1
|
Wani SN, Grewal AK, Khan H, Singh TG. Elucidating the molecular symphony: unweaving the transcriptional & epigenetic pathways underlying neuroplasticity in opioid dependence and withdrawal. Psychopharmacology (Berl) 2024; 241:1955-1981. [PMID: 39254835 DOI: 10.1007/s00213-024-06684-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 09/02/2024] [Indexed: 09/11/2024]
Abstract
The persistent use of opioids leads to profound changes in neuroplasticity of the brain, contributing to the emergence and persistence of addiction. However, chronic opioid use disrupts the delicate balance of the reward system in the brain, leading to neuroadaptations that underlie addiction. Chronic cocaine usage leads to synchronized alterations in gene expression, causing modifications in the Nucleus Accumbens (NAc), a vital part of the reward system of the brain. These modifications assist in the development of maladaptive behaviors that resemble addiction. Neuroplasticity in the context of addiction involves changes in synaptic connectivity, neuronal morphology, and molecular signaling pathways. Drug-evoked neuroplasticity in opioid addiction and withdrawal represents a complicated interaction between environmental, genetic, and epigenetic factors. Identifying specific transcriptional and epigenetic targets that can be modulated to restore normal neuroplasticity without disrupting essential physiological processes is a critical consideration. The discussion in this article focuses on the transcriptional aspects of drug-evoked neuroplasticity, emphasizing the role of key transcription factors, including cAMP response element-binding protein (CREB), ΔFosB, NF-kB, Myocyte-enhancing factor 2 (MEF2), Methyl-CpG binding protein 2 (MeCP2), E2F3a, and FOXO3a. These factors regulate gene expression and lead to the neuroadaptive changes observed in addiction and withdrawal. Epigenetic regulation, which involves modifying gene accessibility by controlling these structures, has been identified as a critical component of addiction development. By unraveling these complex molecular processes, this study provides valuable insights that may pave the way for future therapeutic interventions targeting the mechanisms underlying addiction and withdrawal.
Collapse
Affiliation(s)
- Shahid Nazir Wani
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
- Aman Pharmacy College, Dholakhera, Udaipurwati, Jhunjhunu, Rajasthan, 333307, India
| | - Amarjot Kaur Grewal
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.
| | - Heena Khan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| |
Collapse
|
2
|
Pelletier OB, Brunori G, Wang Y, Robishaw JD. Post-transcriptional regulation and subcellular localization of G-protein γ7 subunit: implications for striatal function and behavioral responses to cocaine. Front Neuroanat 2024; 18:1394659. [PMID: 38764487 PMCID: PMC11100332 DOI: 10.3389/fnana.2024.1394659] [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: 03/01/2024] [Accepted: 04/17/2024] [Indexed: 05/21/2024] Open
Abstract
The striatal D1 dopamine receptor (D1R) and A2a adenosine receptor (A2aR) signaling pathways play important roles in drug-related behaviors. These receptors activate the Golf protein comprised of a specific combination of αolfβ2γ7 subunits. During assembly, the γ7 subunit sets the cellular level of the Golf protein. In turn, the amount of Golf protein determines the collective output from both D1R and A2aR signaling pathways. This study shows the Gng7 gene encodes multiple γ7 transcripts differing only in their non-coding regions. In striatum, Transcript 1 is the predominant isoform. Preferentially expressed in the neuropil, Transcript 1 is localized in dendrites where it undergoes post-transcriptional regulation mediated by regulatory elements in its 3' untranslated region that contribute to translational suppression of the γ7 protein. Earlier studies on gene-targeted mice demonstrated loss of γ7 protein disrupts assembly of the Golf protein. In the current study, morphological analysis reveals the loss of the Golf protein is associated with altered dendritic morphology of medium spiny neurons. Finally, behavioral analysis of conditional knockout mice with cell-specific deletion of the γ7 protein in distinct populations of medium spiny neurons reveals differential roles of the Golf protein in mediating behavioral responses to cocaine. Altogether, these findings provide a better understanding of the regulation of γ7 protein expression, its impact on Golf function, and point to a new potential target and mechanisms for treating addiction and related disorders.
Collapse
Affiliation(s)
- Oliver B. Pelletier
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, United States
| | - Gloria Brunori
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, United States
| | - Yingcai Wang
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, United States
| | - Janet D. Robishaw
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, United States
- Department of Comparative, Diagnostic, and Population Medicine, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| |
Collapse
|
3
|
Khalifa FN, Hussein RF, Mekawy DM, Elwi HM, Alsaeed SA, Elnawawy Y, Shaheen SH. Potential role of the lncRNA "HOTAIR"/miRNA "206"/BDNF network in the alteration in expression of synaptic plasticity gene arc and BDNF level in sera of patients with heroin use disorder through the PI3K/AKT/mTOR pathway compared to the controls. Mol Biol Rep 2024; 51:293. [PMID: 38334898 PMCID: PMC10858136 DOI: 10.1007/s11033-024-09265-3] [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: 10/07/2023] [Accepted: 01/17/2024] [Indexed: 02/10/2024]
Abstract
INTRODUCTION Heroin use disorder (HUD) is a seriously increasing health issue, accounting for most deaths among drug abusers. Studying non-coding ribonucleic acid gene expression among drug abusers is a promising approach, as it may be used in diagnosis and therapeutics. PARTICIPANTS AND METHODS A total of 49 male heroin-dependent patients and 49 male control participants were recruited from Kasr Al Ainy Psychiatry and Addiction outpatient clinics, Faculty of Medicine, Cairo University. Sera were gathered. qRT-PCR was utilized for the detection of gene expression of non-coding RNAs such as "HOX transcript antisense RNA" (HOTAIR), micro-RNA (miRNA-206), phosphatidylinositol 3-kinase (PI3K), protein kinase B (AKT), mechanistic target of rapamycin (mTOR), and Activity Regulated Cytoskeleton Associated Protein (Arc). Sera Brain-Derived Neurotrophic Factor (BDNF) levels were assessed using ELISA. Using a western blot made it possible to determine the protein expression of PI3K, AKT, and mTOR. RESULTS The study demonstrated that gene expressions of HOTAIR, AKT, PI3K, and Arc were considerably lowered between cases and controls, while gene expressions of miR-206 and mTOR1 were significantly raised. PI3K and AKT protein expressions were downregulated, while mTOR expressions were upregulated. BDNF levels were significantly decreased in some cases. CONCLUSION The results of this study suggest that decreased HOTAIR in HUD relieves miR-206 inhibition, which thus increases and affects downstream PI3K/AKT/mTOR, ARC, and BDNF expression. This may be shared in addictive and relapsing behaviors.
Collapse
Affiliation(s)
- Fatma Nada Khalifa
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Cairo University, Kasr Alainy Street, Cairo, 11562, Egypt
| | - Riham F Hussein
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Cairo University, Kasr Alainy Street, Cairo, 11562, Egypt
| | - Dina M Mekawy
- Department of Biochemistry, Faculty of Medicine, Cairo University, Kasr Alainy Street, Cairo, 11562, Egypt
| | - Heba M Elwi
- Department of Biochemistry, Faculty of Medicine, Cairo University, Kasr Alainy Street, Cairo, 11562, Egypt
| | - Shimaa Ahmed Alsaeed
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Cairo University, Kasr Alainy Street, Cairo, 11562, Egypt.
| | - Yassmin Elnawawy
- Department of Psychiatry, Faculty of Medicine, Cairo University, Kasr Alainy Street, Cairo, 11562, Egypt
| | - Somaya H Shaheen
- Department of Psychiatry, Faculty of Medicine, Cairo University, Kasr Alainy Street, Cairo, 11562, Egypt
| |
Collapse
|
4
|
Occhipinti C, La Russa R, Iacoponi N, Lazzari J, Costantino A, Di Fazio N, Del Duca F, Maiese A, Fineschi V. miRNAs and Substances Abuse: Clinical and Forensic Pathological Implications: A Systematic Review. Int J Mol Sci 2023; 24:17122. [PMID: 38069445 PMCID: PMC10707252 DOI: 10.3390/ijms242317122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/17/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
Substance addiction is a chronic and relapsing brain disorder characterized by compulsive seeking and continued substance use, despite adverse consequences. The high prevalence and social burden of addiction are indisputable; however, the available intervention is insufficient. The modulation of gene expression and aberrant adaptation of neural networks are attributed to the changes in brain functions under repeated exposure to addictive substances. Considerable studies have demonstrated that miRNAs are strong modulators of post-transcriptional gene expression in substance addiction. The emerging role of microRNA (miRNA) provides new insights into many biological and pathological processes in the central nervous system: their variable expression in different regions of the brain and tissues may play a key role in regulating the pathophysiological events of addiction. This work provides an overview of the current literature on miRNAs involved in addiction, evaluating their impaired expression and regulatory role in neuroadaptation and synaptic plasticity. Clinical implications of such modulatory capacities will be estimated. Specifically, it will evaluate the potential diagnostic role of miRNAs in the various stages of drug and substance addiction. Future perspectives about miRNAs as potential novel therapeutic targets for substance addiction and abuse will also be provided.
