1
|
Adermark L, Cadeddu D, Lucente E, Danielsson K, Söderpalm B, Ericson M. Morphine self-administration decreases intrinsic excitability of accumbal medium spiny neurons and suppresses the innate immune system in male Wistar rats. Neurochem Int 2025; 186:105965. [PMID: 40127781 DOI: 10.1016/j.neuint.2025.105965] [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: 01/06/2025] [Revised: 03/18/2025] [Accepted: 03/21/2025] [Indexed: 03/26/2025]
Abstract
INTRODUCTION Morphine alleviates severe pain but is addictive and associated with weakened immune system. Interestingly, the immunosuppressive effects have been linked to central circuits including the nucleus accumbens shell (NAc), suggesting that there might be a direct link between reward processing in the NAc and weakened immune system. The overall aim with this study was to assess the impact displayed by morphine self-administration on neuroplasticity in the NAc shell and circulating white blood cells. METHODS Wistar rats received morphine injections over ten days, and locomotor activity was monitored. Next, morphine self-administration, and relapse drinking after forced abstinence, were assessed. Lastly, electrophysiological recordings were conducted in the NAc ex vivo to define neurophysiological adaptations, and hematological analysis were conducted in parallel. RESULTS While ten days of morphine injections were not sufficient to affect morphine self-administration, behavioral sensitization to the locomotor stimulatory properties of morphine was observed and further correlated with the amount of morphine consumed following forced abstinence. Electrophysiological slice recordings demonstrated no effect on excitatory neurotransmission, but the intrinsic excitability of NAc neurons was significantly depressed compared to water drinking controls. In addition, hematological analysis demonstrated a significant decline in the number of white blood cells, especially monocytes and neutrophils, while erythrocytes were not affected. The amount of circulating white blood cells further correlated with morphine intake, but not with neurophysiological parameters. CONCLUSION The data presented here demonstrates that morphine self-administration produces accumbal neuroplasticity and biological transformations that could contribute to the addictive and immunosuppressive properties of morphine.
Collapse
Affiliation(s)
- Louise Adermark
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Davide Cadeddu
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Erika Lucente
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Klara Danielsson
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Bo Söderpalm
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Beroendekliniken, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Mia Ericson
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
2
|
McDonough J, Singhal NK, Getsy PM, Knies K, Knauss ZT, Mueller D, Bates JN, Damron DS, Lewis SJ. The epigenetic signatures of opioid addiction and physical dependence are prevented by D-cysteine ethyl ester and betaine. Front Pharmacol 2024; 15:1416701. [PMID: 39281282 PMCID: PMC11392886 DOI: 10.3389/fphar.2024.1416701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 07/29/2024] [Indexed: 09/18/2024] Open
Abstract
We have reported that D,L-thiol esters, including D-cysteine ethyl ester (D-CYSee), are effective at overcoming opioid-induced respiratory depression (OIRD) in rats. Our on-going studies reveal that co-injections of D-CYSee with multi-day morphine injections markedly diminish spontaneous withdrawal that usually occurs after cessation of multiple injections of morphine in rats. Chronically administered opioids are known (1) to alter cellular redox status, thus inducing an oxidative state, and (2) for an overall decrease in DNA methylation, therefore resulting in the transcriptional activation of previously silenced long interspersed elements (LINE-1) retrotransposon genes. The first objective of the present study was to determine whether D-CYSee and the one carbon metabolism with the methyl donor, betaine, would maintain redox control and normal DNA methylation levels in human neuroblastoma cell cultures (SH-SY5Y) under overnight challenge with morphine (100 nM). The second objective was to determine whether D-CYSee and/or betaine could diminish the degree of physical dependence to morphine in male Sprague Dawley rats. Our data showed that overnight treatment with morphine reduced cellular GSH levels, induced mitochondrial damage, decreased global DNA methylation, and increased LINE-1 mRNA expression. These adverse effects by morphine, which diminished the reducing capacity and compromised the maintenance of the membrane potential of SH-SY5Y cells, was prevented by concurrent application of D-CYSee (100 µM) or betaine (300 µM). Furthermore, our data demonstrated that co-injections of D-CYSee (250 μmol/kg, IV) and to a lesser extent, betaine (250 μmol/kg, IV), markedly diminished the development of physical dependence induced by multi-day morphine injections (escalating daily doses of 10-30 mg/kg, IV), as assessed by the lesser number of withdrawal phenomena elicited by the injection of the opioid receptor antagonist, naloxone (1.5 mg/kg, IV). These findings provide evidence that D-CYSee and betaine prevent the appearance of redox alterations and epigenetic signatures commonly seen in neural cells involved in opioid physical dependence/addiction, and lessen development of physical dependence to morphine.
Collapse
Affiliation(s)
- Jennifer McDonough
- Department of Biological Sciences, Kent State University, Kent, OH, United States
| | - Naveen K Singhal
- Department of Biological Sciences, Kent State University, Kent, OH, United States
| | - Paulina M Getsy
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, United States
| | - Katherine Knies
- Department of Biological Sciences, Kent State University, Kent, OH, United States
| | - Zackery T Knauss
- Department of Biological Sciences, Kent State University, Kent, OH, United States
| | - Devin Mueller
- Department of Biological Sciences, Kent State University, Kent, OH, United States
| | - James N Bates
- Department of Anesthesia, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
| | - Derek S Damron
- Department of Biological Sciences, Kent State University, Kent, OH, United States
| | - Stephen J Lewis
- Department of Biological Sciences, Kent State University, Kent, OH, United States
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, United States
- Functional Electrical Stimulation Center, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
3
|
Sethi MK, Maccioni R, Hogan JD, Kawamura T, Repunte-Canonigo V, Chen J, Zaia J, Sanna PP. Comprehensive Glycomic and Proteomic Analysis of Mouse Striatum and Lateral Hypothalamus Following Repeated Exposures to Cocaine or Methamphetamine. Mol Cell Proteomics 2024; 23:100803. [PMID: 38880242 PMCID: PMC11324981 DOI: 10.1016/j.mcpro.2024.100803] [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: 11/01/2023] [Revised: 05/23/2024] [Accepted: 06/13/2024] [Indexed: 06/18/2024] Open
Abstract
Substance use disorder is a major concern, with few therapeutic options. Heparan sulfate (HS) and chondroitin sulfate (CS) interact with a plethora of growth factors and their receptors and have profound effects on cellular signaling. Thus, targeting these dynamic interactions might represent a potential novel therapeutic modality. In the present study, we performed mass spectrometry-based glycomic and proteomic analysis to understand the effects of cocaine and methamphetamine (METH) on HS, CS, and the proteome of two brain regions critically involved in drug addiction: the lateral hypothalamus and the striatum. We observed that cocaine and METH significantly alter HS and CS abundances as well as sulfate contents and composition. In particular, repeated METH or cocaine treatments reduced CS 4-O-sulfation and increased CS 6-O-sulfation. Since C4S and C6S exercise differential effects on axon growth, regeneration, and plasticity, these changes likely contribute to drug-induced neural plasticity in these brain regions. Notably, we observed that restoring these alterations by increasing CS 4-0 levels in the lateral hypothalamus by adeno-associated virus delivery of an shRNA to arylsulfatase B (N-acetylgalactosamine-4-sulfatase) ameliorated anxiety and prevented the expression of preference for cocaine in a novelty induced conditioned place preference test during cocaine withdrawal. Finally, proteomics analyses revealed a number of aberrant proteins in METH- and cocaine-treated versus saline-treated mice, including myelin proteolipid protein, calcium/calmodulin-dependent protein kinase type II subunit alpha, synapsin-2, tenascin-R, calnexin, annexin A7, hepatoma-derived growth factor, neurocan, and CSPG5, and oxidative phosphorylation among the top perturbed pathway. Taken together, these data support the role of HS, CS, and associated proteins in stimulants abuse and suggest that manipulation of HSPGs can represent a novel therapeutic strategy.
Collapse
Affiliation(s)
- Manveen K Sethi
- Center for Biomedical Mass Spectrometry, Department of Biochemistry and Cell Biology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Riccardo Maccioni
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
| | - John D Hogan
- Bioinformatics Program, Boston University, Boston, Massachusetts, USA
| | - Tomoya Kawamura
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
| | - Vez Repunte-Canonigo
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
| | - Jihuan Chen
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
| | - Joseph Zaia
- Center for Biomedical Mass Spectrometry, Department of Biochemistry and Cell Biology, Boston University School of Medicine, Boston, Massachusetts, USA; Bioinformatics Program, Boston University, Boston, Massachusetts, USA.
| | - Pietro Paolo Sanna
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA.
| |
Collapse
|
4
|
Jang YO, Roh Y, Shin W, Jo S, Koo B, Liu H, Kim MG, Lee HJ, Qiao Z, Lee EY, Lee M, Lee J, Lee EJ, Shin Y. Transferrin-conjugated magnetic nanoparticles for the isolation of brain-derived blood exosomal MicroRNAs: A novel approach for Parkinson's disease diagnosis. Anal Chim Acta 2024; 1306:342623. [PMID: 38692796 DOI: 10.1016/j.aca.2024.342623] [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: 02/04/2024] [Revised: 03/28/2024] [Accepted: 04/17/2024] [Indexed: 05/03/2024]
Abstract
BACKGROUND Brain-derived exosomes circulate in the bloodstream and other bodily fluids, serving as potential indicators of neurological disease progression. These exosomes present a promising avenue for the early and precise diagnosis of neurodegenerative conditions. Notably, miRNAs found in plasma extracellular vesicles (EVs) offer distinct diagnostic benefits due to their stability, abundance, and resistance to breakdown. RESULTS In this study, we introduce a method using transferrin conjugated magnetic nanoparticles (TMNs) to isolate these exosomes from the plasma of patients with neurological disorders. This TMNs technique is both quick (<35 min) and cost-effective, requiring no high-priced ingredients or elaborate equipment for EV extraction. Our method successfully isolated EVs from 33 human plasma samples, including those from patients with Parkinson's disease (PD), Multiple Sclerosis (MS), and Dementia. Using quantitative polymerase chain reaction (PCR) analysis, we evaluated the potential of 8 exosomal miRNA profiles as biomarker candidates. Six exosomal miRNA biomarkers (miR-195-5p, miR-495-3p, miR-23b-3P, miR-30c-2-3p, miR-323a-3p, and miR-27a-3p) were consistently linked with all stages of PD. SIGNIFICANCE The TMNs method provides a practical, cost-efficient way to isolate EVs from biological samples, paving the way for non-invasive neurological diagnoses. Furthermore, the identified miRNA biomarkers in these exosomes may emerge as innovative tools for precise diagnosis in neurological disorders including PD.
Collapse
Affiliation(s)
- Yoon Ok Jang
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Yeonjeong Roh
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Wangyong Shin
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Sungyang Jo
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Bonhan Koo
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Huifang Liu
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Myoung Gyu Kim
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Hyo Joo Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Zhen Qiao
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Eun Yeong Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Minju Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Joonseok Lee
- Department of Chemistry, Hanyang University, Seoul, 04763, Republic of Korea.
| | - Eun-Jae Lee
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea.
| | - Yong Shin
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea.
| |
Collapse
|
5
|
Mitra A, Deats SP, Dickson PE, Zhu J, Gardin J, Nieman BJ, Henkelman RM, Tsai NP, Chesler EJ, Zhang ZW, Kumar V. Tmod2 Is a Regulator of Cocaine Responses through Control of Striatal and Cortical Excitability and Drug-Induced Plasticity. J Neurosci 2024; 44:e1389232024. [PMID: 38508714 PMCID: PMC11063827 DOI: 10.1523/jneurosci.1389-23.2024] [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: 06/27/2023] [Revised: 02/12/2024] [Accepted: 02/24/2024] [Indexed: 03/22/2024] Open
Abstract
Drugs of abuse induce neuroadaptations, including synaptic plasticity, that are critical for transition to addiction, and genes and pathways that regulate these neuroadaptations are potential therapeutic targets. Tropomodulin 2 (Tmod2) is an actin-regulating gene that plays an important role in synapse maturation and dendritic arborization and has been implicated in substance abuse and intellectual disability in humans. Here, we mine the KOMP2 data and find that Tmod2 knock-out mice show emotionality phenotypes that are predictive of addiction vulnerability. Detailed addiction phenotyping shows that Tmod2 deletion does not affect the acute locomotor response to cocaine administration. However, sensitized locomotor responses are highly attenuated in these knock-outs, indicating perturbed drug-induced plasticity. In addition, Tmod2 mutant animals do not self-administer cocaine indicating lack of hedonic responses to cocaine. Whole-brain MR imaging shows differences in brain volume across multiple regions, although transcriptomic experiments did not reveal perturbations in gene coexpression networks. Detailed electrophysiological characterization of Tmod2 KO neurons showed increased spontaneous firing rate of early postnatal and adult cortical and striatal neurons. Cocaine-induced synaptic plasticity that is critical for sensitization is either missing or reciprocal in Tmod2 KO nucleus accumbens shell medium spiny neurons, providing a mechanistic explanation of the cocaine response phenotypes. Combined, these data, collected from both males and females, provide compelling evidence that Tmod2 is a major regulator of plasticity in the mesolimbic system and regulates the reinforcing and addictive properties of cocaine.
