry Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA
- Cousins Center for Psychoneuroimmunology, University of California at Los Angeles, Los Angeles, CA
| | - Lara A. Ray
- Department of Psychology, University of California at Los Angeles, Los Angeles, CA
- Department of Psychiatry and Biobehavioral Sciences, University of California at Los Angeles, Los Angeles, CA
- Brain Research Institute, University of California, Los Angeles, CA
| |
Collapse
|
14
|
Varodayan FP, Pahng AR, Davis TD, Gandhi P, Bajo M, Steinman MQ, Kiosses WB, Blednov YA, Burkart MD, Edwards S, Roberts AJ, Roberto M. Chronic ethanol induces a pro-inflammatory switch in interleukin-1β regulation of GABAergic signaling in the medial prefrontal cortex of male mice. Brain Behav Immun 2023; 110:125-139. [PMID: 36863493 PMCID: PMC10106421 DOI: 10.1016/j.bbi.2023.02.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 02/20/2023] [Accepted: 02/25/2023] [Indexed: 03/04/2023] Open
Abstract
Neuroimmune pathways regulate brain function to influence complex behavior and play a role in several neuropsychiatric diseases, including alcohol use disorder (AUD). In particular, the interleukin-1 (IL-1) system has emerged as a key regulator of the brain's response to ethanol (alcohol). Here we investigated the mechanisms underlying ethanol-induced neuroadaptation of IL-1β signaling at GABAergic synapses in the prelimbic region of the medial prefrontal cortex (mPFC), an area responsible for integrating contextual information to mediate conflicting motivational drives. We exposed C57BL/6J male mice to the chronic intermittent ethanol vapor-2 bottle choice paradigm (CIE-2BC) to induce ethanol dependence, and conducted ex vivo electrophysiology and molecular analyses. We found that the IL-1 system regulates basal mPFC function through its actions at inhibitory synapses on prelimbic layer 2/3 pyramidal neurons. IL-1β can selectively recruit either neuroprotective (PI3K/Akt) or pro-inflammatory (MyD88/p38 MAPK) mechanisms to produce opposing synaptic effects. In ethanol naïve conditions, there was a strong PI3K/Akt bias leading to a disinhibition of pyramidal neurons. Ethanol dependence produced opposite IL-1 effects - enhanced local inhibition via a switch in IL-1β signaling to the canonical pro-inflammatory MyD88 pathway. Ethanol dependence also increased cellular IL-1β in the mPFC, while decreasing expression of downstream effectors (Akt, p38 MAPK). Thus, IL-1β may represent a key neural substrate in ethanol-induced cortical dysfunction. As the IL-1 receptor antagonist (kineret) is already FDA-approved for other diseases, this work underscores the high therapeutic potential of IL-1 signaling/neuroimmune-based treatments for AUD.
Collapse
Affiliation(s)
- F P Varodayan
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA; Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, Binghamton, NY, USA
| | - A R Pahng
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA, USA; Southeast Louisiana Veterans Health Care System, New Orleans, LA, USA
| | - T D Davis
- Department of Chemistry & Biochemistry, University of California, San Diego, La Jolla, CA, USA; Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Binghamton University-SUNY, Binghamton, NY, USA
| | - P Gandhi
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - M Bajo
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - M Q Steinman
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - W B Kiosses
- Microscopy Core Imaging Facility, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Y A Blednov
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX 78712, USA
| | - M D Burkart
- Department of Chemistry & Biochemistry, University of California, San Diego, La Jolla, CA, USA
| | - S Edwards
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - A J Roberts
- Animal Models Core Facility, The Scripps Research Institute, La Jolla, CA, USA
| | - M Roberto
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
15
|
Zeng J, You L, Yang F, Luo Y, Yu S, Yan J, Liu M, Yang X. A meta-analysis of the neural substrates of monetary reward anticipation and outcome in alcohol use disorder. Hum Brain Mapp 2023; 44:2841-2861. [PMID: 36852619 PMCID: PMC10089105 DOI: 10.1002/hbm.26249] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 01/23/2023] [Accepted: 02/13/2023] [Indexed: 03/01/2023] Open
Abstract
The capacity to anticipate and detect rewarding outcomes is fundamental for the development of adaptive decision-making and goal-oriented behavior. Delineating the neural correlates of different stages of reward processing is imperative for understanding the neurobiological mechanism underlying alcohol use disorder (AUD). To examine the neural correlates of monetary anticipation and outcome in AUD patients, we performed two separate voxel-wise meta-analyses of functional neuroimaging studies, including 12 studies investigating reward anticipation and 7 studies investigating reward outcome using the monetary incentive delay task. During the anticipation stage, AUD patients displayed decreased activation in response to monetary cues in mesocortical-limbic circuits and sensory areas, including the ventral striatum (VS), insula, hippocampus, inferior occipital gyrus, supramarginal gyrus, lingual gyrus and fusiform gyrus. During the outcome stage, AUD patients exhibited reduced activation in the dorsal striatum, VS and insula, and increased activation in the orbital frontal cortex and medial temporal area. Our findings suggest that different activation patterns are associated with nondrug rewards during different reward processing stages, potentially reflecting a changed sensitivity to monetary reward in AUD.