Collapse
Affiliation(s)
- Carla Occhipinti
- Department of Surgical Pathology, Medical, Molecular and Critical Area, Institute of Legal Medicine, University of Pisa, 56126 Pisa, Italy; (C.O.); (N.I.); (J.L.); (A.C.)
| | - Raffaele La Russa
- Department of Clinical Medicine, Public Health, Life Sciences, and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy;
| | - Naomi Iacoponi
- Department of Surgical Pathology, Medical, Molecular and Critical Area, Institute of Legal Medicine, University of Pisa, 56126 Pisa, Italy; (C.O.); (N.I.); (J.L.); (A.C.)
| | - Julia Lazzari
- Department of Surgical Pathology, Medical, Molecular and Critical Area, Institute of Legal Medicine, University of Pisa, 56126 Pisa, Italy; (C.O.); (N.I.); (J.L.); (A.C.)
| | - Andrea Costantino
- Department of Surgical Pathology, Medical, Molecular and Critical Area, Institute of Legal Medicine, University of Pisa, 56126 Pisa, Italy; (C.O.); (N.I.); (J.L.); (A.C.)
| | - Nicola Di Fazio
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Viale Regina Elena 336, 00161 Rome, Italy; (N.D.F.); (F.D.D.); (V.F.)
| | - Fabio Del Duca
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Viale Regina Elena 336, 00161 Rome, Italy; (N.D.F.); (F.D.D.); (V.F.)
| | - Aniello Maiese
- Department of Surgical Pathology, Medical, Molecular and Critical Area, Institute of Legal Medicine, University of Pisa, 56126 Pisa, Italy; (C.O.); (N.I.); (J.L.); (A.C.)
| | - Vittorio Fineschi
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Viale Regina Elena 336, 00161 Rome, Italy; (N.D.F.); (F.D.D.); (V.F.)
| |
Collapse
|
5
|
Rezayof A, Ghasemzadeh Z, Sahafi OH. Addictive drugs modify neurogenesis, synaptogenesis and synaptic plasticity to impair memory formation through neurotransmitter imbalances and signaling dysfunction. Neurochem Int 2023; 169:105572. [PMID: 37423274 DOI: 10.1016/j.neuint.2023.105572] [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: 04/19/2023] [Revised: 07/01/2023] [Accepted: 07/05/2023] [Indexed: 07/11/2023]
Abstract
Drug abuse changes neurophysiological functions at multiple cellular and molecular levels in the addicted brain. Well-supported scientific evidence suggests that drugs negatively affect memory formation, decision-making and inhibition, and emotional and cognitive behaviors. The mesocorticolimbic brain regions are involved in reward-related learning and habitual drug-seeking/taking behaviors to develop physiological and psychological dependence on the drugs. This review highlights the importance of specific drug-induced chemical imbalances resulting in memory impairment through various neurotransmitter receptor-mediated signaling pathways. The mesocorticolimbic modifications in the expression levels of brain-derived neurotrophic factor (BDNF) and the cAMP-response element binding protein (CREB) impair reward-related memory formation following drug abuse. The contributions of protein kinases and microRNAs (miRNAs), along with the transcriptional and epigenetic regulation have also been considered in memory impairment underlying drug addiction. Overall, we integrate the research on various types of drug-induced memory impairment in distinguished brain regions and provide a comprehensive review with clinical implications addressing the upcoming studies.
Collapse
Affiliation(s)
- Ameneh Rezayof
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran.
| | - Zahra Ghasemzadeh
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Oveis Hosseinzadeh Sahafi
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| |
Collapse
|
6
|
Zanda MT, Floris G, Daws SE. Orbitofrontal cortex microRNAs support long-lasting heroin seeking behavior in male rats. Transl Psychiatry 2023; 13:117. [PMID: 37031193 PMCID: PMC10082780 DOI: 10.1038/s41398-023-02423-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 03/23/2023] [Accepted: 03/29/2023] [Indexed: 04/10/2023] Open
Abstract
Recovery from opioid use disorder (OUD) and maintenance of abstinence from opioid use is hampered by perseverant drug cravings that may persist for months after cessation of drug use. Drug cravings can intensify during the abstinence period, a phenomenon referred to as the 'incubation of craving' that has been well-described in preclinical studies. We previously reported that animals that self-administered heroin at a dosage of 0.075 mg/kg/infusion (HH) paired with discrete drug cues displayed robust incubation of heroin craving behavior after 21 days (D) of forced abstinence, an effect that was not observed with a lower dosage (0.03 mg/kg/infusion; HL). Here, we sought to elucidate molecular mechanisms underlying long-term heroin seeking behavior by profiling microRNA (miRNA) pathways in the orbitofrontal cortex (OFC), a brain region that modulates incubation of heroin seeking. miRNAs are small noncoding RNAs with long half-lives that have emerged as critical regulators of drug seeking behavior but their expression in the OFC has not been examined in any drug exposure paradigm. We employed next generation sequencing to detect OFC miRNAs differentially expressed after 21D of forced abstinence between HH and HL animals, and proteomics analysis to elucidate miRNA-dependent translational neuroadaptations. We identified 55 OFC miRNAs associated with incubation of heroin craving, including miR-485-5p, which was significantly downregulated following 21D forced abstinence in HH but not HL animals. We bidirectionally manipulated miR-485-5p in the OFC to demonstrate that miR-485-5p can regulate long-lasting heroin seeking behavior after extended forced abstinence. Proteomics analysis identified 45 proteins selectively regulated in the OFC of HH but not HL animals that underwent 21D forced abstinence, of which 7 were putative miR-485-5p target genes. Thus, the miR-485-5p pathway is dysregulated in animals with a phenotype of persistent heroin craving behavior and OFC miR-485-5p pathways may function to support long-lasting heroin seeking.
Collapse
Affiliation(s)
- Mary Tresa Zanda
- Center for Substance Abuse Research, Temple University, Philadelphia, PA, USA
- Department of Neural Sciences, Temple University, Philadelphia, PA, USA
| | - Gabriele Floris
- Center for Substance Abuse Research, Temple University, Philadelphia, PA, USA
- Department of Neural Sciences, Temple University, Philadelphia, PA, USA
| | - Stephanie E Daws
- Center for Substance Abuse Research, Temple University, Philadelphia, PA, USA.
- Department of Neural Sciences, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
7
|
Dykxhoorn DM, Wang H, Da Fonseca Ferreira A, Wei J, Dong C. MicroRNA-423-5p Mediates Cocaine-Induced Smooth Muscle Cell Contraction by Targeting Cacna2d2. Int J Mol Sci 2023; 24:6584. [PMID: 37047559 PMCID: PMC10094933 DOI: 10.3390/ijms24076584] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/23/2023] [Accepted: 03/27/2023] [Indexed: 04/05/2023] Open
Abstract
Cocaine abuse increases the risk of atherosclerotic cardiovascular disease (CVD) and causes acute coronary syndromes (ACS) and hypertension (HTN). Significant research has explored the role of the sympathetic nervous system mediating the cocaine effects on the cardiovascular (CV) system. However, the response of the sympathetic nervous system alone is insufficient to completely account for the CV consequences seen in cocaine users. In this study, we examined the role of microRNAs (miRNAs) in mediating the effect of cocaine on the CV system. MiRNAs regulate many important biological processes and have been associated with both response to cocaine and CV disease development. Multiple miRNAs have altered expression in the CV system (CVS) upon cocaine exposure. To understand the molecular mechanisms underlying the cocaine response in the CV system, we studied the role of miRNA-423-5p and its target Cacna2d2 in the regulation of intracellular calcium concentration and SMC contractility, a critical factor in the modulation of blood pressure (BP). We used in vivo models to evaluate BP and aortic stiffness. In vitro, cocaine treatment decreased miR-423-5p expression and increased Cacna2d2 expression, which led to elevated intracellular calcium concentrations and increased SMC contractility. Overexpression of miR-423-5p, silencing of its target Cacna2d2, and treatment with a calcium channel blocker reversed the elevated SMC contractility caused by cocaine. In contrast, suppression of miR-423-5p increased the intracellular calcium concentration and SMC contractibility. In vivo, smooth muscle-specific overexpression of miR-423-5p ameliorated the increase in BP and aortic stiffness associated with cocaine use. Thus, miR-423-5p regulates SMC contraction by modulating Cacna2d2 expression increasing intracellular calcium concentrations. Modulation of the miR-423-5p-Cacna2d2-Calcium transport pathway may represent a novel therapeutic strategy to improve cocaine-induced HTN and aortic stiffness.
