1
|
Liu M, Si Z. An update: epigenetic mechanisms underlying methamphetamine addiction. Front Cell Dev Biol 2024; 12:1494557. [PMID: 39650725 PMCID: PMC11621221 DOI: 10.3389/fcell.2024.1494557] [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: 09/11/2024] [Accepted: 11/11/2024] [Indexed: 12/11/2024] Open
Abstract
Methamphetamine (METH) is one of the most widely abused illicit drugs globally. Despite its widespread abuse, the effects of methamphetamine on the brain and the precise mechanisms underlying addiction remain poorly understood. Elucidating these biological mechanisms and developing effective treatments is of utmost importance. Researchers have adopted a multi-faceted approach, combining studies at the genetic, molecular, organ, and individual levels, to explore the epigenetic changes that methamphetamine use brings to an organism from both micro and macro perspectives. They utilize a comparative analysis of experimental animal data and clinical cases to ascertain differences and identify potential targets for translating METH addiction research from the experimental to the clinical setting. Recent studies have demonstrated that epigenetic regulation plays a pivotal role in neural mechanisms, encompassing DNA methylation, histone modifications (such as acetylation and methylation), ubiquitination, phosphorylation, and the regulation of non-coding RNA. These epigenetic factors influence an individual's susceptibility and response to methamphetamine addiction by regulating the expression of specific genes. Specifically, methamphetamine use has been observed to cause alterations in DNA methylation status, which in turn affects the expression of genes associated with neuroreward pathways, leading to alterations in brain function and structure. Furthermore, histone modifications have significant implications for the neurotoxicity associated with methamphetamine addiction. For instance, the methylation and acetylation of histone H3 modify chromatin structure, consequently influencing the transcriptional activity of genes. Non-coding RNAs, including microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), also play a pivotal role in methamphetamine addiction by interacting with messenger RNAs (mRNAs) and regulating gene expression. To further advance our understanding, researchers employ advanced technologies such as high-throughput sequencing, chromatin immunoprecipitation sequencing (ChIP-seq), and RNA sequencing (RNA-seq) to comprehensively analyze epigenetic changes in both animal models and human subjects. These technologies enable researchers to identify specific epigenetic markers associated with methamphetamine addiction and to explore their functional consequences. This article reviews the role of these epigenetic mechanisms in methamphetamine addiction and discusses their potential implications for future clinical treatment strategies, particularly in the development of drugs targeting methamphetamine addiction. By deepening our comprehension of these epigenetic regulatory mechanisms, it is anticipated that targeted therapeutic strategies may be devised to reverse the gene expression alterations associated with methamphetamine addiction, thus enhancing the efficacy of addiction treatment and paving the way for future research in this domain.
Collapse
Affiliation(s)
| | - Zizhen Si
- Department of Medicine, Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
2
|
Jasim SA, Altalbawy FMA, Abohassan M, Oghenemaro EF, Bishoyi AK, Singh RP, Kaur P, Sivaprasad GV, Mohammed JS, Hulail HM. Histone Deacetylases (HDACs) Roles in Inflammation-mediated Diseases; Current Knowledge. Cell Biochem Biophys 2024:10.1007/s12013-024-01587-0. [PMID: 39419931 DOI: 10.1007/s12013-024-01587-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2024] [Indexed: 10/19/2024]
Abstract
The histone acetyl transferases (HATs) and histone deacetylases (HDACs), which are mostly recognized for their involvement in regulating chromatin remodeling via histone acetylation/deacetylation, have been shown to also change several non-histone proteins to regulate other cellular processes. Acetylation affects the activity or function of cytokine receptors, nuclear hormone receptors, intracellular signaling molecules, and transcription factors in connection to inflammation. Some small-molecule HDAC inhibitors are utilized as anticancer medications in clinical settings due to their capability to regulate cellular growth arrest, differentiation, and death. Here, we summarize our present knowledge of the innate and adaptive immunological pathways that classical HDAC enzymes control. The aim is to justify the targeted (or non-targeted) use of inhibitors against certain HDAC enzymes in inflammatory diseases such as arthritis, inflammatory bowel diseases (IBD), airways inflammation and neurological diseases.