Collapse
Affiliation(s)
| | | | | | - Jiuhe Zhu
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801
| | | | - Brian J Nieman
- Mouse Imaging Centre and Translational Medicine, Hospital for Sick Children; Ontario Institute for Cancer Research; Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5T 3H7, Canada
| | - R Mark Henkelman
- Mouse Imaging Centre and Translational Medicine, Hospital for Sick Children; Ontario Institute for Cancer Research; Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5T 3H7, Canada
| | - Nien-Pei Tsai
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801
| | | | | | - Vivek Kumar
- The Jackson Laboratory, Bar Harbor, Maine 04609
| |
Collapse
|
6
|
Falconnier C, Caparros-Roissard A, Decraene C, Lutz PE. Functional genomic mechanisms of opioid action and opioid use disorder: a systematic review of animal models and human studies. Mol Psychiatry 2023; 28:4568-4584. [PMID: 37723284 PMCID: PMC10914629 DOI: 10.1038/s41380-023-02238-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 08/17/2023] [Accepted: 08/24/2023] [Indexed: 09/20/2023]
Abstract
In the past two decades, over-prescription of opioids for pain management has driven a steep increase in opioid use disorder (OUD) and death by overdose, exerting a dramatic toll on western countries. OUD is a chronic relapsing disease associated with a lifetime struggle to control drug consumption, suggesting that opioids trigger long-lasting brain adaptations, notably through functional genomic and epigenomic mechanisms. Current understanding of these processes, however, remain scarce, and have not been previously reviewed systematically. To do so, the goal of the present work was to synthesize current knowledge on genome-wide transcriptomic and epigenetic mechanisms of opioid action, in primate and rodent species. Using a prospectively registered methodology, comprehensive literature searches were completed in PubMed, Embase, and Web of Science. Of the 2709 articles identified, 73 met our inclusion criteria and were considered for qualitative analysis. Focusing on the 5 most studied nervous system structures (nucleus accumbens, frontal cortex, whole striatum, dorsal striatum, spinal cord; 44 articles), we also conducted a quantitative analysis of differentially expressed genes, in an effort to identify a putative core transcriptional signature of opioids. Only one gene, Cdkn1a, was consistently identified in eleven studies, and globally, our results unveil surprisingly low consistency across published work, even when considering most recent single-cell approaches. Analysis of sources of variability detected significant contributions from species, brain structure, duration of opioid exposure, strain, time-point of analysis, and batch effects, but not type of opioid. To go beyond those limitations, we leveraged threshold-free methods to illustrate how genome-wide comparisons may generate new findings and hypotheses. Finally, we discuss current methodological development in the field, and their implication for future research and, ultimately, better care.
Collapse
Affiliation(s)
- Camille Falconnier
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives UPR 3212, 67000, Strasbourg, France
| | - Alba Caparros-Roissard
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives UPR 3212, 67000, Strasbourg, France
| | - Charles Decraene
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives UPR 3212, 67000, Strasbourg, France
- Centre National de la Recherche Scientifique, Université de Strasbourg, Laboratoire de Neurosciences Cognitives et Adaptatives UMR 7364, 67000, Strasbourg, France
| | - Pierre-Eric Lutz
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives UPR 3212, 67000, Strasbourg, France.
- Douglas Mental Health University Institute, Montreal, QC, Canada.
| |
Collapse
|
7
|
Vilca SJ, Margetts AV, Pollock TA, Tuesta LM. Transcriptional and epigenetic regulation of microglia in substance use disorders. Mol Cell Neurosci 2023; 125:103838. [PMID: 36893849 PMCID: PMC10247513 DOI: 10.1016/j.mcn.2023.103838] [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: 11/16/2022] [Revised: 02/17/2023] [Accepted: 03/02/2023] [Indexed: 03/09/2023] Open
Abstract
Microglia are widely known for their role in immune surveillance and for their ability to refine neurocircuitry during development, but a growing body of evidence suggests that microglia may also play a complementary role to neurons in regulating the behavioral aspects of substance use disorders. While many of these efforts have focused on changes in microglial gene expression associated with drug-taking, epigenetic regulation of these changes has yet to be fully understood. This review provides recent evidence supporting the role of microglia in various aspects of substance use disorder, with particular focus on changes to the microglial transcriptome and the potential epigenetic mechanisms driving these changes. Further, this review discusses the latest technical advances in low-input chromatin profiling and highlights the current challenges for studying these novel molecular mechanisms in microglia.
Collapse
Affiliation(s)
- Samara J Vilca
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, United States of America; Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136, United States of America
| | - Alexander V Margetts
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, United States of America; Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136, United States of America; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, United States of America
| | - Tate A Pollock
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, United States of America; Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136, United States of America
| | - Luis M Tuesta
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, United States of America; Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136, United States of America; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, United States of America.
| |
Collapse
|
8
|
King'uyu DN, Edgar EL, Figueroa C, Kirkland JM, Kopec AM. Morphine exposure during adolescence induces enduring social changes dependent on adolescent stage of exposure, sex, and social test. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.21.537856. [PMID: 37131669 PMCID: PMC10153224 DOI: 10.1101/2023.04.21.537856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Drug exposure during adolescence, when the 'reward' circuitry of the brain is developing, can permanently impact reward-related behavior. Epidemiological studies show that opioid treatment during adolescence, such as pain management for a dental procedure or surgery, increases the incidence of psychiatric illness including substance use disorders. Moreover, the opioid epidemic currently in the United States is affecting younger individuals raising the impetus to understand the pathogenesis of the negative effects of opioids. One reward-related behavior that develops during adolescence is social behavior. We previously demonstrated that social development occurs in rats during sex-specific adolescent periods: early to mid-adolescence in males (postnatal day (P)30-40) and pre-early adolescence in females (P20-30). We thus hypothesized that morphine exposure during the female critical period would result in adult sociability deficits in females, but not males, and morphine administered during the male critical period would result in adult sociability deficits in males, but not females. We found that morphine exposure during the female critical period primarily resulted in deficits in sociability in females, while morphine exposure during the male critical period primarily resulted in deficits in sociability primarily in males. However, depending on the test performed and the social parameter measured, social alterations could be found in both sexes that received morphine exposure at either adolescent stage. These data indicate that when drug exposure occurs during adolescence, and how the endpoint data are measured, will play a large role in determining the effects of drug exposures on social development.
Collapse
Affiliation(s)
- David N King'uyu
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College
| | - Erin L Edgar
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College
| | - Christopher Figueroa
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College
| | - J M Kirkland
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College
| | - Ashley M Kopec
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College
| |
Collapse
|
9
|
Cabana-Domínguez J, Antón-Galindo E, Fernàndez-Castillo N, Singgih EL, O'Leary A, Norton WH, Strekalova T, Schenck A, Reif A, Lesch KP, Slattery D, Cormand B. The translational genetics of ADHD and related phenotypes in model organisms. Neurosci Biobehav Rev 2023; 144:104949. [PMID: 36368527 DOI: 10.1016/j.neubiorev.2022.104949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 11/02/2022] [Accepted: 11/05/2022] [Indexed: 11/10/2022]
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a highly prevalent neurodevelopmental disorder resulting from the interaction between genetic and environmental risk factors. It is well known that ADHD co-occurs frequently with other psychiatric disorders due, in part, to shared genetics factors. Although many studies have contributed to delineate the genetic landscape of psychiatric disorders, their specific molecular underpinnings are still not fully understood. The use of animal models can help us to understand the role of specific genes and environmental stimuli-induced epigenetic modifications in the pathogenesis of ADHD and its comorbidities. The aim of this review is to provide an overview on the functional work performed in rodents, zebrafish and fruit fly and highlight the generated insights into the biology of ADHD, with a special focus on genetics and epigenetics. We also describe the behavioral tests that are available to study ADHD-relevant phenotypes and comorbid traits in these models. Furthermore, we have searched for new models to study ADHD and its comorbidities, which can be useful to test potential pharmacological treatments.
Collapse
Affiliation(s)
- Judit Cabana-Domínguez
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Spain; Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Catalonia, Spain; Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Catalonia, Spain.
| | - Ester Antón-Galindo
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Spain; Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Catalonia, Spain; Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Catalonia, Spain
| | - Noèlia Fernàndez-Castillo
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Spain; Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Catalonia, Spain; Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Catalonia, Spain
| | - Euginia L Singgih
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Aet O'Leary
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University, Frankfurt, Germany; Division of Neuropsychopharmacology, Department of Psychology, University of Tartu, Tartu, Estonia
| | - William Hg Norton
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Tatyana Strekalova
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany, and Department of Neuropsychology and Psychiatry, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, the Netherlands
| | - Annette Schenck
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Andreas Reif
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University, Frankfurt, Germany
| | - Klaus-Peter Lesch
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany, and Department of Neuropsychology and Psychiatry, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, the Netherlands
| | - David Slattery
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University, Frankfurt, Germany
| | - Bru Cormand
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Spain; Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Catalonia, Spain; Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Catalonia, Spain.
| |
Collapse
|
10
|
Seyednejad SA, Sartor GC. Noncoding RNA therapeutics for substance use disorder. ADVANCES IN DRUG AND ALCOHOL RESEARCH 2022; 2:10807. [PMID: 36601439 PMCID: PMC9808746 DOI: 10.3389/adar.2022.10807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Although noncoding RNAs (ncRNAs) have been shown to regulate maladaptive neuroadaptations that drive compulsive drug use, ncRNA-targeting therapeutics for substance use disorder (SUD) have yet to be clinically tested. Recent advances in RNA-based drugs have improved many therapeutic issues related to immune response, specificity, and delivery, leading to multiple successful clinical trials for other diseases. As the need for safe and effective treatments for SUD continues to grow, novel nucleic acid-based therapeutics represent an appealing approach to target ncRNA mechanisms in SUD. Here, we review ncRNA processes implicated in SUD, discuss recent therapeutic approaches for targeting ncRNAs, and highlight potential opportunities and challenges of ncRNA-targeting therapeutics for SUD.
Collapse
Affiliation(s)
- Seyed Afshin Seyednejad
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, United States
- Connecticut Institute for the Brain and Cognitive Sciences (CT IBACS), Storrs, CT, United States
| | - Gregory C. Sartor
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, United States
- Connecticut Institute for the Brain and Cognitive Sciences (CT IBACS), Storrs, CT, United States
| |
Collapse
|
11
|
Zhao Y, Qin F, Han S, Li S, Zhao Y, Wang H, Tian J, Cen X. MicroRNAs in drug addiction: Current status and future perspectives. Pharmacol Ther 2022; 236:108215. [DOI: 10.1016/j.pharmthera.2022.108215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/11/2022] [Accepted: 05/16/2022] [Indexed: 12/21/2022]
|
12
|
Gillespie A, Mayberry HL, Wimmer ME, Sillivan SE. microRNA expression levels in the nucleus accumbens correlate with morphine-taking but not morphine-seeking behaviour in male rats. Eur J Neurosci 2022; 55:1742-1755. [PMID: 35320877 PMCID: PMC9314918 DOI: 10.1111/ejn.15650] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 03/04/2022] [Accepted: 03/15/2022] [Indexed: 12/01/2022]
Abstract
A powerful motivation to seek opioids remains after drug cessation and intensifies during extended periods of abstinence. Unfortunately, biomarkers associated with continued drug seeking have not been described. Moreover, previous studies have focused on the effects of early abstinence with little exploration into the long-term drug-induced mechanisms that occur after extended abstinence. Here we demonstrated that 30 days (D) of forced abstinence results in a time-dependent increase in morphine seeking in a rat model of morphine self-administration (SA). We measured expression of known drug-responsive microRNAs (miRNAs) in the nucleus accumbens, an area critical for reward-related plasticity, during early or late abstinence in animals that underwent either a cue-induced relapse test or no relapse test. miRNAs are small noncoding RNAs that represent suitable biomarker candidates due to their long-lasting nature. mir-32-5p levels during early abstinence negatively correlated with active lever pressing in both cue-exposed and cue-naïve animals. mir-1298-5p positively correlated with drug SA history after a relapse test during late abstinence. When animals underwent acute abstinence with no relapse test, mir-1298-5p correlated with drug infusions and active lever pressing during SA. In late abstinence with no relapse test, mir-137-3p negatively correlated with drug infusions. Regulation of mir-32-5p target genes and significant correlation of target gene mRNA with mir-32-5p was observed after abstinence. These results indicate that lasting regulation of miRNA expression is associated with drug intake following morphine SA. In addition, we conclude that the miRNA profile undergoes regulation from early to late abstinence and miRNA expression may indicate past drug history.