Collapse
Affiliation(s)
- Derek M. Dykxhoorn
- Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Huilan Wang
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Andrea Da Fonseca Ferreira
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Jianqin Wei
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Chunming Dong
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Section of Cardiology, Miami VA Health Systems, Miami, FL 33136, USA
- Biomedical Research Building, Suite 812, 1501 NW 10th Avenue, Miami, FL 33136, USA
| |
Collapse
|
8
|
Bio-inspired Active Learning method in spiking neural network. Knowl Based Syst 2023. [DOI: 10.1016/j.knosys.2022.110193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
9
|
Li L, Zhan Y, Xia H, Wu Y, Wu X, Chen S. Sevoflurane protects against intracerebral hemorrhage via microRNA-133b/FOXO4/BCL2 axis. Int Immunopharmacol 2023; 114:109453. [PMID: 36476488 DOI: 10.1016/j.intimp.2022.109453] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 11/03/2022] [Accepted: 11/10/2022] [Indexed: 12/12/2022]
Abstract
The application of Sevoflurane (Sev) in neurological diseases has been documented. We herein clarified the role of Sev in intracerebral hemorrhage (ICH). Through bioinformatics analysis, ICH-related microRNA (miRNA) was collected with microRNA-133b (miR-133b) chosen for the study subject. Then, the related downstream gene Forkhead box O4 (FOXO4) was identified. For in vivo assays, an ICH mouse model was established by autologous blood injection. For in vitro assays, hippocampal neurons were extracted from mouse brain tissues, and erythrocyte lysates were employed to simulate in vitro hemorrhage. Interaction between miR-133b and FOXO4 as well as between FOXO4 and BCL2 were assayed. We found decreased miR-133b in the brain tissue of ICH mice and erythrocyte lysate-treated hippocampal neurons. Sev treatment attenuated ICH and hippocampal neuronal apoptosis in mice by upregulating miR-133b. miR-133b targeted FOXO4 expression, and inhibition of FOXO4 attenuated hippocampal neuronal apoptosis by increasing BCL2 expression. Sev attenuated ICH in mice by increasing BCL2 expression through regulation of miR-133b-mediated FOXO4 expression. The findings highlighted the protective effect of Sev on ICH mice through the regulation of miR-133b-mediated FOXO4 expression.
Collapse
Affiliation(s)
- Lei Li
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, PR China
| | - Yanping Zhan
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, PR China
| | - Haimei Xia
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, PR China
| | - Yunkun Wu
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, PR China
| | - Xiongzhi Wu
- Nanchang University, Nanchang 330006, PR China
| | - Shibiao Chen
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, PR China.
| |
Collapse
|
10
|
Vasiliev GV, Ovchinnikov VY, Lisachev PD, Bondar NP, Grinkevich LN. The Expression of miRNAs Involved in Long-Term Memory Formation in the CNS of the Mollusk Helix lucorum. Int J Mol Sci 2022; 24:ijms24010301. [PMID: 36613744 PMCID: PMC9820140 DOI: 10.3390/ijms24010301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/15/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
Mollusks are unique animals with a relatively simple central nervous system (CNS) containing giant neurons with identified functions. With such simple CNS, mollusks yet display sufficiently complex behavior, thus ideal for various studies of behavioral processes, including long-term memory (LTM) formation. For our research, we use the formation of the fear avoidance reflex in the terrestrial mollusk Helix lucorum as a learning model. We have shown previously that LTM formation in Helix requires epigenetic modifications of histones leading to both activation and inactivation of the specific genes. It is known that microRNAs (miRNAs) negatively regulate the expression of genes; however, the role of miRNAs in behavioral regulation has been poorly investigated. Currently, there is no miRNAs sequencing data being published on Helix lucorum, which makes it impossible to investigate the role of miRNAs in the memory formation of this mollusk. In this study, we have performed sequencing and comparative bioinformatics analysis of the miRNAs from the CNS of Helix lucorum. We have identified 95 different microRNAs, including microRNAs belonging to the MIR-9, MIR-10, MIR-22, MIR-124, MIR-137, and MIR-153 families, known to be involved in various CNS processes of vertebrates and other species, particularly, in the fear behavior and LTM. We have shown that in the CNS of Helix lucorum MIR-10 family (26 miRNAs) is the most representative one, including Hlu-Mir-10-S5-5p and Hlu-Mir-10-S9-5p as top hits. Moreover, we have shown the involvement of the MIR-10 family in LTM formation in Helix. The expression of 17 representatives of MIR-10 differentially changes during different periods of LTM consolidation in the CNS of Helix. In addition, using comparative analysis of microRNA expression upon learning in normal snails and snails with deficient learning abilities with dysfunction of the serotonergic system, we identified a number of microRNAs from several families, including MIR-10, which expression changes only in normal animals. The obtained data can be used for further fundamental and applied behavioral research.
Collapse
Affiliation(s)
- Gennady V. Vasiliev
- The Federal Research Center Institute of Cytology and Genetics, Siberian Branch, Russian Academy of Sciences, 10 Lavrentiev Avenue, Novosibirsk 630090, Russia
| | - Vladimir Y. Ovchinnikov
- The Federal Research Center Institute of Cytology and Genetics, Siberian Branch, Russian Academy of Sciences, 10 Lavrentiev Avenue, Novosibirsk 630090, Russia
| | - Pavel D. Lisachev
- Federal Research Center for Information and Computational Technologies, 6 Lavrentiev Avenue, Novosibirsk 630090, Russia
| | - Natalia P. Bondar
- The Federal Research Center Institute of Cytology and Genetics, Siberian Branch, Russian Academy of Sciences, 10 Lavrentiev Avenue, Novosibirsk 630090, Russia
| | - Larisa N. Grinkevich
- The Federal State Budget Scientific Institution Pavlov Institute of Physiology, Russian Academy of Sciences, 6 nab. Makarova, St. Petersburg 199034, Russia
- Correspondence:
| |
Collapse
|
11
|
Abdolahi S, Zare-Chahoki A, Noorbakhsh F, Gorji A. A Review of Molecular Interplay between Neurotrophins and miRNAs in Neuropsychological Disorders. Mol Neurobiol 2022; 59:6260-6280. [PMID: 35916975 PMCID: PMC9463196 DOI: 10.1007/s12035-022-02966-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 07/17/2022] [Indexed: 01/10/2023]
Abstract
Various neurotrophins (NTs), including nerve growth factor, brain-derived neurotrophic factor, neurotrophin-3, and neurotrophin-4, promote cellular differentiation, survival, and maintenance, as well as synaptic plasticity, in the peripheral and central nervous system. The function of microRNAs (miRNAs) and other small non-coding RNAs, as regulators of gene expression, is pivotal for the appropriate control of cell growth and differentiation. There are positive and negative loops between NTs and miRNAs, which exert modulatory effects on different signaling pathways. The interplay between NTs and miRNAs plays a crucial role in the regulation of several physiological and pathological brain procedures. Emerging evidence suggests the diagnostic and therapeutic roles of the interactions between NTs and miRNAs in several neuropsychological disorders, including epilepsy, multiple sclerosis, Alzheimer's disease, Huntington's disease, amyotrophic lateral sclerosis, schizophrenia, anxiety disorders, depression, post-traumatic stress disorder, bipolar disorder, and drug abuse. Here, we review current data regarding the regulatory interactions between NTs and miRNAs in neuropsychological disorders, for which novel diagnostic and/or therapeutic strategies are emerging. Targeting NTs-miRNAs interactions for diagnostic or therapeutic approaches needs to be validated by future clinical studies.
Collapse
Affiliation(s)
- Sara Abdolahi
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | - Ameneh Zare-Chahoki
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Farshid Noorbakhsh
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Gorji
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Neurosurgery, Westfälische Wilhelms-Universität, Münster, Germany.
- Department of Neurology and Institute for Translational Neurology, Westfälische Wilhelms-Universität, Münster, Germany.
- Epilepsy Research Center, Westfälische Wilhelms-Universität, 48149, Münster, Germany.
| |
Collapse
|
12
|
Mohammadi AH, Seyedmoalemi S, Moghanlou M, Akhlagh SA, Talaei Zavareh SA, Hamblin MR, Jafari A, Mirzaei H. MicroRNAs and Synaptic Plasticity: From Their Molecular Roles to Response to Therapy. Mol Neurobiol 2022; 59:5084-5102. [PMID: 35666404 DOI: 10.1007/s12035-022-02907-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 05/25/2022] [Indexed: 12/24/2022]
Abstract
Synaptic plasticity is the ability of synapses to weaken or strengthen over time, in response to changes in the activity of the neurons. It is orchestrated by a variety of genes, proteins, and external and internal factors, especially epigenetic factors. MicroRNAs (miRNAs) are well-acknowledged epigenetic modulators that regulate the translation and degradation of target genes in the nervous system. Increasing evidence has suggested that a number of miRNAs play important roles in modulating various aspects of synaptic plasticity. The deregulation of miRNAs could be associated with pathological alterations in synaptic plasticity, which could lead to different CNS-related diseases. Herein, we provide an update on the role of miRNAs in governing synaptic plasticity. In addition, we also summarize recent researches on the role of miRNAs in drug addiction, and their targets and mechanism of action. Understanding of the way in which miRNAs contribute to synaptic plasticity provides rational clues in establishing the novel biomarkers and new therapeutic strategies for the diagnosis and treatment of plasticity-related diseases and drug addiction.