Collapse
Affiliation(s)
- Saade Abdalkareem Jasim
- Medical Laboratory Techniques department, College of Health and Medical Technology, University of Al-maarif, Anbar, Iraq
| | - Farag M A Altalbawy
- Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia.
| | - Mohammad Abohassan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Enwa Felix Oghenemaro
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Delta State University, Abraka, Delta State, Nigeria
| | - Ashok Kumar Bishoyi
- Department of Microbiology, Faculty of Science, Marwadi University Research Center, Marwadi University, Rajkot, 360003, Gujarat, India
| | - Ravindra Pal Singh
- Department of Pharmaceutics, NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Parjinder Kaur
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - G V Sivaprasad
- Department of Basic Science & Humanities, Raghu Engineering College, Visakhapatnam, India
| | | | - Hanen Mahmod Hulail
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| |
Collapse
|
3
|
Nohesara S, Mostafavi Abdolmaleky H, Thiagalingam S. Substance-Induced Psychiatric Disorders, Epigenetic and Microbiome Alterations, and Potential for Therapeutic Interventions. Brain Sci 2024; 14:769. [PMID: 39199463 PMCID: PMC11352452 DOI: 10.3390/brainsci14080769] [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: 06/23/2024] [Revised: 07/18/2024] [Accepted: 07/25/2024] [Indexed: 09/01/2024] Open
Abstract
Substance use disorders (SUDs) are complex biopsychosocial diseases that cause neurocognitive deficits and neurological impairments by altering the gene expression in reward-related brain areas. Repeated drug use gives rise to alterations in DNA methylation, histone modifications, and the expression of microRNAs in several brain areas that may be associated with the development of psychotic symptoms. The first section of this review discusses how substance use contributes to the development of psychotic symptoms via epigenetic alterations. Then, we present more evidence about the link between SUDs and brain epigenetic alterations. The next section presents associations between paternal and maternal exposure to substances and epigenetic alterations in the brains of offspring and the role of maternal diet in preventing substance-induced neurological impairments. Then, we introduce potential therapeutic agents/approaches such as methyl-rich diets to modify epigenetic alterations for alleviating psychotic symptoms or depression in SUDs. Next, we discuss how substance use-gut microbiome interactions contribute to the development of neurological impairments through epigenetic alterations and how gut microbiome-derived metabolites may become new therapeutics for normalizing epigenetic aberrations. Finally, we address possible challenges and future perspectives for alleviating psychotic symptoms and depression in patients with SUDs by modulating diets, the epigenome, and gut microbiome.
Collapse
Affiliation(s)
- Shabnam Nohesara
- Department of Medicine (Biomedical Genetics), Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA;
- Mental Health Research Center, Psychosocial Health Research Institute, Department of Psychiatry, School of Medicine, Iran University of Medical Sciences, Tehran 14535, Iran
| | - Hamid Mostafavi Abdolmaleky
- Department of Medicine (Biomedical Genetics), Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA;
- Nutrition/Metabolism Laboratory, Department of Surgery, BIDMC, Harvard Medical School, Boston, MA 02215, USA
| | - Sam Thiagalingam
- Department of Medicine (Biomedical Genetics), Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA;
- Department of Pathology & Laboratory Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
4
|
Vincent B, Shukla M. The Common Denominators of Parkinson's Disease Pathogenesis and Methamphetamine Abuse. Curr Neuropharmacol 2024; 22:2113-2156. [PMID: 37691228 PMCID: PMC11337683 DOI: 10.2174/1570159x21666230907151226] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 09/12/2023] Open
Abstract
The pervasiveness and mortality associated with methamphetamine abuse have doubled during the past decade, suggesting a possible worldwide substance use crisis. Epitomizing the pathophysiology and toxicology of methamphetamine abuse proclaims severe signs and symptoms of neurotoxic and neurobehavioral manifestations in both humans and animals. Most importantly, chronic use of this drug enhances the probability of developing neurodegenerative diseases manifolds. Parkinson's disease is one such neurological disorder, which significantly and evidently not only shares a number of toxic pathogenic mechanisms induced by methamphetamine exposure but is also interlinked both structurally and genetically. Methamphetamine-induced neurodegeneration involves altered dopamine homeostasis that promotes the aggregation of α-synuclein protofibrils in the dopaminergic neurons and drives these neurons to make them more vulnerable to degeneration, as recognized in Parkinson's disease. Moreover, the pathologic mechanisms such as mitochondrial dysfunction, oxidative stress, neuroinflammation and decreased neurogenesis detected in methamphetamine abusers dramatically resemble to what is observed in Parkinson's disease cases. Therefore, the present review comprehensively cumulates a holistic illustration of various genetic and molecular mechanisms putting across the notion of how methamphetamine administration and intoxication might lead to Parkinson's disease-like pathology and Parkinsonism.