Collapse
Affiliation(s)
- Aria Gillespie
- Center for Substance Abuse ResearchTemple UniversityPhiladelphiaPennsylvaniaUSA,Department of Neural SciencesTemple UniversityPhiladelphiaPAUSA
| | - Hannah L. Mayberry
- Department of Psychology and Neuroscience Program, College of Liberal ArtsTemple UniversityPhiladelphiaPennsylvaniaUSA
| | - Mathieu E. Wimmer
- Department of Psychology and Neuroscience Program, College of Liberal ArtsTemple UniversityPhiladelphiaPennsylvaniaUSA
| | - Stephanie E. Sillivan
- Center for Substance Abuse ResearchTemple UniversityPhiladelphiaPennsylvaniaUSA,Department of Neural SciencesTemple UniversityPhiladelphiaPAUSA
| |
Collapse
|
13
|
Li X, Xie B, Lu Y, Yang H, Wang J, Yu F, Zhang L, Cong B, Wen D, Ma C. Transcriptomic Analysis of Long Non-coding RNA-MicroRNA-mRNA Interactions in the Nucleus Accumbens Related to Morphine Addiction in Mice. Front Psychiatry 2022; 13:915398. [PMID: 35722589 PMCID: PMC9201067 DOI: 10.3389/fpsyt.2022.915398] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 04/29/2022] [Indexed: 11/13/2022] Open
Abstract
Recent research suggest that some non-coding RNAs (ncRNAs) are important regulators of chromatin dynamics and gene expression in nervous system development and neurological diseases. Nevertheless, the molecular mechanisms of long non-coding RNAs (lncRNAs), acting as competing endogenous RNAs (ceRNAs), underlying morphine addiction are still unknown. In this research, RNA sequencing (RNA-seq) was used to examine the expression profiles of lncRNAs, miRNAs and mRNAs on the nucleus accumbens (NAc) tissues of mice trained with morphine or saline conditioned place preference (CPP), with differential expression of 31 lncRNAs, 393 miRNAs, and 371 mRNAs found. A ceRNA network was established for reciprocal interactions for 9 differentially expressed lncRNAs (DElncRNAs), 10 differentially expressed miRNAs (DEmiRNAs) and 12 differentially expressed mRNAs (DEmRNAs) based on predicted miRNAs shared by lncRNAs and mRNAs. KEGG pathway enrichment analyses were conducted to explore the potential functions of DEmRNAs interacting with lncRNAs in the ceRNA network. These DEmRNAs were enriched in synaptic plasticity-related pathways, including pyrimidine metabolism, ECM-receptor interaction, and focal adhesion. The correlation between the relative expression of lncRNAs, miRNAs and mRNAs was analyzed to further validate predicted ceRNA networks, and the Lnc15qD3-miR-139-3p-Lrp2 ceRNA regulatory interaction was determined. These results suggest that the comprehensive network represents a new insight into the lncRNA-mediated ceRNA regulatory mechanisms underlying morphine addiction and provide new potential diagnostic and prognostic biomarkers for morphine addiction.
Collapse
Affiliation(s)
- Xiaojie Li
- Hebei Key Laboratory of Forensic Medicine, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Collaborative Innovation Center of Forensic Medical Molecular Identification, College of Forensic Medicine, Chinese Academy of Medical Sciences, Hebei Medical University, Shijiazhuang, China
| | - Bing Xie
- Hebei Key Laboratory of Forensic Medicine, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Collaborative Innovation Center of Forensic Medical Molecular Identification, College of Forensic Medicine, Chinese Academy of Medical Sciences, Hebei Medical University, Shijiazhuang, China
| | - Yun Lu
- Hebei Key Laboratory of Forensic Medicine, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Collaborative Innovation Center of Forensic Medical Molecular Identification, College of Forensic Medicine, Chinese Academy of Medical Sciences, Hebei Medical University, Shijiazhuang, China
| | - Hongyu Yang
- Hebei Key Laboratory of Forensic Medicine, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Collaborative Innovation Center of Forensic Medical Molecular Identification, College of Forensic Medicine, Chinese Academy of Medical Sciences, Hebei Medical University, Shijiazhuang, China
| | - Jian Wang
- Hebei Key Laboratory of Forensic Medicine, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Collaborative Innovation Center of Forensic Medical Molecular Identification, College of Forensic Medicine, Chinese Academy of Medical Sciences, Hebei Medical University, Shijiazhuang, China
| | - Feng Yu
- Hebei Key Laboratory of Forensic Medicine, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Collaborative Innovation Center of Forensic Medical Molecular Identification, College of Forensic Medicine, Chinese Academy of Medical Sciences, Hebei Medical University, Shijiazhuang, China
| | - Ludi Zhang
- Hebei Key Laboratory of Forensic Medicine, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Collaborative Innovation Center of Forensic Medical Molecular Identification, College of Forensic Medicine, Chinese Academy of Medical Sciences, Hebei Medical University, Shijiazhuang, China
| | - Bin Cong
- Hebei Key Laboratory of Forensic Medicine, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Collaborative Innovation Center of Forensic Medical Molecular Identification, College of Forensic Medicine, Chinese Academy of Medical Sciences, Hebei Medical University, Shijiazhuang, China
| | - Di Wen
- Hebei Key Laboratory of Forensic Medicine, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Collaborative Innovation Center of Forensic Medical Molecular Identification, College of Forensic Medicine, Chinese Academy of Medical Sciences, Hebei Medical University, Shijiazhuang, China
| | - Chunling Ma
- Hebei Key Laboratory of Forensic Medicine, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Collaborative Innovation Center of Forensic Medical Molecular Identification, College of Forensic Medicine, Chinese Academy of Medical Sciences, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
14
|
Harp SJ, Martini M, Rosenow W, Mesner LD, Johnson H, Farber CR, Rissman EF. Fentanyl-induced acute and conditioned behaviors in two inbred mouse lines: Potential role for Glyoxalase. Physiol Behav 2022; 243:113630. [PMID: 34710466 PMCID: PMC8713069 DOI: 10.1016/j.physbeh.2021.113630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/19/2021] [Accepted: 10/21/2021] [Indexed: 01/03/2023]
Abstract
An increase in opioid-overdose deaths was evident before the COVID-19 pandemic, and has escalated since its onset. Fentanyl, a highly potent synthetic opioid, is the primary driver of these recent trends. The current study used two inbred mouse strains, C57BL/6 J and A/J, to investigate the genetics of behavioral responses to fentanyl. Mice were tested for conditioned place preference and fentanyl-induced locomotor activity. C57BL/6J mice formed a conditioned place preference to fentanyl injections and fentanyl increased their activity. Neither effect was noted in A/J mice. We conducted RNA-sequencing on the nucleus accumbens of mice used for fentanyl-induced locomotor activity. Surprisingly, we noted few differentially expressed genes using treatment as the main factor. However many genes differed between strains. We validated differences in two genes: suppressor APC domain containing 1 (Sapcd1) and Glyoxalase 1 (Glo1), with quantitative PCR on RNA from the nucleus accumbens and prefrontal cortex (). In both regions A/J mice had significantly higher expression of both genes than did C57BL/6 J. In prefrontal cortex, fentanyl treatment decreased Glo1 mRNA. Glyoxalase 1 catalyzes the detoxification of reactive alpha-oxoaldehydes such as glyoxal and methylglyoxal, is associated with anxiety and activity levels, and its inhibition reduces alcohol intake. We suggest that future studies assess the ability of Glo1 and related metabolites to modify opioid intake.
Collapse
Affiliation(s)
- Samuel J. Harp
- Center for Human Health and the Environment Program in Genetics, North Carolina State University, Raleigh, NC USA
| | - Mariangela Martini
- Center for Human Health and the Environment Program in Genetics, North Carolina State University, Raleigh, NC USA
| | - Will Rosenow
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA USA
| | - Larry D. Mesner
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA USA
| | - Hugh Johnson
- Center for Human Health and the Environment Program in Genetics, North Carolina State University, Raleigh, NC USA
| | - Charles R. Farber
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA USA
| | - Emilie F. Rissman
- Center for Human Health and the Environment Program in Genetics, North Carolina State University, Raleigh, NC USA,Corresponding author: Dr. E.F. Rissman, Department of Biological Sciences, Thomas Hall, North Carolina State University, Raleigh, NC 27695, Phone: (919) 515-5807, FAX: (919) 515-
| |
Collapse
|
15
|
Blum K, Steinberg B, Gondre-Lewis MC, Baron D, Modestino EJ, Badgaiyan RD, Downs BW, Bagchi D, Brewer R, McLaughlin T, Bowirrat A, Gold M. A Review of DNA Risk Alleles to Determine Epigenetic Repair of mRNA Expression to Prove Therapeutic Effectiveness in Reward Deficiency Syndrome (RDS): Embracing "Precision Behavioral Management". Psychol Res Behav Manag 2021; 14:2115-2134. [PMID: 34949945 PMCID: PMC8691196 DOI: 10.2147/prbm.s292958] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 11/22/2021] [Indexed: 11/23/2022] Open
Abstract
This is a review of research on "Precision Behavioral Management" of substance use disorder (SUD). America is experiencing a high prevalence of substance use disorder, primarily involving legal and illegal opioid use. A 3000% increase in treatment for substance abuse has occurred between 2000 and 2016. Unfortunately, present day treatment of opioid abuse involves providing replacement therapy with powerful opioids to, at best, induce harm reduction, not prophylaxis. These interventions do not enhance gene expression and restore the balance of the brain reward system's neurotransmitters. We are proposing a generalized approach called "Precision Behavioral Management". This approach includes 1) using the Genetic Addiction Risk Severity (GARS, a 10 candidate polymorphic gene panel shown to predict ASI-alcohol and drug severity) to assess early pre-disposition to substance use disorder; 2) using a validated reward deficiency syndrome (RDS) questionnaire; 3) utilization of the Comprehensive Analysis of Reported Drugs (CARD™) to assess treatment compliance and abstinence from illicit drugs during treatment, and, importantly; 4) utilization of a "Pro-dopamine regulator (KB220)" (via IV or oral [KB220Z] delivery systems) to optimize gene expression, restore the balance of the Brain Reward Cascade's neurotransmitter systems and prevent relapse by induction of dopamine homeostasis, and; 5) utilization of targeted DNA polymorphic reward genes to direct mRNA genetic expression profiling during the treatment process. Incorporation of these events can be applied to not only the under-considered African-American RDS community, but all victims of RDS, as a demonstration of a paradigm shift that uniquely provides a novel putative "standard of care" based on DNA guided precision nutrition therapy to induce "dopamine homeostasis" and rebalance neurotransmitters in the Brain Reward Cascade. We are also developing a Reward Deficiency Syndrome Diagnostic Criteria (RDSDC) to assist in potential tertiary treatment.
Collapse
Affiliation(s)
- Kenneth Blum
- Center for Psychiatry, Medicine & Primary Care, Division of Addiction Research & Education, Graduate College, Western University Health Sciences, Pomona, CA, USA
- Eötvös Loránd University, Institute of Psychology, Budapest, Hungary
- Department of Psychiatry, Wright State University Boonshoft School of Medicine and Dayton VA Medical Center, Dayton, OH (IE), USA
- Department of Psychiatry, University of Vermont, Burlington, VT, USA
- Division of Nutrigenomics, The Kenneth Blum Behavioral & Neurogenetic Institute, Austin, TX, USA
| | | | - Marjorie C Gondre-Lewis
- Developmental Neuropsychopharmacology Laboratory, Department of Anatomy, Howard University College of Medicine, Washington, DC, USA
| | - David Baron
- Center for Psychiatry, Medicine & Primary Care, Division of Addiction Research & Education, Graduate College, Western University Health Sciences, Pomona, CA, USA
| | | | - Rajendra D Badgaiyan
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, South Texas Veteran Health Care System, Audie L. Murphy Memorial VA Hospital, San Antonio, TX, USA
- Long School of Medicine, University of Texas Medical Center, San Antonio, TX, USA
| | - B William Downs
- Division of Nutrigenomics, Victory Nutrition International, Inc., Harleysville, PA, USA
| | - Debasis Bagchi
- Division of Nutrigenomics, Victory Nutrition International, Inc., Harleysville, PA, USA
| | - Raymond Brewer
- Division of Nutrigenomics, The Kenneth Blum Behavioral & Neurogenetic Institute, Austin, TX, USA
| | - Thomas McLaughlin
- Department of Psychopharmacology, Center for Psychiatric Medicine, Lawrence, MA, USA
| | - Abdalla Bowirrat
- Adelson School of Medicine & Department of Molecular Biology, Ariel University, Ariel, Israel
| | - Mark Gold
- Department of Psychiatry, Washington University, School of Medicine, St. Louis, MO, USA
| |
Collapse
|
16
|
Genomic and Personalized Medicine Approaches for Substance Use Disorders (SUDs) Looking at Genome-Wide Association Studies. Biomedicines 2021; 9:biomedicines9121799. [PMID: 34944615 PMCID: PMC8698472 DOI: 10.3390/biomedicines9121799] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/23/2021] [Accepted: 11/25/2021] [Indexed: 12/19/2022] Open
Abstract
Drug addiction, or substance use disorder (SUD), is a chronic, relapsing disorder in which compulsive drug-seeking and drug-taking behaviour persist despite serious negative consequences. Drug abuse represents a problem that deserves great attention from a social point of view, and focuses on the importance of genetic studies to help in understanding the genetic basis of addiction and its medical treatment. Despite the complexity of drug addiction disorders, and the high number of environmental variables playing a role in the onset, recurrence, and duration of the symptoms, several studies have highlighted the non-negligible role of genetics, as demonstrated by heritability and genome-wide association studies. A correlation between the relative risk of addiction to specific substances and heritability has been recently observed, suggesting that neurobiological mechanisms may be, at least in part, inherited. All these observations point towards a scenario where the core neurobiological factors of addiction, involving the reward system, impulsivity, compulsivity, stress, and anxiety response, are transmitted, and therefore, genes and mutations underlying their variation might be detected. In the last few years, the development of new and more efficient sequencing technologies has paved the way for large-scale studies in searching for genetic and epigenetic factors affecting drug addiction disorders and their treatments. These studies have been crucial to pinpoint single nucleotide polymorphisms (SNPs) in genes that affect the reaction to medical treatments. This is critically important to identify pharmacogenomic approaches for substance use disorder, such as OPRM1 SNPs and methadone required doses for maintenance treatment (MMT). Nevertheless, despite the promising results obtained by genome-wide association and pharmacogenomic studies, specific studies related to population genetics diversity are lacking, undermining the overall applicability of the preliminary findings, and thus potentially affecting the portability and the accuracy of the genetic studies. In this review, focusing on cannabis, cocaine and heroin use, we report the state-of-the-art genomics and pharmacogenomics of SUDs, and the possible future perspectives related to medical treatment response in people that ask for assistance in solving drug-related problems.