Collapse
Affiliation(s)
- Amir Hossein Mohammadi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Seyedvahid Seyedmoalemi
- Behavioral Sciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Student Research Committee, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahsa Moghanlou
- Department of Psychiatry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, 2028, South Africa
| | - Ameneh Jafari
- Advanced Therapy Medicinal Product (ATMP) Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran.
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
13
|
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]
|
14
|
Epigenetic Studies for Evaluation of NPS Toxicity: Focus on Synthetic Cannabinoids and Cathinones. Biomedicines 2022; 10:biomedicines10061398. [PMID: 35740419 PMCID: PMC9219842 DOI: 10.3390/biomedicines10061398] [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: 05/09/2022] [Revised: 06/02/2022] [Accepted: 06/06/2022] [Indexed: 11/26/2022] Open
Abstract
In the recent decade, numerous new psychoactive substances (NPSs) have been added to the illicit drug market. These are synthetized to mimic the effects of classic drugs of abuse (i.e., cannabis, cocaine, etc.), with the purpose of bypassing substance legislations and increasing the pharmacotoxicological effects. To date, research into the acute pharmacological effects of new NPSs is ongoing and necessary in order to provide an appropriate contribution to public health. In fact, multiple examples of NPS-related acute intoxication and mortality have been recorded in the literature. Accordingly, several in vitro and in vivo studies have investigated the pharmacotoxicological profiles of these compounds, revealing that they can cause adverse effects involving various organ systems (i.e., cardiovascular, respiratory effects) and highlighting their potential increased consumption risks. In this sense, NPSs should be regarded as a complex issue that requires continuous monitoring. Moreover, knowledge of long-term NPS effects is lacking. Because genetic and environmental variables may impact NPS responses, epigenetics may aid in understanding the processes behind the harmful events induced by long-term NPS usage. Taken together, “pharmacoepigenomics” may provide a new field of combined study on genetic differences and epigenetic changes in drug reactions that might be predictive in forensic implications.
Collapse
|
15
|
Kaplan G, Xu H, Abreu K, Feng J. DNA Epigenetics in Addiction Susceptibility. Front Genet 2022; 13:806685. [PMID: 35145550 PMCID: PMC8821887 DOI: 10.3389/fgene.2022.806685] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 01/06/2022] [Indexed: 12/22/2022] Open
Abstract
Addiction is a chronically relapsing neuropsychiatric disease that occurs in some, but not all, individuals who use substances of abuse. Relatively little is known about the mechanisms which contribute to individual differences in susceptibility to addiction. Neural gene expression regulation underlies the pathogenesis of addiction, which is mediated by epigenetic mechanisms, such as DNA modifications. A growing body of work has demonstrated distinct DNA epigenetic signatures in brain reward regions that may be associated with addiction susceptibility. Furthermore, factors that influence addiction susceptibility are also known to have a DNA epigenetic basis. In the present review, we discuss the notion that addiction susceptibility has an underlying DNA epigenetic basis. We focus on major phenotypes of addiction susceptibility and review evidence of cell type-specific, time dependent, and sex biased effects of drug use. We highlight the role of DNA epigenetics in these diverse processes and propose its contribution to addiction susceptibility differences. Given the prevalence and lack of effective treatments for addiction, elucidating the DNA epigenetic mechanism of addiction vulnerability may represent an expeditious approach to relieving the addiction disease burden.
Collapse
|
16
|
Xie B, Zhang J, Ma C, Yu H, Ni Z, Cong B, Wen D. Roles of miR-592-3p and Its Target Gene, TMEFF1, in the Nucleus Accumbens During Incubation of Morphine Craving. Int J Neuropsychopharmacol 2022; 25:412-424. [PMID: 35020881 PMCID: PMC9154238 DOI: 10.1093/ijnp/pyac004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 01/03/2022] [Accepted: 01/07/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Prolonged forced abstinence from morphine can increase cue-induced cravings for the drug, contributing to a persistent vulnerability to relapse. Previous studies have identified the implications of aberrant microRNA (miRNA) regulation in the pathogenesis of morphine addiction, but the changes in miRNA expression during the incubation of morphine craving are still unknown. METHODS Nucleus accumbens (NAc)-specific altered miRNA transcriptomics was determined in a mouse model of cue-induced incubation of morphine craving following a next-generation sequencing method and verified by RT-qPCR. Bioinformatics analysis was performed to predict the target gene of selected miRNA, and the protein expression of the target gene was detected by western blot. A dual-luciferase assay was performed to confirm the binding sites, and gain- and loss-of-function strategy was applied to understand the mechanism of miRNA and its target gene. RESULTS The miR-592-3p observed to be downregulated in the NAc core was linked to the incubation of morphine craving, and a dual-luciferase assay was performed to confirm the binding sites of miR-592-3p in its target gene, tomoregulin-1 (TMEFF1). Also, gain- and loss-of-function analyses revealed that the inhibition of miR-592-3p expression in the NAc core negatively regulated TMEFF1 expression, thereby enhancing the incubation of morphine craving; however, the overexpression of miR-592-3p in the NAc core resulted in a decreased expression of TMEFF1, thereby reducing the incubation of morphine craving. CONCLUSION Our findings demonstrated that miR-592-3p can improve the incubation of morphine craving by targeting TMEFF1, and thus, it holds a therapeutic potential to inhibit opioid craving.
Collapse
Affiliation(s)
| | | | - Chunling Ma
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Hebei Province, Shijiazhuang, PR China
| | - Hailei Yu
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Hebei Province, Shijiazhuang, PR China
| | - Zhiyu Ni
- School of Basic Medical Science, Hebei University, Hebei Province, Baoding, PR China
| | - Bin Cong
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Hebei Province, Shijiazhuang, PR China
| | - Di Wen
- Correspondence: Di Wen, PhD, College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Province, Shijiazhuang 050017, PR China; Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences Hebei Province, Shijiazhuang 050017, PR China ()
| |
Collapse
|
17
|
Zhou Q, Zhang L. MicroRNA-183-5p protects human derived cell line SH-SY5Y cells from mepivacaine-induced injury. Bioengineered 2021; 12:3177-3187. [PMID: 34180760 PMCID: PMC8806725 DOI: 10.1080/21655979.2021.1946358] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
With the gradual recognition of the side effects of local anesthetics, the nerve injury caused by local anesthetics has received growing attention. This research intended to delve into miR-183-5p changes in mepivacaine-mediated SH-SY5Y cell injury, as well as its modulatory mechanism on cell apoptosis. RT-qPCR was adopted for assaying miR-183-5p and PDCD4 mRNA expression. Our team respectively transfected miR-183-5p mimic and inhibitor to enhance or inhibit miR-183-5p function. We employed Western blot for detecting PDCD4 protein levels, as well as flow cytometry and Hoechst 33342/PI double staining for determining cell apoptosis rate. Additionally, our crew applied an ELISA kit for measuring TNF-α, IL-1β, IL-6, and IL-8 contents. The level of reactive oxygen species (ROS) production was examined by the Image-iT LIVE Green ROS detection Kit. As well as dual-luciferase reporter experiment for verifying the targeting link of miR-183-5p with PDCD4. In mepivacaine-induced cell apoptosis in SH-SY5Y cells, miR-183-5p expression was down-regulated. TNF-α, IL-1β, IL-6, and IL-8 contents were elevated. The rate of apoptosis increased visibly, cleaved caspase-3 and Bax levels waxed, whereas Bcl-2 level waned. MiR-183-5p could alleviate the damaging impact of mepivacaine. Dual-luciferase reporter experiments demonstrated that miR-183-5p directly targeted PDCD4. Collectively, we concluded that a high concentration of mepivacaine can cause SH-SY5Y cell damage, miR-183-5p functions crucially in mepivacaine-mediated cell damage. This study provides a theoretical basis for elucidating the mechanism of mepivacaine-induced nerve cell damage, and overexpressed miR-183-5p likely become a novel strategy to combat mepivacaine-induced nerve damage.Abbreviations:miRNA: Micro RNA; PDCD4: Programmed Cell Death 4; MDA: Malondialdehyde; SOD: Superoxide Dismutase; ROS: Reactive Oxygen Species; WT: Wild Type; Mut: Mutant; UTR: Untranslated Region; IL-6: Interleukin-6; IL-1β: Interleukin-1β; TNF-α: Tumor Necrosis Factor-α; IL-8: Interleukin-8; COX-2: Cyclooxygenase-2; iNOS: inducible NOS; MEP: Mepivacaine.