Collapse
Affiliation(s)
- Jianguang Zeng
- School of Economics and Business AdministrationChongqing UniversityChongqingChina
| | - Lantao You
- School of Economics and Business AdministrationChongqing UniversityChongqingChina
| | - Fan Yang
- Department of Ultrasonography, West China Second University HospitalSichuan UniversityChengduChina
- Chengdu Chenghua District Maternal and Child Health HospitalSichuan UniversityChengduChina
| | - Ya Luo
- Department of Psychiatry, State Key Lab of BiotherapyWest China Hospital of Sichuan UniversityChengduChina
| | - Shuxian Yu
- School of Economics and Business AdministrationChongqing UniversityChongqingChina
| | - Jiangnan Yan
- School of Economics and Business AdministrationChongqing UniversityChongqingChina
| | - Mengqi Liu
- Department of RadiologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Xun Yang
- School of Public AffairsChongqing UniversityChongqingChina
| |
Collapse
|
16
|
Hatoum AS, Colbert SM, Johnson EC, Huggett SB, Deak JD, Pathak G, Jennings MV, Paul SE, Karcher NR, Hansen I, Baranger DA, Edwards A, Grotzinger A, Tucker-Drob EM, Kranzler HR, Davis LK, Sanchez-Roige S, Polimanti R, Gelernter J, Edenberg HJ, Bogdan R, Agrawal A. Multivariate genome-wide association meta-analysis of over 1 million subjects identifies loci underlying multiple substance use disorders. NATURE. MENTAL HEALTH 2023; 1:210-223. [PMID: 37250466 PMCID: PMC10217792 DOI: 10.1038/s44220-023-00034-y] [Citation(s) in RCA: 84] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 02/10/2023] [Indexed: 05/31/2023]
Abstract
Genetic liability to substance use disorders can be parsed into loci that confer general or substance-specific addiction risk. We report a multivariate genome-wide association meta-analysis that disaggregates general and substance-specific loci for published summary statistics of problematic alcohol use, problematic tobacco use, cannabis use disorder, and opioid use disorder in a sample of 1,025,550 individuals of European descent and 92,630 individuals of African descent. Nineteen independent SNPs were genome-wide significant (P < 5e-8) for the general addiction risk factor (addiction-rf), which showed high polygenicity. Across ancestries, PDE4B was significant (among other genes), suggesting dopamine regulation as a cross-substance vulnerability. An addiction-rf polygenic risk score was associated with substance use disorders, psychopathologies, somatic conditions, and environments associated with the onset of addictions. Substance-specific loci (9 for alcohol, 32 for tobacco, 5 for cannabis, 1 for opioids) included metabolic and receptor genes. These findings provide insight into genetic risk loci for substance use disorders that could be leveraged as treatment targets.