Collapse
|
17
|
Radhakrishna U, Vishweswaraiah S, Uppala LV, Szymanska M, Macknis J, Kumar S, Saleem-Rasheed F, Aydas B, Forray A, Muvvala SB, Mishra NK, Guda C, Carey DJ, Metpally RP, Crist RC, Berrettini WH, Bahado-Singh RO. Placental DNA methylation profiles in opioid-exposed pregnancies and associations with the neonatal opioid withdrawal syndrome. Genomics 2021; 113:1127-1135. [PMID: 33711455 DOI: 10.1016/j.ygeno.2021.03.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 12/29/2020] [Accepted: 03/02/2021] [Indexed: 12/11/2022]
Abstract
Opioid abuse during pregnancy can result in Neonatal Opioid Withdrawal Syndrome (NOWS). We investigated genome-wide methylation analyses of 96 placental tissue samples, including 32 prenatally opioid-exposed infants with NOWS who needed therapy (+Opioids/+NOWS), 32 prenatally opioid-exposed infants with NOWS who did not require treatment (+Opioids/-NOWS), and 32 prenatally unexposed controls (-Opioids/-NOWS, control). Statistics, bioinformatics, Artificial Intelligence (AI), including Deep Learning (DL), and Ingenuity Pathway Analyses (IPA) were performed. We identified 17 dysregulated pathways thought to be important in the pathophysiology of NOWS and reported accurate AI prediction of NOWS diagnoses. The DL had an AUC (95% CI) =0.98 (0.95-1.0) with a sensitivity and specificity of 100% for distinguishing NOWS from the +Opioids/-NOWS group and AUCs (95% CI) =1.00 (1.0-1.0) with a sensitivity and specificity of 100% for distinguishing NOWS versus control and + Opioids/-NOWS group versus controls. This study provides strong evidence of methylation dysregulation of placental tissue in NOWS development.
Collapse
Affiliation(s)
- Uppala Radhakrishna
- Department of Obstetrics and Gynecology, Oakland University William Beaumont School of Medicine, Royal Oak, MI, USA.
| | - Sangeetha Vishweswaraiah
- Department of Obstetrics and Gynecology, Oakland University William Beaumont School of Medicine, Royal Oak, MI, USA
| | - Lavanya V Uppala
- College of Information Science & Technology, University of Nebraska at Omaha, Peter Kiewit Institute, Omaha, NE, USA
| | - Marta Szymanska
- Department of Obstetrics and Gynecology, Oakland University William Beaumont School of Medicine, Royal Oak, MI, USA
| | | | - Sandeep Kumar
- Department of Pathology, Beaumont Health System, Royal Oak, MI, USA
| | - Fozia Saleem-Rasheed
- Department of Newborn Medicine, Oakland University William Beaumont School of Medicine, Royal Oak, MI, USA
| | - Buket Aydas
- Department of Healthcare Analytics, Meridian Health Plans, Detroit, MI, USA
| | - Ariadna Forray
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | | | - Nitish K Mishra
- Department of Genetics, Cell Biology & Anatomy College of Medicine, University of Nebraska Medical Center Omaha, NE, USA
| | - Chittibabu Guda
- Department of Genetics, Cell Biology & Anatomy College of Medicine, University of Nebraska Medical Center Omaha, NE, USA
| | - David J Carey
- Department of Molecular and Functional Genomics, Geisinger, Danville, PA, USA
| | - Raghu P Metpally
- Department of Molecular and Functional Genomics, Geisinger, Danville, PA, USA
| | - Richard C Crist
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Wade H Berrettini
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Geisinger Clinic, Danville, PA, USA
| | - Ray O Bahado-Singh
- Department of Obstetrics and Gynecology, Oakland University William Beaumont School of Medicine, Royal Oak, MI, USA
| |
Collapse
|
18
|
Thompson BL, Oscar-Berman M, Kaplan GB. Opioid-induced structural and functional plasticity of medium-spiny neurons in the nucleus accumbens. Neurosci Biobehav Rev 2021; 120:417-430. [PMID: 33152423 PMCID: PMC7855607 DOI: 10.1016/j.neubiorev.2020.10.015] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/16/2020] [Accepted: 10/20/2020] [Indexed: 12/21/2022]
Abstract
Opioid Use Disorder (OUD) is a chronic relapsing clinical condition with tremendous morbidity and mortality that frequently persists, despite treatment, due to an individual's underlying psychological, neurobiological, and genetic vulnerabilities. Evidence suggests that these vulnerabilities may have neurochemical, cellular, and molecular bases. Key neuroplastic events within the mesocorticolimbic system that emerge through chronic exposure to opioids may have a determinative influence on behavioral symptoms associated with OUD. In particular, structural and functional alterations in the dendritic spines of medium spiny neurons (MSNs) within the nucleus accumbens (NAc) and its dopaminergic projections from the ventral tegmental area (VTA) are believed to facilitate these behavioral sequelae. Additionally, glutamatergic neurons from the prefrontal cortex, the basolateral amygdala, the hippocampus, and the thalamus project to these same MSNs, providing an enriched target for synaptic plasticity. Here, we review literature related to neuroadaptations in NAc MSNs from dopaminergic and glutamatergic pathways in OUD. We also describe new findings related to transcriptional, epigenetic, and molecular mechanisms in MSN plasticity in the different stages of OUD.
Collapse
Affiliation(s)
- Benjamin L Thompson
- Department of Radiology & Biomedical Imaging, Yale School of Medicine, New Haven, CT, 06520, USA; Research Service, VA Boston Healthcare System, 150 South Huntington Avenue, Boston, MA 02130, USA.
| | - Marlene Oscar-Berman
- Research Service, VA Boston Healthcare System, 150 South Huntington Avenue, Boston, MA 02130, USA; Department of Anatomy & Neurobiology, Boston University School of Medicine, 72 East Concord Street, Boston, MA, 02118, USA; Department of Psychiatry, Boston University School of Medicine, 720 Harrison Avenue, Boston, MA, 02118, USA; Department of Neurology, Boston University School of Medicine, Boston University Medical Center, 80 East Concord Street, Boston, MA 02118, USA.
| | - Gary B Kaplan
- Department of Psychiatry, Boston University School of Medicine, 720 Harrison Avenue, Boston, MA, 02118, USA; Mental Health Service, VA Boston Healthcare System, 940 Belmont Street, Brockton, MA, 02301, USA; Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, 72 East Concord Street, Boston, MA, 02118, USA.
| |
Collapse
|
19
|
Ravanidis S, Bougea A, Papagiannakis N, Koros C, Simitsi AM, Pachi I, Breza M, Stefanis L, Doxakis E. Validation of differentially expressed brain-enriched microRNAs in the plasma of PD patients. Ann Clin Transl Neurol 2020; 7:1594-1607. [PMID: 32860338 PMCID: PMC7480914 DOI: 10.1002/acn3.51146] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 07/06/2020] [Indexed: 12/16/2022] Open
Abstract
Objective There is a pressing need to identify and validate, minimally invasive, molecular biomarkers that will complement current practices and increase the diagnostic accuracy in Parkinson’s disease (PD). Brain‐enriched miRNAs regulate all aspects of neuron development and function; importantly, they are secreted by neurons in amounts that can be readily detected in the plasma. Τhe aim of the present study was to validate a set of previously identified brain‐enriched miRNAs with diagnostic potential for idiopathic PD and recognize the molecular pathways affected by these deregulated miRNAs. Methods RT‐qPCR was performed in the plasma of 92 healthy controls and 108 idiopathic PD subjects. Statistical and in silico analyses were used to validate deregulated miRNAs and pathways in PD, respectively. Results miR‐22‐3p, miR‐124‐3p, miR‐136‐3p, miR‐154‐5p, and miR‐323a‐3p levels were found to be differentially expressed between healthy controls and PD patients. miR‐330‐5p, miR‐433‐3p, and miR‐495‐3p levels were overall higher in male subjects. Most of these miRNAs are clustered at Chr14q32 displaying CREB1, CEBPB, and MAZ transcription factor binding sites. Gene Ontology annotation analysis of deregulated miRNA targets revealed that “Protein modification,” “Transcription factor activity,” and “Cell death” terms were over‐represented. Kyoto Encyclopedia of Genes and Genome analysis revealed that “Long‐term depression,” “TGF‐beta signaling,” and “FoxO signaling” pathways were significantly affected. Interpretation We validated a panel of brain‐enriched miRNAs that can be used along with other measures for the detection of PD, revealed molecular pathways targeted by these deregulated miRNAs, and identified upstream transcription factors that may be directly implicated in PD pathogenesis.
Collapse
Affiliation(s)
- Stylianos Ravanidis
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, 11527, Greece
| | - Anastasia Bougea
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, 11527, Greece.,Center of Clinical Research, Biomedical Research Foundation, Academy of Athens, Athens, 11527, Greece.,First Department of Neurology, National and Kapodistrian University of Athens Medical School, Athens, 11528, Greece
| | - Nikolaos Papagiannakis
- Center of Clinical Research, Biomedical Research Foundation, Academy of Athens, Athens, 11527, Greece.,First Department of Neurology, National and Kapodistrian University of Athens Medical School, Athens, 11528, Greece
| | - Christos Koros
- First Department of Neurology, National and Kapodistrian University of Athens Medical School, Athens, 11528, Greece
| | - Athina Maria Simitsi
- First Department of Neurology, National and Kapodistrian University of Athens Medical School, Athens, 11528, Greece
| | - Ioanna Pachi
- First Department of Neurology, National and Kapodistrian University of Athens Medical School, Athens, 11528, Greece
| | - Marianthi Breza
- First Department of Neurology, National and Kapodistrian University of Athens Medical School, Athens, 11528, Greece
| | - Leonidas Stefanis
- Center of Clinical Research, Biomedical Research Foundation, Academy of Athens, Athens, 11527, Greece.,First Department of Neurology, National and Kapodistrian University of Athens Medical School, Athens, 11528, Greece
| | - Epaminondas Doxakis
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, 11527, Greece
| |
Collapse
|
20
|
Zhang H, Wang Q, Wang Q, Liu A, Qin F, Sun Q, Li Q, Gu Y, Tang Z, Lu S, Lu Z. Circular RNA expression profiling in the nucleus accumbens: Effects of electroacupuncture treatment on morphine-induced conditioned place preference. Addict Biol 2020; 25:e12794. [PMID: 31240833 DOI: 10.1111/adb.12794] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 04/16/2019] [Accepted: 05/23/2019] [Indexed: 11/30/2022]
Abstract
Electroacupuncture (EA) has been developed on the basis of traditional Chinese acupuncture. EA can suppress craving in opioid addicts and opioid-seeking responses in rodents. However, the molecular mechanism of EA on the rewarding properties of morphine and craving responses is not known. Here, we have applied a conditioned place preference paradigm in mice to measure morphine-induced rewarding effects along with EA treatment. Circular RNAs (circRNAs) can function as micro RNA (miRNA) sponges to effectively regulate gene expression levels. CircRNA profiling within the nucleus accumbens (NAc) was performed in EA-treated and sham-treated mice. Following RNAseq, data were analyzed by gene ontology (GO) and Kyoto Encyclopedia Genes and Genomes (KEGG) tools. We identified 112 significantly differentially expressed circRNAs, including 51 that were up-regulated and 61 that were down-regulated. Our bioinformatics analyses show that these differentially expressed circRNAs map into pathways that are mainly involved with renin secretion and the cGMP-PKG signaling. We further constructed a circRNA-miRNA network that predicts the potential roles of the differentially expressed circRNAs and the interaction of circRNAs with miRNAs. Our secondary sequencing and bioinformatics analysis in the NAc after EA treatment on morphine-induced CPP provides putative novel targets on molecular mechanisms involved in morphine reinforcement and possibly craving.