Collapse
Affiliation(s)
- Bruno Vincent
- Institute of Molecular and Cellular Pharmacology, Laboratory of Excellence DistALZ, Université Côte d'Azur, INSERM, CNRS, Sophia-Antipolis, 06560, Valbonne, France
| | - Mayuri Shukla
- Chulabhorn Graduate Institute, Chulabhorn Royal Academy, 10210, Bangkok, Thailand
| |
Collapse
|
5
|
Perrine SA, Alsharif WF, Harutyunyan A, Kamal S, Viola NT, Gelovani JG. Low- and high-cocaine intake affects the spatial and temporal dynamics of class IIa HDAC expression-activity in the nucleus accumbens and hippocampus of male rats as measured by [18F]TFAHA PET/CT neuroimaging. ADDICTION NEUROSCIENCE 2022; 4:100046. [PMID: 36540409 PMCID: PMC9762729 DOI: 10.1016/j.addicn.2022.100046] [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: 06/17/2023]
Abstract
Repeated cocaine alters neuronal function in the nucleus accumbens (NAc), a brain region involved in cocaine taking, and in hippocampus (HC), known for contextual and associative learning. [18F]TFAHA is a histone deacetylase (HDAC) class IIa-specific radiotracer for positron emission tomography (PET)-imaging developed by our group to study epigenetic mechanisms. Here, [18F]TFAHA was used to conduct PET-imaging coupled with computed tomography (CT) of rat brains at baseline and after repeated cocaine intravenous self-administration (cocaine-IVSA) in low-intake versus high-intake cocaine groups. A 3 h-access FR1-schedule of cocaine-IVSA (0.5 mg/kg/infusion) for 12 continuous days was used with male Sprague Dawley rats following jugular vein catheterization. PET/CT neuroimaging with [18F]TFAHA was acquired in a dynamic mode over 40 min post-radiotracer administration at baseline and on day 12 of cocaine-IVSA using a longitudinal, repeated design. This study shows that high-cocaine intake significantly decreases class IIa HDAC expression-activity in NAc, while low-cocaine intake significantly decreases expression-activity in HC in male rats. These findings suggest the individual rats with low-cocaine intake had epigenetic changes in HC, where drug-associative changes occur. Alternatively, individuals with high-cocaine intake had robust epigenetic changes in NAc, where rewared-related behaviors originate. These findings are the first longitudinal data obtained in vivo to implicate class IIa HDACs in the persistent behavioral effects of cocaine. Furthermore, our results are consistent with published research implicating class IIa HDACs in cocaine-induced brain changes and studies suggesting a relationship between an individual's drug-taking behavior and regional pattern of epigenetic changes in the brain.
Collapse
Affiliation(s)
- Shane A. Perrine
- Psychiatry and Behavioral Neurosciences, Wayne State University, 6135 Woodward Avenue, Suite 3119, Detroit, MI, USA
- Research Services, John D. Dingell VAMC, Detroit, MI, USA
| | | | - Arman Harutyunyan
- Psychiatry and Behavioral Neurosciences, Wayne State University, 6135 Woodward Avenue, Suite 3119, Detroit, MI, USA
| | - Swatabdi Kamal
- Biomedical Engineering, Wayne State University, Detroit, MI, USA
| | - Nerissa T. Viola
- Oncology, Wayne State University, Detroit, MI, USA
- Karmanos Cancer Institute, Detroit, MI, USA
| | - Juri G. Gelovani
- Biomedical Engineering, Wayne State University, Detroit, MI, USA
- Karmanos Cancer Institute, Detroit, MI, USA
| |
Collapse
|
6
|
Wang H, Dong X, Awan MUN, Bai J. Epigenetic mechanisms involved in methamphetamine addiction. Front Pharmacol 2022; 13:984997. [PMID: 36091781 PMCID: PMC9458865 DOI: 10.3389/fphar.2022.984997] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Methamphetamine (METH) is an illicit psychostimulant that is widely abused. The molecular mechanism of METH addiction is complicated and still unknown. METH causes the release of the neurotransmitters including dopamine, glutamate, norepinephrine and serotonin, which activate various brain areas in the central nervous system. METH also induces synaptic plasticity and pathological memory enhancement. Epigenetics plays the important roles in regulating METH addiction. This review will briefly summarize the studies on epigenetics involved in METH addiction.