Collapse
Affiliation(s)
- Qian Zhou
- Department of Anesthesiology, Jingzhou Central Hospital, Jinzhou, Hubei, China
| | - Ling Zhang
- Department of Anesthesiology, Jingzhou Central Hospital, Jinzhou, Hubei, China
| |
Collapse
|
18
|
Kazemi T, Huang S, Avci NG, Akay YM, Akay M. Investigating the effects of chronic perinatal alcohol and combined nicotine and alcohol exposure on dopaminergic and non-dopaminergic neurons in the VTA. Sci Rep 2021; 11:8706. [PMID: 33888815 PMCID: PMC8062589 DOI: 10.1038/s41598-021-88221-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 04/06/2021] [Indexed: 02/02/2023] Open
Abstract
The ventral tegmental area (VTA) is the origin of dopaminergic neurons and the dopamine (DA) reward pathway. This pathway has been widely studied in addiction and drug reinforcement studies and is believed to be the central processing component of the reward circuit. In this study, we used a well-established rat model to expose mother dams to alcohol, nicotine-alcohol, and saline perinatally. DA and non-DA neurons collected from the VTA of the rat pups were used to study expression profiles of miRNAs and mRNAs. miRNA pathway interactions, putative miRNA-mRNA target pairs, and downstream modulated biological pathways were analyzed. In the DA neurons, 4607 genes were differentially upregulated and 4682 were differentially downregulated following nicotine-alcohol exposure. However, in the non-DA neurons, only 543 genes were differentially upregulated and 506 were differentially downregulated. Cell proliferation, differentiation, and survival pathways were enriched after the treatments. Specifically, in the PI3K/AKT signaling pathway, there were 41 miRNAs and 136 mRNAs differentially expressed in the DA neurons while only 16 miRNAs and 20 mRNAs were differentially expressed in the non-DA neurons after the nicotine-alcohol exposure. These results depicted that chronic nicotine and alcohol exposures during pregnancy differentially affect both miRNA and gene expression profiles more in DA than the non-DA neurons in the VTA. Understanding how the expression signatures representing specific neuronal subpopulations become enriched in the VTA after addictive substance administration helps us to identify how neuronal functions may be altered in the brain.
Collapse
Affiliation(s)
- Tina Kazemi
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Shuyan Huang
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Naze G Avci
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Yasemin M Akay
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Metin Akay
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA.
| |
Collapse
|
19
|
Liu H, Xu W, Feng J, Ma H, Zhang J, Xie X, Zhuang D, Shen W, Liu H, Zhou W. Increased Expression of Plasma miRNA-320a and let-7b-5p in Heroin-Dependent Patients and Its Clinical Significance. Front Psychiatry 2021; 12:679206. [PMID: 34267687 PMCID: PMC8275879 DOI: 10.3389/fpsyt.2021.679206] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 06/04/2021] [Indexed: 12/23/2022] Open
Abstract
Heroin use disorder is a chronic and relapsing disease that induces persistent changes in the brain. The diagnoses of heroin use disorders are mainly based on subjective reports and no valid biomarkers available. Recent researches have revealed that circulating miRNAs are useful non-invasive biomarkers for diagnosing brain diseases such as Alzheimer's disease, multiple sclerosis, schizophrenia, and bipolar disorder. However, studies on circulating miRNAs for the diagnosis of heroin use disorders are rarely reported. In this study, we investigated the differential expression of plasma miRNAs in 57 heroin-dependent patients. Based on literature research and microarray analysis, two candidate miRNAs, miR-320a and let-7b-5p, were selected and analyzed by quantitative real-time RT-PCR. The results showed miR-320a and let-7b were significantly upregulated in plasma of the heroin-dependent patients compared to that in healthy controls. The area under curves (AUCs) of receiver operating characteristic (ROC) curves of miR-320a and let-7b-5p were 0.748 and 0.758, respectively. The sensitivities of miR-320a and let-7b-5p were 71.9 and 70.2%, while the specificities of miR-320a and let-7b-5p were 76.1 and 78.3%, respectively. The combination of these two miRNAs predicted heron dependence with an AUC of 0.782 (95% CI 0.687-0.876), with 73.7% sensitivity and 82.6% specificity. Our findings suggest a potential use for circulating miRNAs as biomarkers for the diagnosis of heroin abuse.
Collapse
Affiliation(s)
- Haixiong Liu
- Laboratory of Behavioral Neuroscience, Key Laboratory of Addiction Research of Zhejiang Province, School of Medicine, Ningbo Institute of Microcirculation and Henbane, Ningbo Kangning Hospital, Ningbo University, Ningbo, China
| | - Wenjin Xu
- Molecular Diagnostic Laboratory, Ningbo Institute of Medical Science, The Affiliated Hospital of School of Medicine, Ningbo University, Ningbo, China
| | - Jiying Feng
- Molecular Diagnostic Laboratory, Ningbo Institute of Medical Science, The Affiliated Hospital of School of Medicine, Ningbo University, Ningbo, China
| | - Hong Ma
- Department of Psychiatry, Ningbo Kangning Hospital, Ningbo, China
| | - Jianbin Zhang
- Molecular Diagnostic Laboratory, Ningbo Institute of Medical Science, The Affiliated Hospital of School of Medicine, Ningbo University, Ningbo, China
| | - Xiaohu Xie
- Molecular Diagnostic Laboratory, Ningbo Institute of Medical Science, The Affiliated Hospital of School of Medicine, Ningbo University, Ningbo, China
| | - Dingding Zhuang
- Molecular Diagnostic Laboratory, Ningbo Institute of Medical Science, The Affiliated Hospital of School of Medicine, Ningbo University, Ningbo, China
| | - Wenwen Shen
- Molecular Diagnostic Laboratory, Ningbo Institute of Medical Science, The Affiliated Hospital of School of Medicine, Ningbo University, Ningbo, China
| | - Huifen Liu
- Molecular Diagnostic Laboratory, Ningbo Institute of Medical Science, The Affiliated Hospital of School of Medicine, Ningbo University, Ningbo, China
| | - Wenhua Zhou
- Molecular Diagnostic Laboratory, Ningbo Institute of Medical Science, The Affiliated Hospital of School of Medicine, Ningbo University, Ningbo, China
| |
Collapse
|
20
|
Chavoshi H, Boroujeni ME, Abdollahifar MA, Amini A, Tehrani AM, Moghaddam MH, Norozian M, Farahani RM, Aliaghaei A. From dysregulated microRNAs to structural alterations in the striatal region of METH-injected rats. J Chem Neuroanat 2020; 109:101854. [DOI: 10.1016/j.jchemneu.2020.101854] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 08/05/2020] [Accepted: 08/09/2020] [Indexed: 02/07/2023]
|
21
|
Kazemi T, Huang S, Avci NG, Waits CMK, Akay YM, Akay M. Investigating the influence of perinatal nicotine and alcohol exposure on the genetic profiles of dopaminergic neurons in the VTA using miRNA-mRNA analysis. Sci Rep 2020; 10:15016. [PMID: 32929144 PMCID: PMC7490691 DOI: 10.1038/s41598-020-71875-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 08/04/2020] [Indexed: 12/11/2022] Open
Abstract
Nicotine and alcohol are two of the most commonly used and abused recreational drugs, are often used simultaneously, and have been linked to significant health hazards. Furthermore, patients diagnosed with dependence on one drug are highly likely to be dependent on the other. Several studies have shown the effects of each drug independently on gene expression within many brain regions, including the ventral tegmental area (VTA). Dopaminergic (DA) neurons of the dopamine reward pathway originate from the VTA, which is believed to be central to the mechanism of addiction and drug reinforcement. Using a well-established rat model for both nicotine and alcohol perinatal exposure, we investigated miRNA and mRNA expression of dopaminergic (DA) neurons of the VTA in rat pups following perinatal alcohol and joint nicotine-alcohol exposure. Microarray analysis was then used to profile the differential expression of both miRNAs and mRNAs from DA neurons of each treatment group to further explore the altered genes and related biological pathways modulated. Predicted and validated miRNA-gene target pairs were analyzed to further understand the roles of miRNAs within these networks following each treatment, along with their post transcription regulation points affecting gene expression throughout development. This study suggested that glutamatergic synapse and axon guidance pathways were specifically enriched and many miRNAs and genes were significantly altered following alcohol or nicotine-alcohol perinatal exposure when compared to saline control. These results provide more detailed insight into the cell proliferation, neuronal migration, neuronal axon guidance during the infancy in rats in response to perinatal alcohol/ or nicotine-alcohol exposure.