Collapse
Affiliation(s)
- Han Zhang
- First Clinical Medical College Nanjing University of Chinese Medicine China
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education Nanjing University of Chinese Medicine China
| | - Qian Wang
- International Education college Nanjing University of Chinese Medicine China
| | - Qisheng Wang
- First Clinical Medical College Nanjing University of Chinese Medicine China
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education Nanjing University of Chinese Medicine China
| | - Anlong Liu
- First Clinical Medical College Nanjing University of Chinese Medicine China
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education Nanjing University of Chinese Medicine China
| | - Fenfen Qin
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education Nanjing University of Chinese Medicine China
- College of Pharmacy Nanjing University of Chinese Medicine China
| | - Qinmei Sun
- First Clinical Medical College Nanjing University of Chinese Medicine China
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education Nanjing University of Chinese Medicine China
| | - Qian Li
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education Nanjing University of Chinese Medicine China
| | - Yun Gu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education Nanjing University of Chinese Medicine China
| | - Zongxiang Tang
- School of Medicine and Life Science Nanjing University of Chinese Medicine China
| | - Shengfeng Lu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education Nanjing University of Chinese Medicine China
| | - Zhigang Lu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education Nanjing University of Chinese Medicine China
- College of Pharmacy Nanjing University of Chinese Medicine China
| |
Collapse
|
21
|
Demirel G, Guzel E, Creighton CJ, Ozturk YE, Kucuk C, Asliyuksek H, Yurdun T. MDMA Abuse in Relation to MicroRNA Variation in Human Brain Ventral Tegmental Area and Nucleus Accumbens. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2020; 18:1989-1999. [PMID: 32184864 PMCID: PMC7059052 DOI: 10.22037/ijpr.2019.15097.12874] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
3,4-methylenedioxymethamphetamine (MDMA) is one of the most widespread illegal drugs, that have been used particularly by young people in the 15-34 age group. MicroRNAs (miRNAs) are endogenously synthesized, non-coding, and small RNAs that post-transcriptionally regulate their target genes' expression by inhibiting protein translation or degradation. miRNAs are increasingly implicated in drug-related gene expressions and functions. Notably, there are no reports of miRNA variation in the human brain in MDMA abuse. We here present a miRNA profiling study - the first such study, to the best of our knowledge - into the post-mortem human brains of a sample of people with MDMA abuse, along with non-drug dependent controls. The miRNA profiling of nucleus accumbens (NAc) and ventral tegmental areas (VTA) was performed by microarray analysis. Subsequently, two candidate miRNA putative biomarkers were selected according to significant regional differential expression (miR-1202 and miR-7975), using quantitative reverse-transcription PCR (qRT-PCR). We showed that the expression level of miR-7975 was significantly lower in the VTA regions of the 30 MDMA users, as compared with the 30 control samples. Another significantly deregulated miR-1202 was down-regulated in the NAc regions of 30 MDMA samples in comparison to the control samples. Alteration of these miRNAs can potentially serve as novel biomarkers for MDMA abuse, and warrant further research in independent and larger samples of patients.
Collapse
Affiliation(s)
- Goksun Demirel
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Biruni University, Istanbul, Turkey.,Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Çukurova University, Adana, Turkey.,G D and E G. These authors contributed equally to this work
| | - Esra Guzel
- Department of Molecular Biology and Genetics, Institute of Health Sciences, University of Health Sciences, Istanbul, Turkey.,G D and E G. These authors contributed equally to this work
| | - Chad J Creighton
- Department of Medicine, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Cancer Center Division of Biostatistics, Houston, Texas
| | - Yeter Erol Ozturk
- Chemistry Department, Council of Forensic Medicine, Istanbul, Turkey
| | | | | | - Türkan Yurdun
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Marmara University, Istanbul, Turkey
| |
Collapse
|
22
|
Lu Z, Xu J, Wang Q, Pan YX. Morphine modulates the expression of mu-opioid receptor exon 5-associated full-length C-terminal splice variants by upregulating miR-378a-3p. FASEB J 2020; 34:4540-4556. [PMID: 31999011 DOI: 10.1096/fj.201901879rr] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 01/16/2020] [Accepted: 01/16/2020] [Indexed: 11/11/2022]
Abstract
The mu-opioid receptor gene, OPRM1, undergoes extensive alternative splicing, creating an array of splice variants that are conserved from rodent to human. Both mouse and human OPRM1 have five exon 5-associated seven transmembrane full-length carboxyl terminal variants, MOR-1B1, MOR-1B2, MOR-1B3, MOR-1B4, and MOR-1B5, all of which are derived from alternative 3' splicing from exon 3 to alternative sites within exon 5. The functional relevance of these exon 5-associated MOR-1Bs has been demonstrated in mu agonist-induced G protein coupling, adenylyl cyclase activity, receptor internalization and desensitization, and post-endocytic sorting, as well as region-specific expression at the mRNA level. In the present study, we mapped a polyadenylation site for both mouse and human MOR-1Bs that defines the 3'-untranslated regions (3'-UTR) of MOR-1Bs and stabilizes mMOR-1Bs mRNAs. We identified a conserved miR378a-3p sequence in the 3'-UTR of both mouse and human MOR-1BS transcripts through which miR-378a-3p can regulate the expression of MOR-1Bs at the mRNA level. Chronic morphine treatment significantly increased the miR-378-3p level in Be(2)C cells and the brainstem of the morphine tolerant mice, contributing to the decreased expression of the mouse and human MOR-1B3 and MOR-1B4. Our study provides new insights into the role of miRNAs and Oprm1 splice variants in morphine tolerance.
Collapse
Affiliation(s)
- Zhigang Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,The Affiliated Hospital of Nanjing University of Chinese Medicine, First College of Clinical Medicine, Nanjing, China.,Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jin Xu
- Department of Neurology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA.,Program in Molecular Pharmacology and Chemistry, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Qian Wang
- International Education College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ying-Xian Pan
- Department of Neurology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA.,Program in Molecular Pharmacology and Chemistry, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
23
|
Browne CJ, Godino A, Salery M, Nestler EJ. Epigenetic Mechanisms of Opioid Addiction. Biol Psychiatry 2020; 87:22-33. [PMID: 31477236 PMCID: PMC6898774 DOI: 10.1016/j.biopsych.2019.06.027] [Citation(s) in RCA: 142] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 06/18/2019] [Accepted: 06/19/2019] [Indexed: 12/20/2022]
Abstract
Opioid use kills tens of thousands of Americans each year, devastates families and entire communities, and cripples the health care system. Exposure to opioids causes long-term changes to brain regions involved in reward processing and motivation, leading vulnerable individuals to engage in pathological drug seeking and drug taking that can remain a lifelong struggle. The persistence of these neuroadaptations is mediated in part by epigenetic remodeling of gene expression programs in discrete brain regions. Although the majority of work examining how epigenetic modifications contribute to addiction has focused on psychostimulants such as cocaine, research into opioid-induced changes to the epigenetic landscape is emerging. This review summarizes our knowledge of opioid-induced epigenetic modifications and their consequential changes to gene expression. Current evidence points toward opioids promoting higher levels of permissive histone acetylation and lower levels of repressive histone methylation as well as alterations to DNA methylation patterns and noncoding RNA expression throughout the brain's reward circuitry. Additionally, studies manipulating epigenetic enzymes in specific brain regions are beginning to build causal links between these epigenetic modifications and changes in addiction-related behavior. Moving forward, studies must leverage advanced chromatin analysis and next-generation sequencing approaches combined with bioinformatics pipelines to identify novel gene networks regulated by particular epigenetic modifications. Improved translational relevance also requires increased focus on volitional drug-intake models and standardization of opioid exposure paradigms. Such work will significantly advance our understanding of how opioids cause persistent changes to brain function and will provide a platform on which to develop interventions for treating opioid addiction.
Collapse
Affiliation(s)
- Caleb J Browne
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
| | - Arthur Godino
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L Levy Place, New York, NY 10029, USA
| | - Marine Salery
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L Levy Place, New York, NY 10029, USA
| | - Eric J Nestler
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
24
|
Goetzl L, Thompson-Felix T, Darbinian N, Merabova N, Merali S, Merali C, Sanserino K, Tatevosian T, Fant B, Wimmer ME. Novel biomarkers to assess in utero effects of maternal opioid use: First steps toward understanding short- and long-term neurodevelopmental sequelae. GENES BRAIN AND BEHAVIOR 2019; 18:e12583. [PMID: 31119847 DOI: 10.1111/gbb.12583] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 05/17/2019] [Accepted: 05/21/2019] [Indexed: 12/24/2022]
Abstract
Maternal opioid use disorder is common, resulting in significant neonatal morbidity and cost. Currently, it is not possible to predict which opioid-exposed newborns will require pharmacotherapy for neonatal abstinence syndrome. Further, little is known regarding the effects of maternal opioid use disorder on the developing human brain. We hypothesized that novel methodologies utilizing fetal central nervous system-derived extracellular vesicles isolated from maternal blood can address these gaps in knowledge. Plasma from opioid users and controls between 9 and 21 weeks was precipitated and extracellular vesicles were isolated. Mu opioid and cannabinoid receptor levels were quantified. Label-free proteomics studies and unbiased small RNA next generation sequencing was performed in paired fetal brain tissue. Maternal opioid use disorder increased mu opioid receptor protein levels in extracellular vesicles independent of opioid equivalent dose. Moreover, cannabinoid receptor levels in extracellular vesicles were upregulated with opioid exposure indicating cross talk with endocannabinoids. Maternal opioid use disorder was associated with significant changes in extracellular vesicle protein cargo and fetal brain micro RNA expression, especially in male fetuses. Many of the altered cargo molecules and micro RNAs identified are associated with adverse clinical neurodevelopmental outcomes. Our data suggest that assays relying on extracellular vesicles isolated from maternal blood extracellular vesicles may provide information regarding fetal response to opioids in the setting of maternal opioid use disorder. Prospective clinical studies are needed to evaluate the association between extracellular vesicle biomarkers, risk of neonatal abstinence syndrome and neurodevelopmental outcomes.
Collapse
Affiliation(s)
- Laura Goetzl
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Texas Health Sciences Center, Houston, Texas
| | - Tara Thompson-Felix
- Department of Psychiatry and Behavioral Science, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Nune Darbinian
- Shriners Pediatric Research Center, Center for Neural Repair and Rehabilitation, Temple University, Philadelphia, Pennsylvania
| | - Nana Merabova
- Shriners Pediatric Research Center, Center for Neural Repair and Rehabilitation, Temple University, Philadelphia, Pennsylvania
| | - Salim Merali
- School of Pharmacy, Temple University, Philadelphia, Pennsylvania
| | - Carmen Merali
- School of Pharmacy, Temple University, Philadelphia, Pennsylvania
| | - Kathryne Sanserino
- Department of Obstetrics & Gynecology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Tamara Tatevosian
- Shriners Pediatric Research Center, Center for Neural Repair and Rehabilitation, Temple University, Philadelphia, Pennsylvania
| | - Bruno Fant
- Department of Psychiatry, Center for Neurobiology and Behavior, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mathieu E Wimmer
- Department of Psychology, Temple University, Philadelphia, Pennsylvania
| |
Collapse
|
25
|
Oliver RJ, Mandyam CD. Regulation of Adult Neurogenesis by Non-coding RNAs: Implications for Substance Use Disorders. Front Neurosci 2018; 12:849. [PMID: 30524229 PMCID: PMC6261985 DOI: 10.3389/fnins.2018.00849] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 10/30/2018] [Indexed: 12/25/2022] Open
Abstract
The discovery of non-coding RNAs (ncRNAs)has been one of the central findings from early genomic sequencing studies. Not only was the presence of these genes unknown previously, it was the staggering disproportionate share of the genome that was predicted to be encoded by ncRNAs that was truly significant in genomic research. Over the years the function of various classes of these ncRNAs has been revealed. One of the first and enduring regulatory programs associated with these factors was development. In the neurosciences, the discovery of adult derived populations of dividing cells within the brain was equally substantial. The brain was hypothesized to be plastic only in its neuronal connectivity, but the discovery of the generation of new neurons was a novel mechanism of neuronal and behavioral plasticity. The process of adult neurogenesis resembles early neuronal development and has been found to share many parallels in the proper stages of specified genetic programs. Adult neurogenesis has also been found to play a role in learning and memory involved in particular hippocampal-dependent behaviors. Substance use disorders (SUDs) are an example of a behavioral condition that is associated with and possibly driven by hippocampal alterations. Our laboratory has determined that hippocampal adult neurogenesis is necessary for a rodent model of methamphetamine relapse. Due to the previous research on ncRNAs in development and in other brain regions involved in SUDs, we posit that ncRNAs may play a role in adult neurogenesis associated with this disorder. This review will cover the regulatory mechanisms of various classes of ncRNAs on the coordinated genetic program associated with adult neurogenesis with a special focus on how these programs could be dysregulated in SUDs.