Collapse
|
7
|
Sex Differences in Psychostimulant Abuse: Implications for Estrogen Receptors and Histone Deacetylases. Genes (Basel) 2022; 13:genes13050892. [PMID: 35627277 PMCID: PMC9140379 DOI: 10.3390/genes13050892] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/11/2022] [Accepted: 05/13/2022] [Indexed: 02/04/2023] Open
Abstract
Substance abuse is a chronic pathological disorder that negatively affects many health and neurological processes. A growing body of literature has revealed gender differences in substance use. Compared to men, women display distinct drug-use phenotypes accompanied by recovery and rehabilitation disparities. These observations have led to the notion that sex-dependent susceptibilities exist along the progression to addiction. Within this scope, neuroadaptations following psychostimulant exposure are thought to be distinct for each sex. This review summarizes clinical findings and animal research reporting sex differences in the subjective and behavioral responses to cocaine, methamphetamine, and nicotine. This discussion is followed by an examination of epigenetic and molecular alterations implicated in the addiction process. Special consideration is given to histone deacetylases and estrogen receptor-mediated gene expression.
Collapse
|
8
|
Sivalingam K, Doke M, Khan MA, Samikkannu T. Influence of psychostimulants and opioids on epigenetic modification of class III histone deacetylase (HDAC)-sirtuins in glial cells. Sci Rep 2021; 11:21335. [PMID: 34716387 PMCID: PMC8556237 DOI: 10.1038/s41598-021-00836-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 09/24/2021] [Indexed: 01/29/2023] Open
Abstract
Substance abuse affects the central nervous system (CNS) and remains a global health problem. Psychostimulants, such as cocaine and methamphetamine (METH), and opioids affect neuronal function and lead to behavioral impairments via epigenetic modification. Epigenetic changes occur via classical pathways, especially the class III histone deacetylase (HDAC)-sirtuin (SIRT) family, that act as cellular sensors to regulate energy homeostasis and coordinate cellular responses to maintain genome integrity. However, SIRT family (1-7)-associated neurodegeneration has not been elucidated in the context of energy metabolism. The present study examined the effects of psychostimulants, such as cocaine and METH, and opioids, such as morphine, on SIRT family (1-7) [class I, II, III and IV] expression and cellular translocation-mediated dysfunction in astrocytes and microglial cells. The "nootropic" drug piracetam played a preventative role against psychostimulant- and opioid-induced SIRT (1-7) expression in astrocytes. These results indicate that cocaine, METH, and morphine affected deacetylation and cellular function, and these changes were prevented by piracetam in astrocytes.
Collapse
Affiliation(s)
- Kalaiselvi Sivalingam
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University, 1010 W Avenue B, Kingsville, TX, 78363, USA
| | - Mayur Doke
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University, 1010 W Avenue B, Kingsville, TX, 78363, USA
| | - Mansoor A Khan
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University, 1010 W Avenue B, Kingsville, TX, 78363, USA
| | - Thangavel Samikkannu
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University, 1010 W Avenue B, Kingsville, TX, 78363, USA.
| |
Collapse
|
9
|
Epigenetic Regulatory Dynamics in Models of Methamphetamine-Use Disorder. Genes (Basel) 2021; 12:genes12101614. [PMID: 34681009 PMCID: PMC8535492 DOI: 10.3390/genes12101614] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 10/08/2021] [Accepted: 10/10/2021] [Indexed: 02/07/2023] Open
Abstract
Methamphetamine (METH)-use disorder (MUD) is a very serious, potentially lethal, biopsychosocial disease. Exposure to METH 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 behavior that can remain a lifelong struggle. It is crucial to elucidate underlying mechanisms by which exposure to METH leads to molecular neuroadaptive changes at transcriptional and translational levels. Changes in gene expression are controlled by post-translational modifications via chromatin remodeling. This review article focuses on the brain-region specific combinatorial or distinct epigenetic modifications that lead to METH-induced changes in gene expression.