Collapse
Affiliation(s)
- Tina Kazemi
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Shuyan Huang
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Naze G Avci
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Charlotte Mae K Waits
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Yasemin M Akay
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Metin Akay
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA.
| |
Collapse
|
22
|
Lim ZM, Chie QT, Teh LK. Influence of dopamine receptor gene on eating behaviour and obesity in Malaysia. Meta Gene 2020. [DOI: 10.1016/j.mgene.2020.100736] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
23
|
Patrício P, Mateus-Pinheiro A, Alves ND, Morais M, Rodrigues AJ, Bessa JM, Sousa N, Pinto L. miR-409 and miR-411 Modulation in the Adult Brain of a Rat Model of Depression and After Fluoxetine Treatment. Front Behav Neurosci 2020; 14:136. [PMID: 32848656 PMCID: PMC7427047 DOI: 10.3389/fnbeh.2020.00136] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/14/2020] [Indexed: 12/14/2022] Open
Abstract
Depression is a chronic debilitating disorder predicted to affect around 20% of the world population. Both brain and peripheral changes, including neuroplastic changes have been shown to occur in the brains of depressed individuals and animal models of depression. Over the past few decades, growing evidence has supported the role of miRNAs as regulators of critical aspects of brain plasticity and function, namely in the context of depression. These molecules are not only highly expressed in the brain, but are also relatively stable in bodily fluids, including blood. Previous microarray analysis from our group has disclosed molecular players in the hippocampal dentate gyrus (DG), in the context of depression and antidepressant treatment. Two miRNAs in particular-miR-409-5p and miR-411-5p-were significantly up-regulated in the DG of an unpredictable chronic mild stress (CMS) rat model of depression and reversed by antidepressant treatment. Here, we further analyzed the levels of these miRNAs along the DG longitudinal axis and in other brain regions involved in the pathophysiology of depression, as well as in peripheral blood of CMS-exposed rats and after fluoxetine treatment. The effects of CMS and fluoxetine treatment on miR-409-5p and miR-411-5p levels varied across brain regions, and miR-411-5p was significantly decreased in the blood of fluoxetine-treated rats. Additional bioinformatic analyses revealed target genes and pathways of these miRNAs related to neurotransmitter signaling and neuroplasticity functions; an implication of the two miRNAs in the regulation of the cellular and molecular changes observed in these brain regions in depression is worth further examination.
Collapse
Affiliation(s)
- Patrícia Patrício
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory (AL), Braga/Guimarães, Portugal
| | - António Mateus-Pinheiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory (AL), Braga/Guimarães, Portugal
| | - Nuno Dinis Alves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory (AL), Braga/Guimarães, Portugal
| | - Mónica Morais
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory (AL), Braga/Guimarães, Portugal
| | - Ana João Rodrigues
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory (AL), Braga/Guimarães, Portugal
| | - João Miguel Bessa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory (AL), Braga/Guimarães, Portugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory (AL), Braga/Guimarães, Portugal
| | - Luísa Pinto
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory (AL), Braga/Guimarães, Portugal
| |
Collapse
|
24
|
Zhang Y, Zhai H. Bilobalide assuages morphine-induced addiction in hippocampal neuron cells through upregulation of microRNA-101. J Biochem Mol Toxicol 2020; 34:e22493. [PMID: 32319158 DOI: 10.1002/jbt.22493] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 12/24/2019] [Accepted: 03/03/2020] [Indexed: 11/09/2022]
Abstract
Bilobalide exhibits many biological activities, but its effects on morphine stimulation have not been elucidated. The research aims to explore the function and underlying mechanisms of bilobalide in morphine-led hippocampal neuron cells. Cells were treated with or without morphine or oxaliplatin (OXA), bilobalide, or SCH772984 dilutions. miR-101 inhibitor and negative control were transfected into cells. Western blot and quantitative reverse transcription-polymerase chain reaction were, respectively, conducted to measure the relative expression of proteins or RNAs. Morphine improved the expression levels of orexin1 receptor (OX1R) and c-FOS, the p/t-ERK/PKC as well. The c-FOS protein level and p/t-ERK/PKC were significantly elevated by morphine + OXA. Bilobalide had no effect on OX1R and p/t-PKC but evidently decreased the c-FOS and p/t-ERK. The p-ERK and the c-FOS accumulation levels were remarkably reduced by SCH772984. The production of miR-101 was promoted by bilobalide but inhibited by the miR-101 inhibitor. miR-101 inhibitor abolished bilobalide's inhibitory effects on p/t-ERK. Bilobalide exhibited morphine-induced effects on hippocampal neuron cells by upregulating miR-101.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Pathology, Huaihe Hospital, Henan University, Kaifeng, China
| | - Hongyin Zhai
- Department of Children Rehabilitation Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
25
|
Shi M, Zhang T, Zeng Y. A Curiosity-Based Learning Method for Spiking Neural Networks. Front Comput Neurosci 2020; 14:7. [PMID: 32116621 PMCID: PMC7020337 DOI: 10.3389/fncom.2020.00007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 01/20/2020] [Indexed: 01/12/2023] Open
Abstract
Spiking Neural Networks (SNNs) have shown favorable performance recently. Nonetheless, the time-consuming computation on neuron level and complex optimization limit their real-time application. Curiosity has shown great performance in brain learning, which helps biological brains grasp new knowledge efficiently and actively. Inspired by this leaning mechanism, we propose a curiosity-based SNN (CBSNN) model, which contains four main learning processes. Firstly, the network is trained with biologically plausible plasticity principles to get the novelty estimations of all samples in only one epoch; secondly, the CBSNN begins to repeatedly learn the samples whose novelty estimations exceed the novelty threshold and dynamically update the novelty estimations of samples according to the learning results in five epochs; thirdly, in order to avoid the overfitting of the novel samples and forgetting of the learned samples, CBSNN retrains all samples in one epoch; finally, step two and step three are periodically taken until network convergence. Compared with the state-of-the-art Voltage-driven Plasticity-centric SNN (VPSNN) under standard architecture, our model achieves a higher accuracy of 98.55% with only 54.95% of its computation cost on the MNIST hand-written digit recognition dataset. Similar conclusion can also be found out in other datasets, i.e., Iris, NETtalk, Fashion-MNIST, and CIFAR-10, respectively. More experiments and analysis further prove that such curiosity-based learning theory is helpful in improving the efficiency of SNNs. As far as we know, this is the first practical combination of the curiosity mechanism and SNN, and these improvements will make the realistic application of SNNs possible on more specific tasks within the von Neumann framework.
Collapse
Affiliation(s)
- Mengting Shi
- Research Center for Brain-inspired Intelligence, Institute of Automation, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Tielin Zhang
- Research Center for Brain-inspired Intelligence, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Yi Zeng
- Research Center for Brain-inspired Intelligence, Institute of Automation, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
26
|
Gu WJ, Zhang C, Zhong Y, Luo J, Zhang CY, Zhang C, Wang C. Altered serum microRNA expression profile in subjects with heroin and methamphetamine use disorder. Biomed Pharmacother 2020; 125:109918. [PMID: 32036213 DOI: 10.1016/j.biopha.2020.109918] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 12/02/2019] [Accepted: 12/18/2019] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES Drug abuse is one of the most severe global social and public health problems, especially in China. However, objective blood biomarkers that are easy to detect are still in great need. This study was aim to explore the expression pattern of circulating microRNAs (miRNAs) in subjects with drug addiction and test the potential of altered serum miRNAs as noninvasive diagnostic tools for drug abuse. METHODS Serum samples were obtained from 42 heroin abusers, 42 methamphetamine (MA) abusers and 42 controls. Microarray-based miRNA analysis was first applied to screen unique serum miRNA profiles in drug abusers on a training set of serum samples from 12 heroin abusers, 12 MA abusers and 12 control subjects. The expression levels of selected candidate miRNAs were subsequently verified in individual samples of the training set and further confirmed independently in a validation set of samples from 30 heroin abusers, 30 MA abusers and 30 controls using real-time quantitative polymerase chain reaction (RT-qPCR). RESULTS Microarray analysis identified 116 and 109 significantly altered miRNAs in heroin abusers and MA abusers, respectively. Three miRNAs, including let-7b-5p, miR-206 and miR-486-5p, were verified to be significantly and steadily increased in heroin abusers, and miR-9-3p was significantly increased in MA abusers compared with normal controls. The areas under the curve (AUCs) of the ROC curve of these miRNAs ranged from 0.718 to 0.867. CONCLUSIONS Our study raises the possibility that the altered serum miRNAs could potentially be used as an auxiliary tool to identify individuals in drug abuse and addiction.
Collapse
Affiliation(s)
- Wan-Jian Gu
- Department of Clinical Laboratory, Jinling Hospital, State Key Laboratory of Analytical Chemistry for Life Science, Medical School of Nanjing University, Nanjing, 210002, China; Department of Clinical Laboratory, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Cuiping Zhang
- Department of Clinical Laboratory, Jinling Hospital, State Key Laboratory of Analytical Chemistry for Life Science, Medical School of Nanjing University, Nanjing, 210002, China; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Advanced Institute for Life Sciences, Nanjing University School of Life Sciences, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, Nanjing University, Nanjing, 210046, China
| | - Yujie Zhong
- Department of Clinical Laboratory, Jinling Hospital, State Key Laboratory of Analytical Chemistry for Life Science, Medical School of Nanjing University, Nanjing, 210002, China; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Advanced Institute for Life Sciences, Nanjing University School of Life Sciences, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, Nanjing University, Nanjing, 210046, China
| | - Jun Luo
- Central Laboratory of Jiangsu Health Vocational College, Nanjing, 210029, China
| | - Chen-Yu Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Advanced Institute for Life Sciences, Nanjing University School of Life Sciences, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, Nanjing University, Nanjing, 210046, China
| | - Chunni Zhang
- Department of Clinical Laboratory, Jinling Hospital, State Key Laboratory of Analytical Chemistry for Life Science, Medical School of Nanjing University, Nanjing, 210002, China; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Advanced Institute for Life Sciences, Nanjing University School of Life Sciences, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, Nanjing University, Nanjing, 210046, China.