Collapse
Affiliation(s)
- Robert J Oliver
- VA San Diego Healthcare System, San Diego, CA, United States
| | - Chitra D Mandyam
- VA San Diego Healthcare System, San Diego, CA, United States
- Department of Anesthesiology, University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
26
|
Chronic Oxycodone Self-administration Altered Reward-related Genes in the Ventral and Dorsal Striatum of C57BL/6J Mice: An RNA-seq Analysis. Neuroscience 2018; 393:333-349. [PMID: 30059705 DOI: 10.1016/j.neuroscience.2018.07.032] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 07/18/2018] [Accepted: 07/19/2018] [Indexed: 12/28/2022]
Abstract
Prescription opioid abuse, for example of oxycodone, is a pressing public health issue. This study focuses on how chronic oxycodone self-administration (SA) affects the reward pathways in the mouse brain. In this study, we tested the hypothesis that the expression of reward-related genes in the ventral and dorsal striatum, areas involved in different aspects of opioid addiction models, was altered within 1 h after chronic oxycodone SA, using transcriptome-wide sequencing (RNA-seq). Based on results from earlier human genetic and rodent preclinical studies, we focused on a set of genes that may be associated with the development of addictive diseases and the rewarding effect of drugs of abuse, primarily in the opioid, stress response and classical neurotransmitter systems. We found that 32 transcripts in the ventral striatum, and 7 in the dorsal striatum, were altered significantly in adult mice that had self-administered oxycodone (n = 5) for 14 consecutive days (4 h/day) compared with yoked saline controls (n = 5). The following 5 genes in the ventral striatum showed experiment-wise significant changes: proopiomelanocortin (Pomc) and serotonin 5-HT-2A receptor (Htr2a) were upregulated; serotonin receptor 7 (Htr7), galanin receptor1 (Galr1) and glycine receptor 1 (Glra1) were downregulated. Some genes detected by RNA-seq were confirmed by quantitative polymerase chain reaction (qPCR). Conclusion: A RNA-seq study shows that chronic oxycodone SA alters the expression of several reward-related genes in the dorsal and ventral striatum. These results suggest potential mechanisms underlying neuronal adaptation to chronic oxycodone self-exposure, of relevance to our mechanistic understanding of prescription opioid abuse.
Collapse
|
27
|
Smith ACW, Kenny PJ. MicroRNAs regulate synaptic plasticity underlying drug addiction. GENES, BRAIN, AND BEHAVIOR 2018; 17:e12424. [PMID: 28873276 PMCID: PMC5837931 DOI: 10.1111/gbb.12424] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 08/11/2017] [Accepted: 09/01/2017] [Indexed: 12/22/2022]
Abstract
Chronic use of drugs of abuse results in neurochemical, morphological and behavioral plasticity that underlies the emergence of compulsive drug seeking and vulnerability to relapse during periods of attempted abstinence. Identifying and reversing addiction-relevant plasticity is seen as a potential point of pharmacotherapeutic intervention in drug-addicted individuals. Despite considerable advances in our understanding of the actions of drugs of abuse in the brain, this information has thus far yielded few novel treatment options addicted individuals. MicroRNAs are small noncoding RNAs that can each regulate the translation of hundreds to thousands of messenger RNAs. The highly pleiotropic nature of miRNAs has focused attention on their contribution to addiction-relevant structural and functional plasticity in the brain and their potential utility as targets for medications development. In this review, we discuss the roles of miRNAs in synaptic plasticity underlying the development of addiction and then briefly discuss the possibility of using circulating miRNA as biomarkers for addiction.
Collapse
Affiliation(s)
- A. C. W. Smith
- The Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - P. J. Kenny
- The Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
28
|
MicroRNA-124 regulates the expression of MEKK3 in the inflammatory pathogenesis of Parkinson's disease. J Neuroinflammation 2018; 15:13. [PMID: 29329581 PMCID: PMC5767033 DOI: 10.1186/s12974-018-1053-4] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 01/02/2018] [Indexed: 11/21/2022] Open
Abstract
Background Parkinson’s disease (PD) is the most prevalent neurodegenerative disorder that is characterised by selective loss of midbrain dopaminergic (DA) neurons. Chronic inflammation of the central nervous system is mediated by microglial cells and plays a critical role in the pathological progression of PD. Brain-specific microRNA-124 (miR-124) expression is significantly downregulated in lipopolysaccharide (LPS)-treated BV2 cells and in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) model of PD. However, whether abnormal miR-124 expression could regulate the activation of microglia remains poorly understood. Methods BV2 cells were activated by exposure to LPS, and the expression levels of miR-124, mitogen-activated protein kinase kinase kinase 3 (MEKK3), and the nuclear factor of kappaB (NF-κB) p-p65 were analysed. Over-expression and knockdown studies of miR-124 were performed to observe the effects on MEKK3/NF-κB signalling pathways, and the induction of pro-inflammatory and neurotoxic factors was assessed. In addition, a luciferase reporter assay was conducted to confirm whether MEKK3 is a direct target of miR-124. Meanwhile, production of miR-124, MEKK3, and p-p65; midbrain DA neuronal death; or activation of microglia were analysed when treated with or without miR-124 in the MPTP-induced model of PD. Results We found that the knockdown of MEKK3 could inhibit the activation of microglia by regulating NF-κB expression. Over-expression of miR-124 could effectively attenuate the LPS-induced expression of pro-inflammatory cytokines and promote the secretion of neuroprotective factors. We also first identified a unique role of miR-124 in mediating the microglial inflammatory response by targeting MEKK3/NF-κB signalling pathways. In the microglial culture supernatant (MCS) transfer model, over-expression of the miR-124 or knockdown of MEKK3 in BV2 cells prevented SH-SY5Y from apoptosis and death. Moreover, MEKK3 and p-p65 were abundantly expressed in the midbrain. Furthermore, their expression levels increased and microglial activation was observed in the MPTP-induced model of PD. In addition, exogenous delivery of miR-124 could suppress MEKK3 and p-p65 expression and attenuate the activation of microglia in the substantia nigra pars compacta of MPTP-treated mice. miR-124 also could prevent MPTP-dependent apoptotic midbrain DA cell death in a MPTP-induced PD model. Conclusions Taken together, our data suggest that miR-124 can inhibit neuroinflammation in the development of PD by regulating the MEKK3/NF-κB signalling pathways and implicate miR-124 as a potential therapeutic target for regulating the inflammatory response in PD.
Collapse
|
29
|
Yuferov V, Zhang Y, Liang Y, Zhao C, Randesi M, Kreek MJ. Oxycodone Self-Administration Induces Alterations in Expression of Integrin, Semaphorin and Ephrin Genes in the Mouse Striatum. Front Psychiatry 2018; 9:257. [PMID: 29946272 PMCID: PMC6005861 DOI: 10.3389/fpsyt.2018.00257] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Accepted: 05/24/2018] [Indexed: 11/28/2022] Open
Abstract
Oxycodone is one a commonly used medication for pain, and is also a widely abused prescription opioid, like other short-acting MOPr agonists. Neurochemical and structural adaptations in brain following chronic MOPr-agonist administration are thought to underlie pathogenesis and persistence of opiate addiction. Many axon guidance molecules, such as integrins, semaphorins, and ephrins may contribute to oxycodone-induced neuroadaptations through alterations in axon-target connections and synaptogenesis, that may be implicated in the behaviors associated with opiate addiction. However, little is known about this important area. The aim of this study is to investigate alterations in expression of selected integrin, semaphorin, ephrins, netrin, and slit genes in the nucleus accumbens (NAc) and caudate putamen (CPu) of mice following extended 14-day oxycodone self-administration (SA), using RNAseq. Methods: Total RNA from the NAc and CPu were isolated from adult male C57BL/6J mice within 1 h after the last session of oxycodone in a 14-day self-administration paradigm (4h/day, 0.25 mg/kg/infusion, FR1) or from yoked saline controls. Gene expressions were examined using RNA sequencing (RNA-Seq) technology. RNA-Seq libraries were prepared using Illumina's TruSeq® Stranded Total RNA LT kit. The reads were aligned to the mouse reference genome (version mm10) using STAR. DESeq2 was applied to the counts of protein coding genes to estimate the fold change between the treatment groups. False Discovery Rate (FDR) q < 0.1 were used to select genes that have a significant expression change. For selection of a subset of genes related to axon guidance pathway, REACTOME was used. Results: Among 38 known genes of the integrin, semaphorin, and ephrin gene families, RNA-seq data revealed up-regulation of six genes in the NAc: heterodimer receptor, integrins Itgal, Itgb2, and Itgam, and its ligand semaphorin Sema7a, two semaphorin receptors, plexins Plxnd1 and Plxdc1. There was down-regulation of eight genes in this region: two integrin genes Itga3 and Itgb8, semaphorins Sema3c, Sema4g, Sema6a, Sema6d, semaphorin receptor neuropilin Nrp2, and ephrin receptor Epha3. In the CPu, there were five differentially expressed axon guidance genes: up-regulation of three integrin genes, Itgal, Itgb2, Itga1, and down-regulation of Itga9 and ephrin Efna3 were thus observed. No significant alterations in expression of Netrin-1 or Slit were observed. Conclusion: We provide evidence for alterations in the expression of selective axon guidance genes in adult mouse brain following chronic self-administration of oxycodone. Further examination of oxycodone-induced changes in the expression of these specific axon guidance molecules and integrin genes in relation to behavior may provide new insights into development of addiction to oxycodone.
Collapse
Affiliation(s)
- Vadim Yuferov
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY, United States
| | - Yong Zhang
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY, United States
| | - Yupu Liang
- Research Bioinformatics, Clinical and Translational Science Award, The Rockefeller University, New York, NY, United States
| | - Connie Zhao
- Genomic Resource Center, The Rockefeller University, New York, NY, United States
| | - Matthew Randesi
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY, United States
| | - Mary J Kreek
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY, United States
| |
Collapse
|
30
|
Geng L, Liu W, Chen Y. miR-124-3p attenuates MPP +-induced neuronal injury by targeting STAT3 in SH-SY5Y cells. Exp Biol Med (Maywood) 2017; 242:1757-1764. [PMID: 28958159 PMCID: PMC5714150 DOI: 10.1177/1535370217734492] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 08/09/2017] [Indexed: 01/12/2023] Open
Abstract
Ample evidence has demonstrated the involvement of microRNAs in Parkinson's disease pathogenesis. miR-124-3p was reported to be able to improve neural functional recovery. However, the underlying mechanism of miR-124-3p in Parkinson's disease progression was not well established. This study was designed to investigate the role of miR-124-3p in methyl phenyl pyridinium iodide (MPP)+-induced SH-SY5Y cells, an in vitro Parkinson's disease model. It is observed that miR-124-3p expression was decreased, and STAT3 expression was increased in MPP+-induced SH-SY5Y cells. miR-124-3p overexpression attenuated MPP+-induced neuronal injury, displayed as increased cell viability and superoxide dismutase activities, as well as reduced cell apoptosis, Caspase-3 activity, lactate dehydrogenase activity, inflammatory factors TNF-α, and IL-1β levels and reactive oxygen species generation. Moreover, STAT3 was confirmed to be a miR-124-3p target. Restored STAT3 expression reversed miR-124-3p-induced neuroprotective effects against MPP+-mediated neuronal injury. These data demonstrated that miR-124-3p contributed to neuroprotective effects in MPP+-induced Parkinson's disease cell model by targeting STAT3. Impact statement PD affects millions of people in the world, causing uncontrolled tremors. MicroRNAs, a class of endogenous single-stranded non-coding transcript with approximately 22 nucleotides, could bind to the 3″ UTR of their targets. The functional action of miR-124-3p in PD was not fully elucidated. Our study found that ectopic expression miR-124-3p attenuated MPP+-induced injury in PD model in vitro by suppressing neurotoxicity, neuronal apoptosis, neuroinflammation, and oxidative stress. Moreover, we validated that miR-124-3p could bind to STAT3 mediating the neuroprotective effect of miR-124-3p. We believe this study will be interesting for readers of this area.
Collapse
Affiliation(s)
- Lijiao Geng
- 1 Department of Rehabilitation Medicine, Huaihe Hospital of Henan University, Kaifeng 475000, China
| | - Wei Liu
- 2 Department of Neurology, Huaihe Hospital of Henan University, Kaifeng 475000, China
| | - Yong Chen
- 1 Department of Rehabilitation Medicine, Huaihe Hospital of Henan University, Kaifeng 475000, China
| |
Collapse
|
31
|
Marie-Claire C, Jourdaine C, Lépine JP, Bellivier F, Bloch V, Vorspan F. Pharmacoepigenomics of opiates and methadone maintenance treatment: current data and perspectives. Pharmacogenomics 2017; 18:1359-1372. [DOI: 10.2217/pgs-2017-0040] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Current treatments of opioid addiction include primarily maintenance medications such as methadone. Chronic exposure to opiate and/or long-lasting maintenance treatment induce modulations of gene expression in brain and peripheral tissues. There is increasing evidence that epigenetic modifications underlie these modulations. This review summarizes published results on opioid-induced epigenetic changes in animal models and in patients. The epigenetic modifications observed with other drugs of abuse often used by opiate abusers are also outlined. Specific methadone maintenance treatment induced epigenetic modifications at different treatment stages may be combined with the ones resulting from patients’ substance use history. Therefore, research comparing groups of addicts with similar history and substances use disorders but contrasting for well-characterized treatment phenotypes should be encouraged.