Collapse
|
10
|
Histone Deacetylases and Immediate Early Genes: Key Players in Psychostimulant-Induced Neuronal Plasticity. Neurotox Res 2021; 39:2134-2140. [PMID: 34581974 DOI: 10.1007/s12640-021-00420-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 07/13/2021] [Accepted: 09/17/2021] [Indexed: 11/27/2022]
Abstract
IEGs play a critical functional role of in molecular, cellular, and behavioral alterations induced by psychostimulants. IEGs appear to have specific chromatin structures that may contribute to the rapid activation of their transcription. HDAC enzymes regulate reversible acetylation of lysine residues of histones and non-histone proteins. Dysregulation of HDACs has been proposed to modulate the establishment and maintenance of aberrant transcriptional programs and behaviors associated with cognitive dysfunctions and drug addiction. In this mini-review we focus our attention on recent discoveries concerning networks of protein-protein interactions for the two classes of HDAC protein family members that are highly expressed in neurons, class I and IIa HDACs. Because dynamic histone acetylation appears to be critical to IEG expression in the brain, we discuss the role of these epigenetic regulators on IEG expression induced by cocaine and methamphetamine intake.
Collapse
|
11
|
Histone deacetylase 10, a potential epigenetic target for therapy. Biosci Rep 2021; 41:228655. [PMID: 33997894 PMCID: PMC8182986 DOI: 10.1042/bsr20210462] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 05/13/2021] [Accepted: 05/14/2021] [Indexed: 11/17/2022] Open
Abstract
Histone deacetylase (HDAC) 10, a class II family, has been implicated in various tumors and non-tumor diseases, which makes the discovery of biological functions and novel inhibitors a fundamental endeavor. In cancers, HDAC10 plays crucial roles in regulating various cellular processes through its epigenetic functions or targeting some decisive molecular or signaling pathways. It also has potential clinical utility for targeting tumors and non-tumor diseases, such as renal cell carcinoma, prostate cancer, immunoglobulin A nephropathy (IgAN), intracerebral hemorrhage, human immunodeficiency virus (HIV) infection and schizophrenia. To date, relatively few studies have investigated HDAC10-specific inhibitors. Therefore, it is important to study the biological functions of HDAC10 for the future development of specific HDAC10 inhibitors. In this review, we analyzed the biological functions, mechanisms and inhibitors of HDAC10, which makes HDAC10 an appealing therapeutic target.
Collapse
|
12
|
Epigenetics of addiction. Neurochem Int 2021; 147:105069. [PMID: 33992741 DOI: 10.1016/j.neuint.2021.105069] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 04/16/2021] [Accepted: 05/09/2021] [Indexed: 11/22/2022]
Abstract
Substance use disorders are complex biopsychosocial disorders that have substantial negative neurocognitive impact in various patient populations. These diseases involve the compulsive use of licit or illicit substances despite adverse medicolegal consequences and appear to be secondary to long-lasting epigenetic and transcriptional adaptations in brain reward and non-reward circuits. The accumulated evidence supports the notion that repeated drug use causes changes in post-translational histone modifications and in DNA methylation/hydroxymethylation processes in several brain regions. This review provides an overview of epigenetic changes reported in models of cocaine, methamphetamine, and opioid use disorders. The accumulated data suggest that future therapeutic interventions should focus on the development of epigenetic drugs against addictive diseases.