| | - Cheng Wang
- Department of Clinical Laboratory, Jinling Hospital, State Key Laboratory of Analytical Chemistry for Life Science, Medical School of Nanjing University, Nanjing, 210002, China; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Advanced Institute for Life Sciences, Nanjing University School of Life Sciences, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, Nanjing University, Nanjing, 210046, China.
| |
Collapse
|
27
|
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
|
28
|
Szechtman H, Harvey BH, Woody EZ, Hoffman KL. The Psychopharmacology of Obsessive-Compulsive Disorder: A Preclinical Roadmap. Pharmacol Rev 2020; 72:80-151. [PMID: 31826934 DOI: 10.1124/pr.119.017772] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
This review evaluates current knowledge about obsessive-compulsive disorder (OCD), with the goal of providing a roadmap for future directions in research on the psychopharmacology of the disorder. It first addresses issues in the description and diagnosis of OCD, including the structure, measurement, and appropriate description of the disorder and issues of differential diagnosis. Current pharmacotherapies for OCD are then reviewed, including monotherapy with serotonin reuptake inhibitors and augmentation with antipsychotic medication and with psychologic treatment. Neuromodulatory therapies for OCD are also described, including psychosurgery, deep brain stimulation, and noninvasive brain stimulation. Psychotherapies for OCD are then reviewed, focusing on behavior therapy, including exposure and response prevention and cognitive therapy, and the efficacy of these interventions is discussed, touching on issues such as the timing of sessions, the adjunctive role of pharmacotherapy, and the underlying mechanisms. Next, current research on the neurobiology of OCD is examined, including work probing the role of various neurotransmitters and other endogenous processes and etiology as clues to the neurobiological fault that may underlie OCD. A new perspective on preclinical research is advanced, using the Research Domain Criteria to propose an adaptationist viewpoint that regards OCD as the dysfunction of a normal motivational system. A systems-design approach introduces the security motivation system (SMS) theory of OCD as a framework for research. Finally, a new perspective on psychopharmacological research for OCD is advanced, exploring three approaches: boosting infrastructure facilities of the brain, facilitating psychotherapeutic relearning, and targeting specific pathways of the SMS network to fix deficient SMS shut-down processes. SIGNIFICANCE STATEMENT: A significant proportion of patients with obsessive-compulsive disorder (OCD) do not achieve remission with current treatments, indicating the need for innovations in psychopharmacology for the disorder. OCD may be conceptualized as the dysfunction of a normal, special motivation system that evolved to manage the prospect of potential danger. This perspective, together with a wide-ranging review of the literature, suggests novel directions for psychopharmacological research, including boosting support systems of the brain, facilitating relearning that occurs in psychotherapy, and targeting specific pathways in the brain that provide deficient stopping processes in OCD.
Collapse
Affiliation(s)
- Henry Szechtman
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada (H.S.); SAMRC Unit on Risk Resilience in Mental Disorders, Department of Psychiatry, University of Cape Town, and Center of Excellence for Pharmaceutical Sciences, School of Pharmacy, North-West University (Potchefstroom Campus), Potchefstroom, South Africa (B.H.H.); Department of Psychology, University of Waterloo, Waterloo, Ontario, Canada (E.Z.W.); and Centro de Investigación en Reproducción Animal, CINVESTAV-Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico (K.L.H.)
| | - Brian H Harvey
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada (H.S.); SAMRC Unit on Risk Resilience in Mental Disorders, Department of Psychiatry, University of Cape Town, and Center of Excellence for Pharmaceutical Sciences, School of Pharmacy, North-West University (Potchefstroom Campus), Potchefstroom, South Africa (B.H.H.); Department of Psychology, University of Waterloo, Waterloo, Ontario, Canada (E.Z.W.); and Centro de Investigación en Reproducción Animal, CINVESTAV-Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico (K.L.H.)
| | - Erik Z Woody
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada (H.S.); SAMRC Unit on Risk Resilience in Mental Disorders, Department of Psychiatry, University of Cape Town, and Center of Excellence for Pharmaceutical Sciences, School of Pharmacy, North-West University (Potchefstroom Campus), Potchefstroom, South Africa (B.H.H.); Department of Psychology, University of Waterloo, Waterloo, Ontario, Canada (E.Z.W.); and Centro de Investigación en Reproducción Animal, CINVESTAV-Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico (K.L.H.)
| | - Kurt Leroy Hoffman
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada (H.S.); SAMRC Unit on Risk Resilience in Mental Disorders, Department of Psychiatry, University of Cape Town, and Center of Excellence for Pharmaceutical Sciences, School of Pharmacy, North-West University (Potchefstroom Campus), Potchefstroom, South Africa (B.H.H.); Department of Psychology, University of Waterloo, Waterloo, Ontario, Canada (E.Z.W.); and Centro de Investigación en Reproducción Animal, CINVESTAV-Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico (K.L.H.)
| |
Collapse
|
29
|
Decreased Level of Blood MicroRNA-133b in Men with Opioid Use Disorder on Methadone Maintenance Therapy. J Clin Med 2019; 8:jcm8081105. [PMID: 31349687 PMCID: PMC6722972 DOI: 10.3390/jcm8081105] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 06/14/2019] [Accepted: 07/24/2019] [Indexed: 12/20/2022] Open
Abstract
Although previous animal studies have indicated that certain micro ribonucleic acids (microRNAs) play a part in the pathway of opioid addiction, whether such findings extend to human models is yet unknown. This study aims to investigate the important microRNA expressions in patients with opioid use disorder (OUD) on methadone maintenance treatment (MMT) compared to healthy controls and analyze the correlation between microRNAs and opioid characteristics among the patients. We recruited 50 patients and 25 controls, and both groups were matched regarding gender, age, and body mass index. Serum microRNAs (miR-133b, miR-23b, miR-190, miR-206, miR-210, and miR-21) were measured. The age of OUD onset, duration of MMT participation, and recent daily methadone dosage were considered the opioid characteristics. We adopted the t-test to compare the difference between patients and controls and Pearson's correlation to evaluate the association between microRNAs and opioid profiles. Only the level of miR-133b in OUD patients on MMT was significantly lower than that in healthy controls. We did not detect differences of any other microRNA expressions between the two groups. Furthermore, we found no evidence to support the association between microRNAs and opioid characteristics. This study indicates that miR-133b values may be decreased in OUD patients on MMT.
Collapse
|
30
|
Pharmacological Transdifferentiation of Human Nasal Olfactory Stem Cells into Dopaminergic Neurons. Stem Cells Int 2019; 2019:2945435. [PMID: 31236114 PMCID: PMC6545791 DOI: 10.1155/2019/2945435] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 02/25/2019] [Indexed: 01/01/2023] Open
Abstract
The discovery of novel drugs for neurodegenerative diseases has been a real challenge over the last decades. The development of patient- and/or disease-specific in vitro models represents a powerful strategy for the development and validation of lead candidates in preclinical settings. The implementation of a reliable platform modeling dopaminergic neurons will be an asset in the study of dopamine-associated pathologies such as Parkinson's disease. Disease models based on cell reprogramming strategies, using either human-induced pluripotent stem cells or transcription factor-mediated transdifferentiation, are among the most investigated strategies. However, multipotent adult stem cells remain of high interest to devise direct conversion protocols and establish in vitro models that could bypass certain limitations associated with reprogramming strategies. Here, we report the development of a six-step chemically defined protocol that drives the transdifferentiation of human nasal olfactory stem cells into dopaminergic neurons. Morphological changes were progressively accompanied by modifications matching transcript and protein dopaminergic signatures such as LIM homeobox transcription factor 1 alpha (LMX1A), LMX1B, and tyrosine hydroxylase (TH) expression, within 42 days of differentiation. Phenotypic changes were confirmed by the production of dopamine from differentiated neurons. This new strategy paves the way to develop more disease-relevant models by establishing reprogramming-free patient-specific dopaminergic cell models for drug screening and/or target validation for neurodegenerative diseases.
Collapse
|
31
|
Kuatsjah E, Khoshnam M, Menon C. Investigation on the effect of noisy galvanic vestibular stimulation on fine motor skills during a visuomotor task in healthy participants. PLoS One 2019; 14:e0216214. [PMID: 31048906 PMCID: PMC6497271 DOI: 10.1371/journal.pone.0216214] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 04/16/2019] [Indexed: 11/24/2022] Open
Abstract
Noisy galvanic vestibular stimulation (nGVS) has been shown to improve dynamic walking stability, affect postural responses, enhance balance in healthy subjects, and influence motor performance in individuals with Parkinson’s disease. Although the studies to fully characterize the effect of nGVS are still ongoing, stochastic resonance theory which states that the addition of noisy signal may enhance a weak sensory input signals transmission in a non-linear system may provide a possible explanation for the observed positive effects of nGVS. This study explores the effect of nGVS on fine tracking behavior in healthy subjects. Ten healthy participants performed a computer-based visuomotor task by controlling an object with a joystick to follow an amplitude-modulated signal path while simultaneously receiving a sham or pink noise nGVS. The stimulation was generated to have a zero-mean, linearly detrended 1/f-type power spectrum, Gaussian distribution within 0.1–10 Hz range, and a standard deviation (SD) set to 90% based on each participant’s cutaneous threshold value. Results show that simultaneous nGVS delivery statistically improved the tracking performance with a decreased root-mean-squared error of 5.71±6.20% (mean±SD), a decreased time delay of 11.88±9.66% (mean±SD), and an increased signal-to-noise ratio of 2.93% (median, interquartile range (IQR) 3.31%). This study showed evidence that nGVS may be beneficial in improving sensorimotor performance during a fine motor tracking task requiring fine wrist movement in healthy subjects. Further research with a more comprehensive subset of tasks is required to fully characterize the effects of nGVS on fine motor skills.