Collapse
Affiliation(s)
- Cynthia Marie-Claire
- Variabilité de réponse aux psychotropes, INSERMU1144/Faculté de Pharmacie de Paris/Université Paris Descartes/Université ParisDiderot/Université Sorbonne Paris Cité, Paris, France
| | - Clément Jourdaine
- AP-HP, GH Saint-Louis – Lariboisière – F. Widal, Pôle de Psychiatrie et de Médecine Addictologique, 75475 Paris cedex 10, France
| | - Jean-Pierre Lépine
- AP-HP, GH Saint-Louis – Lariboisière – F. Widal, Pôle de Psychiatrie et de Médecine Addictologique, 75475 Paris cedex 10, France
| | - Frank Bellivier
- Variabilité de réponse aux psychotropes, INSERMU1144/Faculté de Pharmacie de Paris/Université Paris Descartes/Université ParisDiderot/Université Sorbonne Paris Cité, Paris, France
- AP-HP, GH Saint-Louis – Lariboisière – F. Widal, Pôle de Psychiatrie et de Médecine Addictologique, 75475 Paris cedex 10, France
| | - Vanessa Bloch
- Variabilité de réponse aux psychotropes, INSERMU1144/Faculté de Pharmacie de Paris/Université Paris Descartes/Université ParisDiderot/Université Sorbonne Paris Cité, Paris, France
| | - Florence Vorspan
- Variabilité de réponse aux psychotropes, INSERMU1144/Faculté de Pharmacie de Paris/Université Paris Descartes/Université ParisDiderot/Université Sorbonne Paris Cité, Paris, France
- AP-HP, GH Saint-Louis – Lariboisière – F. Widal, Pôle de Psychiatrie et de Médecine Addictologique, 75475 Paris cedex 10, France
| |
Collapse
|
32
|
Motti D, Lerch JK, Danzi MC, Gans JH, Kuo F, Slepak TI, Bixby JL, Lemmon VP. Identification of miRNAs involved in DRG neurite outgrowth and their putative targets. FEBS Lett 2017; 591:2091-2105. [PMID: 28626869 PMCID: PMC5864114 DOI: 10.1002/1873-3468.12718] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 06/07/2017] [Accepted: 06/11/2017] [Indexed: 12/14/2022]
Abstract
Peripheral neurons regenerate their axons after injury. Transcriptional regulation by microRNAs (miRNAs) is one possible mechanism controlling regeneration. We profiled miRNA expression in mouse dorsal root ganglion neurons after a sciatic nerve crush, and identified 49 differentially expressed miRNAs. We evaluated the functional role of each miRNA using a phenotypic analysis approach. To predict the targets of the miRNAs we employed RNA-Sequencing and examined transcription at the isoform level. We identify thousands of differentially expressed isoforms and bioinformatically associate the miRNAs that modulate neurite growth with their putative target isoforms to outline a network of regulatory events underlying peripheral nerve regeneration. MiR-298, let-7a, and let-7f enhance neurite growth and target the majority of isoforms in the differentially expressed network.
Collapse
Affiliation(s)
- Dario Motti
- The Miami Project To Cure Paralysis, The University of Miami Miller School of Medicine, Miami, FL
| | - Jessica K. Lerch
- The Department of Neuroscience, The Ohio State University, Columbus, OH
| | - Matt C. Danzi
- The Miami Project To Cure Paralysis, The University of Miami Miller School of Medicine, Miami, FL
| | - Jared H. Gans
- The Miami Project To Cure Paralysis, The University of Miami Miller School of Medicine, Miami, FL
| | - Frank Kuo
- The Miami Project To Cure Paralysis, The University of Miami Miller School of Medicine, Miami, FL
| | - Tatiana I. Slepak
- The Miami Project To Cure Paralysis, The University of Miami Miller School of Medicine, Miami, FL
| | - John L. Bixby
- The Miami Project To Cure Paralysis, The University of Miami Miller School of Medicine, Miami, FL
- The Department of Molecular and Cellular Pharmacology, The University of Miami Miller School of Medicine, Miami, FL
- The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, FL
- The Center for Computational Science, The University of Miami, Miami, FL
| | - Vance P. Lemmon
- The Miami Project To Cure Paralysis, The University of Miami Miller School of Medicine, Miami, FL
- The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, FL
- The Center for Computational Science, The University of Miami, Miami, FL
| |
Collapse
|
33
|
Cahill CM, Walwyn W, Taylor AMW, Pradhan AAA, Evans CJ. Allostatic Mechanisms of Opioid Tolerance Beyond Desensitization and Downregulation. Trends Pharmacol Sci 2016; 37:963-976. [PMID: 27670390 DOI: 10.1016/j.tips.2016.08.002] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 08/18/2016] [Accepted: 08/19/2016] [Indexed: 12/20/2022]
Abstract
Mechanisms of opioid tolerance have focused on adaptive modifications within cells containing opioid receptors, defined here as cellular allostasis, emphasizing regulation of the opioid receptor signalosome. We review additional regulatory and opponent processes involved in behavioral tolerance, and include mechanistic differences both between agonists (agonist bias), and between μ- and δ-opioid receptors. In a process we will refer to as pass-forward allostasis, cells modified directly by opioid drugs impute allostatic changes to downstream circuitry. Because of the broad distribution of opioid systems, every brain cell may be touched by pass-forward allostasis in the opioid-dependent/tolerant state. We will implicate neurons and microglia as interactive contributors to the cumulative allostatic processes creating analgesic and hedonic tolerance to opioid drugs.
Collapse
Affiliation(s)
- Catherine M Cahill
- Department of Anesthesiology and Perioperative Care, University of California, Irvine, 837 Health Sciences Road, Irvine, CA 92697, USA
| | - Wendy Walwyn
- Hatos Center for Neuropharmacology, Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, 675 Charles E. Young Drive South, Los Angeles, CA 90095, USA
| | - Anna M W Taylor
- Hatos Center for Neuropharmacology, Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, 675 Charles E. Young Drive South, Los Angeles, CA 90095, USA
| | - Amynah A A Pradhan
- Department of Psychiatry, University of Illinois at Chicago, 1601 West Taylor Street, Chicago, IL 60612, USA
| | - Christopher J Evans
- Hatos Center for Neuropharmacology, Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, 675 Charles E. Young Drive South, Los Angeles, CA 90095, USA.
| |
Collapse
|
34
|
Gong X, Wang H, Ye Y, Shu Y, Deng Y, He X, Lu G, Zhang S. miR-124 regulates cell apoptosis and autophagy in dopaminergic neurons and protects them by regulating AMPK/mTOR pathway in Parkinson's disease. Am J Transl Res 2016; 8:2127-2137. [PMID: 27347320 PMCID: PMC4891425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 02/29/2016] [Indexed: 06/06/2023]
Abstract
The important roles of miR-124 in the development and progression of various diseases are being increasing recognized. This study was aimed to investigate the potential roles of miR-124 in dopaminergic (DA) neuronal apoptosis and autophagy in Parkinson's disease (PD) and to explore their mechanisms. Human SH-SY5Y cells that are treated with MPTP were transfected with mature miR-124 vector and control empty vector. The effect of MPTP on miR-124 mRNA level was analyzed using RT-PCR analysis. Furthermore, the effects of miR-124 expression on neuronal apoptosis and autophagy, as well as the expression of proteins in the AMPK/mTOR pathway, were analyzed using RT-PCR and western blotting. This study found that miR-124 was down-regulated in the MPTP-treated (100 μM) neurons, and miR-124 suppression significantly increased cell apoptosis and induced autophagy-associated protein expression, including that of Beclin 1 and increased the ratio of LC3 II/LC3 I compared with that in controls. In addition, in vitro rescue of miR-124 significantly decreased the percentage of apoptotic cells and the ratio of LC3 II/LC3 I, findings that were approximately equal to the controls. Moreover, miR-124 suppression increased p-AMPK but decreased p-mTOR levels in neurons. Our study suggested that miR-124 functions as a protector of DA neurons during PD through the involvement of cell apoptosis and autophagy by regulating the AMPK/mTOR pathway.
Collapse
Affiliation(s)
- Xin Gong
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University; The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and RegenerationGuangzhou 510282, Guangdong, China
- Department of Neurosurgery, Hunan Provincial People’s HospitalChangsha 410005, Hunan, China
| | - Huiqing Wang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University; The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and RegenerationGuangzhou 510282, Guangdong, China
| | - Yongyi Ye
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University; The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and RegenerationGuangzhou 510282, Guangdong, China
| | - Yugao Shu
- Department of Neurosurgery, Hunan Provincial People’s HospitalChangsha 410005, Hunan, China
| | - Yongwen Deng
- Department of Neurosurgery, Hunan Provincial People’s HospitalChangsha 410005, Hunan, China
| | - Xiaozheng He
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University; The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and RegenerationGuangzhou 510282, Guangdong, China
| | - Guohui Lu
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University; The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and RegenerationGuangzhou 510282, Guangdong, China
| | - Shizhong Zhang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University; The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and RegenerationGuangzhou 510282, Guangdong, China
| |
Collapse
|
35
|
Tapocik JD, Ceniccola K, Mayo CL, Schwandt ML, Solomon M, Wang BD, Luu TV, Olender J, Harrigan T, Maynard TM, Elmer GI, Lee NH. MicroRNAs Are Involved in the Development of Morphine-Induced Analgesic Tolerance and Regulate Functionally Relevant Changes in Serpini1. Front Mol Neurosci 2016; 9:20. [PMID: 27047334 PMCID: PMC4805586 DOI: 10.3389/fnmol.2016.00020] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 02/29/2016] [Indexed: 12/23/2022] Open
Abstract
Long-term opioid treatment results in reduced therapeutic efficacy and in turn leads to an increase in the dose required to produce equivalent pain relief and alleviate break-through or insurmountable pain. Altered gene expression is a likely means for inducing long-term neuroadaptations responsible for tolerance. Studies conducted by our laboratory (Tapocik et al., 2009) revealed a network of gene expression changes occurring in canonical pathways involved in neuroplasticity, and uncovered miRNA processing as a potential mechanism. In particular, the mRNA coding the protein responsible for processing miRNAs, Dicer1, was positively correlated with the development of analgesic tolerance. The purpose of the present study was to test the hypothesis that miRNAs play a significant role in the development of analgesic tolerance as measured by thermal nociception. Dicer1 knockdown, miRNA profiling, bioinformatics, and confirmation of high value targets were used to test the proposition. Regionally targeted Dicer1 knockdown (via shRNA) had the anticipated consequence of eliminating the development of tolerance in C57BL/6J (B6) mice, thus supporting the involvement of miRNAs in the development of tolerance. MiRNA expression profiling identified a core set of chronic morphine-regulated miRNAs (miR's 27a, 9, 483, 505, 146b, 202). Bioinformatics approaches were implemented to identify and prioritize their predicted target mRNAs. We focused our attention on miR27a and its predicted target serpin peptidase inhibitor clade I (Serpini1) mRNA, a transcript known to be intricately involved in dendritic spine density regulation in a manner consistent with chronic morphine's consequences and previously found to be correlated with the development of analgesic tolerance. In vitro reporter assay confirmed the targeting of the Serpini1 3'-untranslated region by miR27a. Interestingly miR27a was found to positively regulate Serpini1 mRNA and protein levels in multiple neuronal cell lines. Lastly, Serpini1 knockout mice developed analgesic tolerance at a slower rate than wild-type mice thus confirming a role for the protein in analgesic tolerance. Overall, these results provide evidence to support a specific role for miR27a and Serpini1 in the behavioral response to chronic opioid administration (COA) and suggest that miRNA expression and mRNA targeting may underlie the neuroadaptations that mediate tolerance to the analgesic effects of morphine.
Collapse
Affiliation(s)
- Jenica D. Tapocik
- National Institute of Alcohol Abuse and Alcoholism, National Institutes of HealthBethesda, MD, USA
| | - Kristin Ceniccola
- Department of Pharmacology and Physiology, The George Washington UniversityWashington, DC, USA
| | - Cheryl L. Mayo
- Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of MedicineBaltimore, MD, USA
| | - Melanie L. Schwandt
- National Institute of Alcohol Abuse and Alcoholism, National Institutes of HealthBethesda, MD, USA
| | - Matthew Solomon
- National Institute of Alcohol Abuse and Alcoholism, National Institutes of HealthBethesda, MD, USA
| | - Bi-Dar Wang
- Department of Pharmacology and Physiology, The George Washington UniversityWashington, DC, USA
| | - Truong V. Luu
- Department of Pharmacology and Physiology, The George Washington UniversityWashington, DC, USA
| | - Jacqueline Olender
- Department of Pharmacology and Physiology, The George Washington UniversityWashington, DC, USA
| | - Thomas Harrigan
- Department of Pharmacology and Physiology, The George Washington UniversityWashington, DC, USA
| | - Thomas M. Maynard
- Department of Pharmacology and Physiology, The George Washington UniversityWashington, DC, USA
| | - Greg I. Elmer
- Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of MedicineBaltimore, MD, USA
| | - Norman H. Lee
- Department of Pharmacology and Physiology, The George Washington UniversityWashington, DC, USA
| |
Collapse
|
36
|
Heyer MP, Kenny PJ. Corticostriatal microRNAs in addiction. Brain Res 2015; 1628:2-16. [DOI: 10.1016/j.brainres.2015.07.047] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 07/11/2015] [Accepted: 07/25/2015] [Indexed: 01/28/2023]
|
37
|
McAdams RM, McPherson RJ, Beyer RP, Bammler TK, Farin FM, Juul SE. Dose-dependent effects of morphine exposure on mRNA and microRNA (miR) expression in hippocampus of stressed neonatal mice. PLoS One 2015; 10:e0123047. [PMID: 25844808 PMCID: PMC4386824 DOI: 10.1371/journal.pone.0123047] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 02/18/2015] [Indexed: 12/02/2022] Open
Abstract
Morphine is used to sedate critically ill infants to treat painful or stressful conditions associated with intensive care. Whether neonatal morphine exposure affects microRNA (miR) expression and thereby alters mRNA regulation is unknown. We tested the hypothesis that repeated morphine treatment in stress-exposed neonatal mice alters hippocampal mRNA and miR expression. C57BL/6 male mice were treated from postnatal day (P) 5 to P9 with morphine sulfate at 2 or 5 mg/kg ip twice daily and then exposed to stress consisting of hypoxia (100% N2 1 min and 100% O2 5 min) followed by 2h maternal separation. Control mice were untreated and dam-reared. mRNA and miR expression profiling was performed on hippocampal tissues at P9. Overall, 2 and 5 mg/kg morphine treatment altered expression of a total of 150 transcripts (>1.5 fold change, P<0.05) from which 100 unique mRNAs were recognized (21 genes were up- and 79 genes were down-regulated), and 5 mg/kg morphine affected 63 mRNAs exclusively. The most upregulated mRNAs were fidgetin, arginine vasopressin, and resistin-like alpha, and the most down-regulated were defensin beta 11, aquaporin 1, calmodulin-like 4, chloride intracellular channel 6, and claudin 2. Gene Set Enrichment Analysis revealed that morphine treatment affected pathways related to cell cycle, membrane function, signaling, metabolism, cell death, transcriptional regulation, and immune response. Morphine decreased expression of miR-204-5p, miR-455-3p, miR-448-5p, and miR-574-3p. Nine morphine-responsive mRNAs that are involved in neurodevelopment, neurotransmission, and inflammation are predicted targets of the aforementioned differentially expressed miRs. These data establish that morphine produces dose-dependent changes in both hippocampal mRNA and miR expression in stressed neonatal mice. If permanent, morphine–mediated neuroepigenetic effects may affect long-term hippocampal function, and this provides a mechanism for the neonatal morphine-related impairment of adult learning.