Collapse
Affiliation(s)
- Alexander S. Hatoum
- Washington University School of Medicine, Department of
Psychiatry, Saint Louis, USA
| | - Sarah M.C. Colbert
- Washington University School of Medicine, Department of
Psychiatry, Saint Louis, USA
| | - Emma C. Johnson
- Washington University School of Medicine, Department of
Psychiatry, Saint Louis, USA
| | | | - Joseph D. Deak
- Department of Psychiatry, Division of Human Genetics, Yale
School of Medicine, New Haven, CT, USA
- Veterans Affairs Connecticut Healthcare System, West Haven,
CT, USA
| | - Gita Pathak
- Department of Psychiatry, Division of Human Genetics, Yale
School of Medicine, New Haven, CT, USA
| | - Mariela V. Jennings
- UC San Diego School of Medicine, Department of Psychiatry,
San Diego, CA, USA
| | - Sarah E. Paul
- Department of Psychological & Brain Sciences,
Washington University in St. Louis
| | - Nicole R. Karcher
- Washington University School of Medicine, Department of
Psychiatry, Saint Louis, USA
| | - Isabella Hansen
- Department of Psychological & Brain Sciences,
Washington University in St. Louis
| | - David A.A. Baranger
- Washington University School of Medicine, Department of
Psychiatry, Saint Louis, USA
| | - Alexis Edwards
- Virginia Institute of Psychiatric and Behavioral Genetics,
Virginia Commonwealth University, Richmond, VA, USA
| | - Andrew Grotzinger
- University of Colorado-Boulder, Institute for Behavioral
Genetics, Boulder, CO, USA
| | | | - Elliot M. Tucker-Drob
- University of Texas at Austin, Department of Psychology and
Population Research Center, Austin, TX, USA
| | - Henry R. Kranzler
- Center for Studies of Addiction, Department of
Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia,
PA, USA
- VISN 4 MIRECC, Crescenz VAMC, Philadelphia, PA, USA
| | - Lea K. Davis
- Department of Medicine, Division of Genetic Medicine,
Vanderbilt University, Nashville, TN, USA
- Department of Psychiatry and Behavioral Sciences,
Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Biomedical Informatics, Vanderbilt
University Medical Center, Nashville, TN, USA
| | - Sandra Sanchez-Roige
- UC San Diego School of Medicine, Department of Psychiatry,
San Diego, CA, USA
- Department of Medicine, Division of Genetic Medicine,
Vanderbilt University, Nashville, TN, USA
| | - Renato Polimanti
- Department of Psychiatry, Division of Human Genetics, Yale
School of Medicine, New Haven, CT, USA
- Veterans Affairs Connecticut Healthcare System, West Haven,
CT, USA
| | - Joel Gelernter
- Department of Psychiatry, Division of Human Genetics, Yale
School of Medicine, New Haven, CT, USA
- University of Texas at Austin, Department of Psychology and
Population Research Center, Austin, TX, USA
- Department of Genetics, Yale School of Medicine, New
Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New
Haven, CT, USA
| | - Howard J. Edenberg
- Department of Medical and Molecular Genetics, Indiana
University School of Medicine, Indianapolis, IN, USA
- Department of Biochemistry and Molecular Biology, Indiana
University School of Medicine, Indianapolis, IN, USA
| | - Ryan Bogdan
- Department of Psychological & Brain Sciences,
Washington University in St. Louis
| | - Arpana Agrawal
- Washington University School of Medicine, Department of
Psychiatry, Saint Louis, USA
| |
Collapse
|
17
|
Adams C, Perry N, Conigrave J, Hurzeler T, Stevens J, Yacou Dunbar KP, Sweeney A, Lee K, Sutherland G, Haber P, Morley KC. Central markers of neuroinflammation in alcohol use disorder: A meta-analysis of neuroimaging, cerebral spinal fluid, and postmortem studies. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2023; 47:197-208. [PMID: 36852781 DOI: 10.1111/acer.14992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/23/2022] [Accepted: 11/30/2022] [Indexed: 03/01/2023]
Abstract
INTRODUCTION AND AIMS There is emerging evidence that heavy long-term alcohol consumption may alter the neuroimmune profile. We conducted a meta-analysis of the association between alcohol use disorder (AUD) and the extent of neuroinflammation using cerebrospinal (CSF), PET (Positron Emission Tomography), and postmortem studies. DESIGN AND METHODS A comprehensive search of electronic databases was conducted using the Preferred Reporting Items for Systematic Review and Meta-Analysis Protocols (PRISMA-P) for AUD-related terms in combination with neuroinflammatory markers and cytokine- and chemokine-related terms for CSF, PET, and postmortem studies. Participants had to meet established criteria for AUD and/or heavy alcohol consumption with dependence features and be compared with healthy controls. Papers retrieved were assessed for inclusion criteria and a critical appraisal was completed using the Newcastle-Ottawa Scale. A meta-analysis was conducted on postmortem and PET studies. RESULTS Eleven papers met the inclusion criteria with CSF, PET, and postmortem studies included in the final analysis. Postmortem studies demonstrate significant heterogeneity (𝑄 (14) = 62.02, 𝑝 < 0.001), with the alcohol group showing higher levels of neuroimmune markers than controls (𝑑 = 1.50 [95% CI 0.56, 2.45]). PET studies demonstrated a lower [11 C] PBR28 total volume of distribution (V T ) for translocator protein in the hippocampus (g = -1.95 [95% CI -2.72, -1.18], p < 0.001) of the alcohol group compared to controls. CONCLUSION There is emerging evidence across multiple diagnostic modalities that alcohol impacts neuroimmune signaling in the human brain.