Collapse
|
13
|
Zelli S, Brancato A, Mattioli F, Pepe M, Alleva E, Carbone C, Cannizzaro C, Adriani W. A new "sudden fright paradigm" to explore the role of (epi)genetic modulations of the DAT gene in fear-induced avoidance behavior. GENES BRAIN AND BEHAVIOR 2020; 20:e12709. [PMID: 33070435 DOI: 10.1111/gbb.12709] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 10/13/2020] [Accepted: 10/14/2020] [Indexed: 12/21/2022]
Abstract
Alterations in dopamine (DA) reuptake are involved in several psychiatric disorders whose symptoms can be investigated in knock out rats for the DA transporter (DAT-KO). Recent studies evidenced the role of epigenetic DAT modulation in depressive-like behavior. Accordingly, we used heterozygous (HET) rats born from both HET parents (termed MIX-HET), compared to HET rats born from WT-mother and KO-father (MAT-HET), implementing the role of maternal care on DAT modulation. We developed a "sudden fright" paradigm (based on dark-light test) to study reaction to fearful inputs in the DAT-KO, MAT-HET, MIX-HET, and WT groups. Rats could freely explore the whole 3-chambers apparatus; then, they were gently confined in one room where they experienced the fright; finally, they could freely move again. As expected, after the fearful stimulus only MAT-HET rats showed a different behavior consisting of avoidance towards the fear-associated chamber, compared to WT rats. Furthermore, ex-vivo immuno-fluorescence reveals higher prefrontal DAT levels in MAT-HET compared to MIX-HET and WT rats. Immuno-fluorescence shows also a different histone deacetylase (HDAC) enzymes concentration. Since HDAC concentration could modulate gene expression, within MAT-HET fore brain, the enhanced expression of DAT could well impair the corticostriatal-thalamic circuit, thus causing aberrant avoidance behavior (observed only in MAT-HET rats). DAT expression seems to be linked to a simply different breeding condition, which points to a reduced care by HET dams for epigenetic regulation. This could imply significant prefronto-cortical influences onto the emotional processes: hence an excessively frightful response, even to mild stressful agents, may draw developmental trajectories toward anxious and depressed-like behavior.
Collapse
Affiliation(s)
- Silvia Zelli
- Center Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy.,Faculty of Psychology, Università Telematica Internazionale "Uninettuno", Rome, Italy
| | - Anna Brancato
- Department of Sciences for Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties "G. D'Alessandro," University of Palermo, Palermo, Italy
| | - Francesca Mattioli
- Faculty of Psychology, Università Telematica Internazionale "Uninettuno", Rome, Italy
| | - Martina Pepe
- Center Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| | - Enrico Alleva
- Center Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| | - Cristiana Carbone
- Center Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| | - Carla Cannizzaro
- Department of Sciences for Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties "G. D'Alessandro," University of Palermo, Palermo, Italy
| | - Walter Adriani
- Center Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy.,Faculty of Psychology, Università Telematica Internazionale "Uninettuno", Rome, Italy
| |
Collapse
|
14
|
Liu SS, Wu F, Jin YM, Chang WQ, Xu TM. HDAC11: a rising star in epigenetics. Biomed Pharmacother 2020; 131:110607. [PMID: 32841898 DOI: 10.1016/j.biopha.2020.110607] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/31/2020] [Accepted: 08/02/2020] [Indexed: 02/08/2023] Open
Abstract
Epigenetic mechanisms, such as acetylation, methylation, and succinylation, play pivotal roles in the regulation of multiple normal biological processes, including neuron regulation, hematopoiesis, bone cell maturation, and metabolism. In addition, epigenetic mechanisms are closely associated with the pathological processes of various diseases, such as metabolic diseases, autoimmune diseases and cancers. Epigenetic changes may precede genetic mutation, so research on epigenetic changes and regulation may be important for the early detection and diagnosis of disease. Histone deacetylase11 (HDAC11) is the newest member of the histone deacetylase (HDAC) family and the only class IV histone deacetylase. HDAC11 has different expression levels and biological functions in different systems of the human body and is among the top 1 to 4% of genes overexpressed in cancers, such as breast cancer, hepatocellular carcinoma and renal pelvis urothelial carcinoma. This article analyzes the role and mechanism of HDAC11 in disease, especially in tumorigenesis, in an attempt to provide new ideas for clinical and basic research.
Collapse
Affiliation(s)
- Shan-Shan Liu
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, 130041, Jilin Province, China.
| | - Fei Wu
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, 130041, Jilin Province, China.
| | - Yue-Mei Jin
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, 130041, Jilin Province, China.
| | - Wei-Qin Chang
- Department of Surgery, The Second Hospital of Jilin University, 218 Ziqiang Road, Changchun, 130041, Jilin Province, China.
| | - Tian-Min Xu
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, 130041, Jilin Province, China.