Collapse
Affiliation(s)
- Ryan M. McAdams
- Department of Pediatrics, Division of Neonatology, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| | - Ronald J. McPherson
- Department of Pediatrics, Division of Neonatology, University of Washington, Seattle, Washington, United States of America
| | - Richard P. Beyer
- Dept of Environmental & Occupational Health Sciences, University of Washington, Seattle, Washington, United States of America
| | - Theo K. Bammler
- Dept of Environmental & Occupational Health Sciences, University of Washington, Seattle, Washington, United States of America
| | - Frederico M. Farin
- Dept of Environmental & Occupational Health Sciences, University of Washington, Seattle, Washington, United States of America
| | - Sandra E. Juul
- Department of Pediatrics, Division of Neonatology, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
38
|
García-Pérez D, López-Bellido R, Hidalgo JM, Rodríguez RE, Laorden ML, Núñez C, Milanés MV. Morphine regulates Argonaute 2 and TH expression and activity but not miR-133b in midbrain dopaminergic neurons. Addict Biol 2015; 20:104-19. [PMID: 23927484 DOI: 10.1111/adb.12083] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Epigenetic changes such as microRNAs (miRs)/Ago2-induced gene silencing represent complex molecular signature that regulate cellular plasticity. Recent studies showed involvement of miRs and Ago2 in drug addiction. In this study, we show that changes in gene expression induced by morphine and morphine withdrawal occur with concomitant epigenetic modifications in the mesolimbic dopaminergic (DA) pathway [ventral tegmental area (VTA)/nucleus accumbens (NAc) shell], which is critically involved in drug-induced dependence. We found that acute or chronic morphine administration as well as morphine withdrawal did not modify miR-133b messenger RNA (mRNA) expression in the VTA, whereas Ago2 protein levels were decreased and increased in morphine-dependent rats and after morphine withdrawal, respectively. These changes were paralleled with enhanced and decreased NAc tyrosine hydroxylase (TH) protein (an early DA marker) in morphine-dependent rats and after withdrawal, respectively. We also observed changes in TH mRNA expression in the VTA that could be related to Ago2-induced translational repression of TH mRNA during morphine withdrawal. However, the VTA number of TH-positive neurons suffered no alterations after the different treatment. Acute morphine administration produced a marked increase in TH activity and DA turnover in the NAc (shell). In contrast, precipitated morphine withdrawal decreased TH activation and did not change DA turnover. These findings provide new information into the possible correlation between Ago2/miRs complex regulation and DA neurons plasticity during opiate addiction.
Collapse
Affiliation(s)
- Daniel García-Pérez
- Group of Cellular and Molecular Pharmacology; Faculty of Medicine; University of Murcia; Murcia Spain
- IMIB (Murcia Institute of Biomedical Investigation); Murcia Spain
| | - Roger López-Bellido
- Department of Biochemistry and Molecular Biology; Institute of Neurosciences; University of Salamanca; Salamanca Spain
| | - Juana M. Hidalgo
- Group of Cellular and Molecular Pharmacology; Faculty of Medicine; University of Murcia; Murcia Spain
- IMIB (Murcia Institute of Biomedical Investigation); Murcia Spain
| | - Raquel E. Rodríguez
- Department of Biochemistry and Molecular Biology; Institute of Neurosciences; University of Salamanca; Salamanca Spain
| | - Maria Luisa Laorden
- Group of Cellular and Molecular Pharmacology; Faculty of Medicine; University of Murcia; Murcia Spain
- IMIB (Murcia Institute of Biomedical Investigation); Murcia Spain
| | - Cristina Núñez
- Group of Cellular and Molecular Pharmacology; Faculty of Medicine; University of Murcia; Murcia Spain
- IMIB (Murcia Institute of Biomedical Investigation); Murcia Spain
| | - Maria Victoria Milanés
- Group of Cellular and Molecular Pharmacology; Faculty of Medicine; University of Murcia; Murcia Spain
- IMIB (Murcia Institute of Biomedical Investigation); Murcia Spain
| |
Collapse
|
39
|
Abstract
This paper is the thirty-sixth consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2013 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior, and the roles of these opioid peptides and receptors in pain and analgesia; stress and social status; tolerance and dependence; learning and memory; eating and drinking; alcohol and drugs of abuse; sexual activity and hormones, pregnancy, development and endocrinology; mental illness and mood; seizures and neurologic disorders; electrical-related activity and neurophysiology; general activity and locomotion; gastrointestinal, renal and hepatic functions; cardiovascular responses; respiration and thermoregulation; and immunological responses.
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, Flushing, NY 11367, United States.
| |
Collapse
|
40
|
Herrero-Turrión MJ, Rodríguez-Martín I, López-Bellido R, Rodríguez RE. Whole-genome expression profile in zebrafish embryos after chronic exposure to morphine: identification of new genes associated with neuronal function and mu opioid receptor expression. BMC Genomics 2014; 15:874. [PMID: 25294025 PMCID: PMC4201762 DOI: 10.1186/1471-2164-15-874] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 09/24/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND A great number of studies have investigated changes induced by morphine exposure in gene expression using several experimental models. In this study, we examined gene expression changes during chronic exposure to morphine during maturation and differentiation of zebrafish CNS. RESULTS Microarray analysis showed 254 genes whose expression was identified as different by at least 1.3 fold change following chronic morphine exposure as compared to controls. Of these, several novel genes (grb2, copb2, otpb, magi1b, grik-l, bnip4 and sox19b) have been detected for the first time in an experimental animal model treated with morphine. We have also identified a subset of genes (dao.1, wls, bnip4 and camk1γb) differentially expressed by chronic morphine exposure whose expression is related to mu opioid receptor gene expression. Altered expression of copb2, bnip4, sox19b, otpb, dao.1, grik-l and wls is indicative of modified neuronal development, CNS patterning processes, differentiation and dopaminergic neurotransmission, serotonergic signaling pathway, and glutamatergic neurotransmission. The deregulation of camk1γb signaling genes suggests an activation of axonogenesis and dendritogenesis. CONCLUSIONS Our study identified different functional classes of genes and individual candidates involved in the mechanisms underlying susceptibility to morphine actions related to CNS development. These results open new lines to study the treatment of pain and the molecular mechanisms involved in addiction. We also found a set of zebrafish-specific morphine-induced genes, which may be putative targets in human models for addiction and pain processes.
Collapse
Affiliation(s)
| | | | | | - Raquel E Rodríguez
- Instituto de Neurociencias de Castilla y León, University of Salamanca, Salamanca 37007, Spain.
| |
Collapse
|
41
|
Lu Z, Xu J, Xu M, Pasternak GW, Pan YX. Morphine regulates expression of μ-opioid receptor MOR-1A, an intron-retention carboxyl terminal splice variant of the μ-opioid receptor (OPRM1) gene via miR-103/miR-107. Mol Pharmacol 2014; 85:368-80. [PMID: 24302561 PMCID: PMC3913361 DOI: 10.1124/mol.113.089292] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Accepted: 12/03/2013] [Indexed: 01/27/2023] Open
Abstract
The μ-opioid receptor (MOR-1) gene OPRM1 undergoes extensive alternative splicing, generating an array of splice variants. Of these variants, MOR-1A, an intron-retention carboxyl terminal splice variant identical to MOR-1 except for the terminal intracellular tail encoded by exon 3b, is quite abundant and conserved from rodent to humans. Increasing evidence indicates that miroRNAs (miRNAs) regulate MOR-1 expression and that μ agonists such as morphine modulate miRNA expression. However, little is known about miRNA regulation of the OPRM1 splice variants. Using 3'-rapid amplification cDNA end and Northern blot analyses, we identified the complete 3'-untranslated region (3'-UTR) for both mouse and human MOR-1A and their conserved polyadenylation site, and defined the role the 3'-UTR in mRNA stability using a luciferase reporter assay. Computer models predicted a conserved miR-103/107 targeting site in the 3'-UTR of both mouse and human MOR-1A. The functional relevance of miR-103/107 in regulating expression of MOR-1A protein through the consensus miR-103/107 binding sites in the 3'-UTR was established by using mutagenesis and a miR-107 inhibitor in transfected human embryonic kidney 293 cells and Be(2)C cells that endogenously express human MOR-1A. Chronic morphine treatment significantly upregulated miR-103 and miR-107 levels, leading to downregulation of polyribosome-associated MOR-1A in both Be(2)C cells and the striatum of a morphine-tolerant mouse, providing a new perspective on understanding the roles of miRNAs and OPRM1 splice variants in modulating the complex actions of morphine in animals and humans.
Collapse
Affiliation(s)
- Zhigang Lu
- Department of Neurology and the Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, New York
| | | | | | | | | |
Collapse
|
42
|
Han Z, Liu Q, Huang Z, Cui W, Tian Y, Yan W, Wu Q. Expression and imprinting analysis of AK044800, a transcript from the Dlk1-Dio3 imprinted gene cluster during mouse embryogenesis. Mol Cells 2013; 35:285-90. [PMID: 23515577 PMCID: PMC3887882 DOI: 10.1007/s10059-013-2275-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 01/25/2013] [Accepted: 02/04/2013] [Indexed: 01/11/2023] Open
Abstract
Recent advances of induced pluripotent stem cells (iPSCs) has demonstrated that full development potential is closely related with the expression state of noncoding RNAs (ncRNAs) of the Dlk1-Dio3 imprinted gene cluster. However, few of them, especially the long noncoding RNAs (lncRNAs), have been characterized in detail. AK044800 is a transcript from the Dlk1-Dio3 imprinted region with little known information. This study reports original data on the expression pattern of AK044800 during embryogenesis. Expression analysis showed that AK044800 was specifically expressed in the brain at mid-gestation, E9.5 and E11.5. And at E15.5, its expression was mainly concentrated in the forebrain. In the late-gestation stage (E18.5), AK044800 expression was weaker in the brain and began to emerge in some other tissues during this period. Notably, the expression of AK044800 was biallelic in the brain, unlike other noncoding transcripts from this imprinted region. In addition, its expression was dependent on inbred mouse strains. This may be the first lncRNA that has been identified with a different expression between inbred mouse strains. This study may provide useful clues for further investigations of expression regulation and functions of lncRNAs of the Dlk1-Dio3 imprinted region.
Collapse
Affiliation(s)
- Zhengbin Han
- School of Life Science and Technology, Harbin Institute of Technology, Science Park of Harbin Institute of Technology, Harbin,
China
| | - Qi Liu
- School of Life Science and Technology, Harbin Institute of Technology, Science Park of Harbin Institute of Technology, Harbin,
China
| | - Zhijun Huang
- School of Life Science and Technology, Harbin Institute of Technology, Science Park of Harbin Institute of Technology, Harbin,
China
| | - Wei Cui
- School of Life Science and Technology, Harbin Institute of Technology, Science Park of Harbin Institute of Technology, Harbin,
China
| | - Yijun Tian
- School of Life Science and Technology, Harbin Institute of Technology, Science Park of Harbin Institute of Technology, Harbin,
China
| | - Weili Yan
- School of Life Science and Technology, Harbin Institute of Technology, Science Park of Harbin Institute of Technology, Harbin,
China
| | - Qiong Wu
- School of Life Science and Technology, Harbin Institute of Technology, Science Park of Harbin Institute of Technology, Harbin,
China
| |
Collapse
|