Collapse
Affiliation(s)
- Claire Adams
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Edith Collins Centre (Translational Research in Alcohol Drugs and Toxicology), Drug Health Services, Sydney Local Health District, Sydney, New South Wales, Australia
| | - Nina Perry
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - James Conigrave
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Edith Collins Centre (Translational Research in Alcohol Drugs and Toxicology), Drug Health Services, Sydney Local Health District, Sydney, New South Wales, Australia
| | - Tristan Hurzeler
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Edith Collins Centre (Translational Research in Alcohol Drugs and Toxicology), Drug Health Services, Sydney Local Health District, Sydney, New South Wales, Australia
| | - Julia Stevens
- NSW Brain Tissue Resource Centre and School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Kristiane P Yacou Dunbar
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Alicia Sweeney
- NSW Brain Tissue Resource Centre and School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Kylie Lee
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Edith Collins Centre (Translational Research in Alcohol Drugs and Toxicology), Drug Health Services, Sydney Local Health District, Sydney, New South Wales, Australia
| | - Greg Sutherland
- NSW Brain Tissue Resource Centre and School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Paul Haber
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Edith Collins Centre (Translational Research in Alcohol Drugs and Toxicology), Drug Health Services, Sydney Local Health District, Sydney, New South Wales, Australia
| | - Kirsten C Morley
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Edith Collins Centre (Translational Research in Alcohol Drugs and Toxicology), Drug Health Services, Sydney Local Health District, Sydney, New South Wales, Australia
| |
Collapse
|
18
|
Correll CU, Solmi M, Cortese S, Fava M, Højlund M, Kraemer HC, McIntyre RS, Pine DS, Schneider LS, Kane JM. The future of psychopharmacology: a critical appraisal of ongoing phase 2/3 trials, and of some current trends aiming to de-risk trial programmes of novel agents. World Psychiatry 2023; 22:48-74. [PMID: 36640403 PMCID: PMC9840514 DOI: 10.1002/wps.21056] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/14/2022] [Indexed: 01/15/2023] Open
Abstract
Despite considerable progress in pharmacotherapy over the past seven decades, many mental disorders remain insufficiently treated. This situation is in part due to the limited knowledge of the pathophysiology of these disorders and the lack of biological markers to stratify and individualize patient selection, but also to a still restricted number of mechanisms of action being targeted in monotherapy or combination/augmentation treatment, as well as to a variety of challenges threatening the successful development and testing of new drugs. In this paper, we first provide an overview of the most promising drugs with innovative mechanisms of action that are undergoing phase 2 or 3 testing for schizophrenia, bipolar disorder, major depressive disorder, anxiety and trauma-related disorders, substance use disorders, and dementia. Promising repurposing of established medications for new psychiatric indications, as well as variations in the modulation of dopamine, noradrenaline and serotonin receptor functioning, are also considered. We then critically discuss the clinical trial parameters that need to be considered in depth when developing and testing new pharmacological agents for the treatment of mental disorders. Hurdles and perils threatening success of new drug development and testing include inadequacy and imprecision of inclusion/exclusion criteria and ratings, sub-optimally suited clinical trial participants, multiple factors contributing to a large/increasing placebo effect, and problems with statistical analyses. This information should be considered in order to de-risk trial programmes of novel agents or known agents for novel psychiatric indications, increasing their chances of success.