| |
Collapse
|
15
|
Yu Z, Wang J, Zhang P, Wang J, Cui J, Wang H. Enriched environment improves sevoflurane-induced cognitive impairment during late-pregnancy via hippocampal histone acetylation. ACTA ACUST UNITED AC 2020; 53:e9861. [PMID: 32813852 PMCID: PMC7433840 DOI: 10.1590/1414-431x20209861] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 06/08/2020] [Indexed: 12/28/2022]
Abstract
Fetal exposure to sevoflurane induces long-term cognitive impairment. Histone acetylation regulates the transcription of genes involved in memory formation. We investigated whether sevoflurane exposure during late-pregnancy induces neurocognitive impairment in offspring, and if this is related to histone acetylation dysfunction. We determined whether the effects could be reversed by an enriched environment (EE). Pregnant rats were exposed to 2.5% sevoflurane or control for 1, 3, or 6 h on gestational day 18 (G18). Sevoflurane reduced brain-derived neurotrophic factor (BDNF), acetyl histone H3 (Ac-H3), and Ac-H4 levels and increased histone deacetylases-2 (HDAC2) and HDAC3 levels in the hippocampus of the offspring on postnatal day 1 (P1) and P35. Long-term potentiation was inhibited, and spatial learning and memory were impaired in the 6-h sevoflurane group at P35. EE alleviated sevoflurane-induced cognitive dysfunction and increased hippocampal BDNF, Ac-H3, and Ac-H4. Exposure to 2.5% sevoflurane for 3 h during late-pregnancy decreased hippocampal BDNF, Ac-H3, and Ac-H4 in the offspring but had no effect on cognitive function. However, when the exposure time was 6 h, impaired spatial learning and memory were linked to reduced BDNF, Ac-H3, and Ac-H4, which could be reversed by EE.
Collapse
Affiliation(s)
- Zhiqiang Yu
- Department of Anesthesiology, Tianjin Central Hospital of Gynecology and Obstetrics, Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin, China
| | - Jinxin Wang
- Department of Anesthesiology, The Third Central Clinical College of Tianjin Medical University, Tianjin Third Central Hospital, Nankai University Affinity the Third Central Hospital, Tianjin Institute of Hepatobiliary Disease, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
| | - Peijun Zhang
- Department of Anesthesiology, Tianjin Central Hospital of Gynecology and Obstetrics, Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin, China
| | - Jianbo Wang
- Department of Anesthesiology, Tianjin Central Hospital of Gynecology and Obstetrics, Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin, China
| | - Jian Cui
- Department of Anesthesiology, Tianjin Central Hospital of Gynecology and Obstetrics, Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin, China
| | - Haiyun Wang
- Department of Anesthesiology, The Third Central Clinical College of Tianjin Medical University, Tianjin Third Central Hospital, Nankai University Affinity the Third Central Hospital, Tianjin Institute of Hepatobiliary Disease, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
| |
Collapse
|
16
|
Bernardi A, Torres OV, Sosa M, Muñiz JA, Urbano FJ, Cadet JL, Bisagno V. Acute Regulation of the Arousal-Enhancing Drugs Caffeine and Modafinil on Class IIa HDACs In Vivo and In Vitro: Focus on HDAC7. Neurotox Res 2020; 38:498-507. [PMID: 32367472 DOI: 10.1007/s12640-020-00200-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/24/2020] [Accepted: 03/31/2020] [Indexed: 01/15/2023]
Abstract
Psychostimulant drugs, such as modafinil and caffeine, induce transcriptional alterations through the dysregulation of epigenetic mechanisms. We have previously demonstrated that acute modafinil administration is accompanied by multiple changes in the expression of histone deacetylases (HDACs) within the mouse medial prefrontal cortex (mPFC). Herein, we compared alterations in class IIa HDACs in the mouse mPFC and dorsal striatum (DS) after a single exposure to each psychostimulant. We treated male C57BL/6 mice with modafinil (90 mg/kg, i.p.), caffeine (10 mg/kg, i.p.), or vehicle and evaluated locomotor activity. Following, we examined hdac4, hdac5, and hdac7 mRNA expression using qRT-PCR and HDAC7, pHDAC7, and pHDACs4/5/7 using Western blot. Last, we explored generalized effects in N2a cell line using modafinil (100 μM and 1 mM) or caffeine (80 μM and 800 μM). Our results indicate that modafinil had greater effects on locomotor activity compared with caffeine. qRT-PCR experiments revealed that modafinil decreased hdac5 and hdac7 mRNA expression in the DS, while caffeine had no effects. In the mPFC, modafinil increased hdac7 mRNA expression, with no effects observed for caffeine. Western blot revealed that within the DS, modafinil induced increases in HDAC7, pHDAC7, and pHDACs4/5/7 protein expression, while, in the mPFC, caffeine induced decreases in HDAC7, pHDAC7, and pHDACs4/5/7 protein levels. In vitro studies revealed that modafinil increased hdac4, hdac5, and hdac7 mRNA levels in N2a, while caffeine only increased hdac5 at a higher dose. These findings support the notion that modafinil and caffeine exert distinct regulation of class IIa HDAC family members and that these transcriptional and translational consequences are region-specific.