Collapse
Affiliation(s)
- Christoph U Correll
- Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin Berlin, Berlin, Germany
- Department of Psychiatry, Zucker Hillside Hospital, Northwell Health, Glen Oaks, NY, USA
- Department of Psychiatry and Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
- Center for Psychiatric Neuroscience, Feinstein Institute for Medical Research, Manhasset, NY, USA
| | - Marco Solmi
- Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin Berlin, Berlin, Germany
- Department of Psychiatry, University of Ottawa, Ottawa, ON, Canada
- Department of Mental Health, Ottawa Hospital, Ottawa, ON, Canada
- Ottawa Hospital Research Institute (OHRI) Clinical Epidemiology Program, University of Ottawa, Ottawa, ON, Canada
- School of Epidemiology and Public Health, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Centre for Innovation in Mental Health, School of Psychology, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK
| | - Samuele Cortese
- Centre for Innovation in Mental Health, School of Psychology, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK
- Clinical and Experimental Sciences (CNS and Psychiatry), Faculty of Medicine, University of Southampton, Southampton, UK
- Solent NHS Trust, Southampton, UK
- Division of Psychiatry and Applied Psychology, School of Medicine, University of Nottingham, Nottingham, UK
- Hassenfeld Children's Hospital at NYU Langone, New York University Child Study Center, New York, NY, USA
| | - Maurizio Fava
- Depression Clinical and Research Program, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Mikkel Højlund
- Department of Public Health, Clinical Pharmacology, Pharmacy and Environmental Medicine, University of Southern Denmark, Odense, Denmark
- Mental Health Services in the Region of Southern Denmark, Department of Psychiatry Aabenraa, Aabenraa, Denmark
| | - Helena C Kraemer
- Department of Psychiatry and Behavioral Sciences, Stanford University, Cupertino, CA, USA
| | - Roger S McIntyre
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Canadian Rapid Treatment Center of Excellence, Mississauga, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Department of Pharmacology, University of Toronto, Toronto, ON, Canada
- Brain and Cognition Discovery Foundation, Toronto, ON, Canada
| | - Daniel S Pine
- Section on Developmental Affective Neuroscience, National Institute of Mental Health, Bethesda, MD, USA
| | - Lon S Schneider
- Department of Psychiatry and Behavioral Sciences, and Department of Neurology, Keck School of Medicine, and L. Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - John M Kane
- Department of Psychiatry, Zucker Hillside Hospital, Northwell Health, Glen Oaks, NY, USA
- Department of Psychiatry and Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
- Center for Psychiatric Neuroscience, Feinstein Institute for Medical Research, Manhasset, NY, USA
| |
Collapse
|
19
|
Hou X, Rong C, Zhang Q, Song S, Cong Y, Zhang HT. Cyclic Nucleotide Phosphodiesterases in Alcohol Use Disorders: Involving Gut Microbiota. Int J Neuropsychopharmacol 2022; 26:70-79. [PMID: 36087271 PMCID: PMC9850663 DOI: 10.1093/ijnp/pyac060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 08/18/2022] [Accepted: 09/09/2022] [Indexed: 01/22/2023] Open
Abstract
Alcohol abuse is 1 of the most significant public health problems in the world. Chronic, excessive alcohol consumption not only causes alcohol use disorder (AUD) but also changes the gut and lung microbiota, including bacterial and nonbacterial types. Both types of microbiota can release toxins, further damaging the gastrointestinal and respiratory tracts; causing inflammation; and impairing the functions of the liver, lung, and brain, which in turn deteriorate AUD. Phosphodiesterases (PDEs) are critical in the control of intracellular cyclic nucleotides, including cyclic adenosine monophosphate and cyclic guanosine monophosphate. Inhibition of certain host PDEs reduces alcohol consumption and attenuates alcohol-related impairment. These PDEs are also expressed in the microbiota and may play a role in controlling microbiota-associated inflammation. Here, we summarize the influences of alcohol on gut/lung bacterial and nonbacterial microbiota as well as on the gut-liver/brain/lung axis. We then discuss the relationship between gut and lung microbiota-mediated PDE signaling and AUD consequences in addition to highlighting PDEs as potential targets for treatment of AUD.
Collapse
Affiliation(s)
- Xueqin Hou
- Correspondence: Xueqin Hou, PhD, Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong 271016, P.R. China ()
| | | | - Qiwei Zhang
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, Shandong 271016, P.R. China
| | - Shuangshuang Song
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, Shandong 271016, P.R. China
| | - Yifan Cong
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, Shandong 271016, P.R. China
| | - Han-Ting Zhang
- Han-Ting Zhang, MD, PhD, Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, Shandong 266073, P.R. China ()
| |
Collapse
|
20
|
Nieto SJ, Grodin EN, Burnette EM, Cahill CM, Ray LA. Pain Catastrophizing Is Associated With Increased Alcohol Cue-Elicited Neural Activity Among Individuals With Alcohol Use Disorder. Alcohol Alcohol 2022; 57:727-733. [PMID: 35788255 DOI: 10.1093/alcalc/agac029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 05/31/2022] [Accepted: 06/03/2022] [Indexed: 11/13/2022] Open
Abstract
AIMS The current study examined the association between pain catastrophizing and alcohol cue-elicited brain activation in individuals with alcohol use disorder (AUD). METHODS Non-treatment seeking heavy drinkers with AUD (n = 45; 28 males) completed self-report measures of pain catastrophizing and alcohol use/problems as part of a clinical trial of the neuroimmune modulator ibudilast. Participants were randomized to either placebo (n = 25) or ibudilast (n = 20) and completed an functional magnetic resonance imaging (fMRI) scan to assess neural activation to alcohol cues 1 week into the medication trial. Multiple linear regression examined whether pain catastrophizing predicted cue-induced activation in a priori regions of interest, namely the dorsal and ventral striatum (VS). An exploratory whole-brain analysis was conducted to assess the relationship between pain catastrophizing and neural alcohol cue reactivity. RESULTS Pain catastrophizing predicted greater cue-induced activation in the dorsal (b = 0.006; P = 0.03) but not VS controlling for medication. Pain catastrophizing was positively associated with neural activation to alcohol cues in regions including the bilateral thalamus, left precuneus and left frontal pole. CONCLUSION Greater pain catastrophizing is associated with greater cue-induced neural activation in brain regions sub-serving habits and compulsive alcohol use. These findings provide initial support for a neural mechanism by which pain catastrophizing may drive alcohol craving among individuals with AUD.
Collapse
Affiliation(s)
- Steven J Nieto
- Department of Psychology, University of California at Los Angeles, Los Angeles, CA, USA
| | - Erica N Grodin
- Department of Psychology, University of California at Los Angeles, Los Angeles, CA, USA
| | - Elizabeth M Burnette
- Department of Psychology, University of California at Los Angeles, Los Angeles, CA, USA
| | - Catherine M Cahill
- Department of Psychiatry and Biobehavioral Sciences, University of California at Los Angeles, Los Angeles, CA, USA.,Shirley & Stefan Hatos Center for Neuropharmacology, University of California at Los Angeles, Los Angeles, CA, USA.,Jane & Terry Semel Institute for Neuroscience and Human Behavior, University of California at Los Angeles, Los Angeles, CA, USA
| | - Lara A Ray
- Department of Psychology, University of California at Los Angeles, Los Angeles, CA, USA.,Department of Psychiatry and Biobehavioral Sciences, University of California at Los Angeles, Los Angeles, CA, USA.,Shirley & Stefan Hatos Center for Neuropharmacology, University of California at Los Angeles, Los Angeles, CA, USA.,Jane & Terry Semel Institute for Neuroscience and Human Behavior, University of California at Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
21
|
Barney TM, Vore AS, Deak T. Acute Ethanol Challenge Differentially Regulates Expression of Growth Factors and miRNA Expression Profile of Whole Tissue of the Dorsal Hippocampus. Front Neurosci 2022; 16:884197. [PMID: 35706690 PMCID: PMC9189295 DOI: 10.3389/fnins.2022.884197] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 03/31/2022] [Indexed: 02/02/2023] Open