Collapse
Affiliation(s)
- Alejandra Bernardi
- Instituto de Investigaciones Farmacológicas, Universidad de Buenos Aires - Consejo Nacional de Investigaciones Científicas y Técnicas, Junín 956, piso 5, Ciudad Autónoma de Buenos Aires, C1113, Buenos Aires, Argentina
| | - Oscar V Torres
- Department of Behavioral Sciences, San Diego Mesa College, San Diego, CA, USA
| | - Maximo Sosa
- Instituto de Investigaciones Farmacológicas, Universidad de Buenos Aires - Consejo Nacional de Investigaciones Científicas y Técnicas, Junín 956, piso 5, Ciudad Autónoma de Buenos Aires, C1113, Buenos Aires, Argentina
| | - Javier A Muñiz
- Instituto de Investigaciones Farmacológicas, Universidad de Buenos Aires - Consejo Nacional de Investigaciones Científicas y Técnicas, Junín 956, piso 5, Ciudad Autónoma de Buenos Aires, C1113, Buenos Aires, Argentina
| | - Francisco J Urbano
- Laboratorio de Fisiología y Biología Molecular, Instituto de Fisiología, Biología Molecular y Neurociencias, Universidad de Buenos Aires - Consejo Nacional de Investigaciones Científicas y Técnicas, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Jean Lud Cadet
- Molecular Neuropsychiatry Research Branch, NIH/NIDA Intramural Research Program, Baltimore, MD, USA
| | - Veronica Bisagno
- Instituto de Investigaciones Farmacológicas, Universidad de Buenos Aires - Consejo Nacional de Investigaciones Científicas y Técnicas, Junín 956, piso 5, Ciudad Autónoma de Buenos Aires, C1113, Buenos Aires, Argentina.
| |
Collapse
|
17
|
Davis KC, Saito K, Rodeghiero SR, Toth BA, Lutter M, Cui H. Behavioral Alterations in Mice Carrying Homozygous HDAC4 A778T Missense Mutation Associated With Eating Disorder. Front Neurosci 2020; 14:139. [PMID: 32153359 PMCID: PMC7046559 DOI: 10.3389/fnins.2020.00139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 02/04/2020] [Indexed: 11/13/2022] Open
Abstract
Eating disorders (EDs) are serious mental illnesses thought to arise from the complex gene-environment interactions. DNA methylation patterns in histone deacetylase 4 (HDAC4) locus have been associated with EDs and we have previously identified a missense mutation in the HDAC4 gene (HDAC4A786T) that increases the risk of developing an ED. In order to evaluate the biological consequences of this variant and establish a useful mouse model of EDs, here we performed behavioral characterization of mice homozygous for Hdac4A778T (corresponding to human HDAC4A786T) that were further backcrossed onto C57BL/6 background. When fed high-fat diet, male, but not female, homozygous mice showed a trend toward decreased weight gain compared to their wild-type littermates. Behaviorally, male, but not female, homozygous mice spent less time in eating and exhibited reduced motivation to work for palatable food and light phase-specific decrease in locomotor activity. Additionally, homozygous Hdac4A778T female, but not male, mice display social subordination when subjected to a tube dominance test. Collectively, these results reveal a complex sex- and circadian-dependent role of ED-associated Hdac4A778T mutation in affecting mouse behaviors. Homozygous Hdac4A778T mice could therefore be a useful animal model to gain insight into the neurobiological basis of EDs.
Collapse
Affiliation(s)
- Kevin C Davis
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Kenji Saito
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Samuel R Rodeghiero
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Brandon A Toth
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Michael Lutter
- Eating Recovery Center of San Antonio, San Antonio, TX, United States
| | - Huxing Cui
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States.,F.O.E. Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, United States.,Obesity Research and Educational Initiative, University of Iowa Carver College of Medicine, Iowa City, IA, United States.,Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| |
Collapse
|