Abstract
Acute ethanol exposure produces rapid alterations in neuroimmune gene expression that are both time- and cytokine-dependent. Interestingly, adolescent rats, who often consume binge-like quantities of alcohol, displayed reduced neuroimmune responses to acute ethanol challenge. However, it is not known whether growth factors, a related group of signaling factors, respond to ethanol similarly in adults and adolescents. Therefore, Experiment 1 aimed to assess the growth factor response to ethanol in both adolescents and adults. To test this, adolescent (P29-P34) and adult (P70-P80) Sprague Dawley rats of both sexes were injected with either ethanol (3.5 g/kg) or saline, and brains were harvested 3 h post-injection for assessment of growth factor, cytokine, or miRNA expression. As expected, acute ethanol challenge significantly increased IL-6 and IκBα expression in the hippocampus and amygdala, replicating our prior findings. Acute ethanol significantly decreased BDNF and increased FGF2 regardless of age condition. PDGF was unresponsive to ethanol, but showed heightened expression among adolescent males. Because recent work has focused on the PDE4 inhibitor ibudilast for treatment in alcohol use disorder, Experiment 2 tested whether ibudilast would alter ethanol-evoked gene expression changes in cytokines and growth factors in the CNS. Ibudilast (9.0 mg/kg s.c.) administration 1 h prior to ethanol had no effect on ethanol-induced changes in cytokine or growth factor changes in the hippocampus or amygdala. To further explore molecular alterations evoked by acute ethanol challenge in the adult rat hippocampus, Experiment 3 tested whether acute ethanol would change the miRNA expression profile of the dorsal hippocampus using RNASeq, which revealed a rapid suppression of 12 miRNA species 3 h after acute ethanol challenge. Of the miRNA affected by ethanol, the majority were related to inflammation or cell survival and proliferation factors, including FGF2, MAPK, NFκB, and VEGF. Overall, these findings suggest that ethanol-induced, rapid alterations in neuroimmune gene expression were (i) muted among adolescents; (ii) independent of PDE4 signaling; and (iii) accompanied by changes in several growth factors (increased FGF2, decreased BDNF). In addition, ethanol decreased expression of multiple miRNA species, suggesting a dynamic molecular profile of changes in the hippocampus within a few short hours after acute ethanol challenge. Together, these findings may provide important insight into the molecular consequences of heavy drinking in humans.
Collapse
|
22
|
Abstract
Alcohol use disorder (AUD) is a highly prevalent but severely under-treated disorder, with only three widely-approved pharmacotherapies. Given that AUD is a very heterogeneous disorder, it is unlikely that one single medication will be effective for all individuals with an AUD. As such, there is a need to develop new, more effective, and diverse pharmacological treatment options for AUD with the hopes of increasing utilization and improving care. In this qualitative literature review, we discuss the efficacy, mechanism of action, and tolerability of approved, repurposed, and novel pharmacotherapies for the treatment of AUD with a clinical perspective. Pharmacotherapies discussed include: disulfiram, acamprosate, naltrexone, nalmefene, topiramate, gabapentin, varenicline, baclofen, sodium oxybate, aripiprazole, ondansetron, mifepristone, ibudilast, suvorexant, prazosin, doxazosin, N-acetylcysteine, GET73, ASP8062, ABT-436, PF-5190457, and cannabidiol. Overall, many repurposed and novel agents discussed in this review demonstrate clinical effectiveness and promise for the future of AUD treatment. Importantly, these medications also offer potential improvements towards the advancement of precision medicine and personalized treatment for the heterogeneous AUD population. However, there remains a great need to improve access to treatment, increase the menu of approved pharmacological treatments, and de-stigmatize and increase treatment-seeking for AUD.
Collapse
|