Collapse
Affiliation(s)
- Eunice Kuatsjah
- Menrva Research Group, Schools of Mechatronic Systems Engineering and Engineering Science, Simon Fraser University, Metro Vancouver, British Columbia, Canada
| | - Mahta Khoshnam
- Menrva Research Group, Schools of Mechatronic Systems Engineering and Engineering Science, Simon Fraser University, Metro Vancouver, British Columbia, Canada
| | - Carlo Menon
- Menrva Research Group, Schools of Mechatronic Systems Engineering and Engineering Science, Simon Fraser University, Metro Vancouver, British Columbia, Canada
- * E-mail:
| |
Collapse
|
32
|
Anoushirvani AA, Aghabozorgi R, Ahmadi A, Arjomandzadegan M, Khalili S, Sahraei M, Fereydouni T, Khademi Z. The Relationship Between rs3212986C>A Polymorphism and Tumor Stage in Lung Cancer Patients. Cureus 2019; 11:e4423. [PMID: 31245210 PMCID: PMC6559387 DOI: 10.7759/cureus.4423] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Background The nucleotide excision repair (NER) system is one of the most important deoxyribonucleic acid (DNA) repair mechanisms and is critical for chemotherapy resistance. We conducted the present study to investigate the association between two polymorphisms of excision of repair cross-complementing group 1 (ERCC1), the key component of the NER pathway, and the clinicopathological features of patients with non-small cell lung cancer (NSCLC). Methods A total of 38 patients with confirmed NSCLC were included in our study. DNA was extracted from peripheral blood. ERCC1 rs3212986 (8092) and rs11615 (118) were genotyped using molecular assays including polymerase chain reaction (PCR) with restriction fragment length polymorphism (by MboII and HpyCH4 enzymes) and sequencing. Results The PCR results indicated the correct performance of the genomics extraction and molecular protocols. The distribution of C/C, C/A and A/A genotypes at position 8092 was 42.10%, 47.36%, and 10.52% respectively (P=0.03). Multivariate regression analysis showed that there was a significant correlation between C8092A (rs3212986) polymorphism and metastasis, grade of the tumor, and response to treatment. Individuals carrying the rs3212986 CA genotype and A allele had a significantly worse response to the treatment. Also, the correlation between alteration at this genomics location and patients with NSCLC who used to smoke cigarettes was positive. However, no significant association was detected between rs11615 C118>T polymorphism and demographic characteristics of patients with NSCLC. Conclusion We concluded that in lung cancer patients there is a relationship between tumor stage and rs3212986C>A polymorphism.
Collapse
Affiliation(s)
| | - Reza Aghabozorgi
- Internal Medicine, Arak University of Medical Sciences, Arak, IRN
| | - Azam Ahmadi
- Genetics, Arak University of Medical Sciences, Arak, IRN
| | | | - Sara Khalili
- Microbiology, Arak University of Medical Sciences, Arak, IRN
| | - Maryam Sahraei
- Genetics, Arak University of Medical Sciences, Arak, IRN
| | - Taha Fereydouni
- Internal Medicine, Arak University of Medical Sciences, Arak, IRN
| | - Zoha Khademi
- Internal Medicine, Arak University of Medical Sciences, Arak, IRN
| |
Collapse
|
33
|
Kong C, Shin J, Koh CS, Lee J, Yoon MS, Cho Y, Kim S, Jun S, Jung H, Chang J. Optimization of Medial Forebrain Bundle Stimulation Parameters for Operant Conditioning of Rats. Stereotact Funct Neurosurg 2019; 97:1-9. [DOI: 10.1159/000497151] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 01/18/2019] [Indexed: 11/19/2022]
|
34
|
Su H, Zhu L, Li J, Wang R, Liu D, Han W, Cadet JL, Chen T. Regulation of microRNA-29c in the nucleus accumbens modulates methamphetamine -induced locomotor sensitization in mice. Neuropharmacology 2019; 148:160-168. [PMID: 30639389 DOI: 10.1016/j.neuropharm.2019.01.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 01/04/2019] [Accepted: 01/09/2019] [Indexed: 12/21/2022]
Abstract
Changes in microRNA (miRNA)-mediated gene expression in the nucleus accumbens (NAc) may play important roles in regulating drug addiction. MiR-29c is a highly expressed miRNA in the human and rodent nervous systems where it plays a broad regulatory role. As the first step towards investigating potential functions of miR-29c in methamphetamine (METH) addiction, we used C57BL/6 mice in a model of METH-induced locomotor sensitization. We measured miR-29c expression changes in the NAc of the mice after repeated-intermittent METH exposure and acute METH administration respectively by using quantitative real-time PCR (qPCR). We found that miR-29c expression was significantly down-regulated in the NAc of METH-sensitized mice but not in the acute METH-treated mice. Then, we tested the respective effects of miR-29c over-expression and inhibition in the NAc on METH-induced locomotor sensitization. To reach this goal, we constructed adeno-associated virus (AAV)-expressing miR-29c (AAV-miR-29c) and its corresponding inhibitor - tough decoy (AAV-anti-miR-29c TuD) to over-express and inhibit miR-29c, respectively. We found that AAV-miR-29c over-expression in the NAc enhanced METH-induced locomotor sensitization, whereas AAV inhibition of miR-29c expression in the NAc attenuated the effects of METH. Moreover, we observed the participation of Dnmt3a, Dnmt3b, and Meg3 in the effects of miR-29c on METH sensitization. Our results suggest that miR-29c is an important epigenetic regulator of METH-induced behavioural sensitization and changes in gene expression. These data further suggest a potential role of miR-29c in regulating long-term METH-induced adaptation in the brain.
Collapse
Affiliation(s)
- Hang Su
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, PR China; The Key Laboratory of Health Ministry for Forensic Medicine, Xi'an Jiaotong University, Shaanxi, 710061, PR China
| | - Li Zhu
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, PR China; The Key Laboratory of Health Ministry for Forensic Medicine, Xi'an Jiaotong University, Shaanxi, 710061, PR China
| | - Jiaqi Li
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, PR China; The Key Laboratory of Health Ministry for Forensic Medicine, Xi'an Jiaotong University, Shaanxi, 710061, PR China
| | - Rui Wang
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, PR China; The Key Laboratory of Health Ministry for Forensic Medicine, Xi'an Jiaotong University, Shaanxi, 710061, PR China
| | - Dan Liu
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, PR China; The Key Laboratory of Health Ministry for Forensic Medicine, Xi'an Jiaotong University, Shaanxi, 710061, PR China
| | - Wei Han
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, PR China; The Key Laboratory of Health Ministry for Forensic Medicine, Xi'an Jiaotong University, Shaanxi, 710061, PR China
| | - Jean Lud Cadet
- Molecular Neuropsychiatry Research Branch, National Institute on Drug Abuse/NIH/DHHS, Bayview Boulevard, Maryland, 21224, USA
| | - Teng Chen
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, PR China; The Key Laboratory of Health Ministry for Forensic Medicine, Xi'an Jiaotong University, Shaanxi, 710061, PR China.
| |
Collapse
|
35
|
Chen SJ, Liao DL, Chen CH, Wang TY, Chen KC. Construction and Analysis of Protein-Protein Interaction Network of Heroin Use Disorder. Sci Rep 2019; 9:4980. [PMID: 30899073 PMCID: PMC6428805 DOI: 10.1038/s41598-019-41552-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 03/11/2019] [Indexed: 12/17/2022] Open
Abstract
Heroin use disorder (HUD) is a complex disease resulting from interactions among genetic and other factors (e.g., environmental factors). The mechanism of HUD development remains unknown. Newly developed network medicine tools provide a platform for exploring complex diseases at the system level. This study proposes that protein–protein interactions (PPIs), particularly those among proteins encoded by casual or susceptibility genes, are extremely crucial for HUD development. The giant component of our constructed PPI network comprised 111 nodes with 553 edges, including 16 proteins with large degree (k) or high betweenness centrality (BC), which were further identified as the backbone of the network. JUN with the largest degree was suggested to be central to the PPI network associated with HUD. Moreover, PCK1 with the highest BC and MAPK14 with the secondary largest degree and 9th highest BC might be involved in the development HUD and other substance diseases.
Collapse
Affiliation(s)
- Shaw-Ji Chen
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan.,Department of Psychiatry, Mackay Memorial Hospital, Taitung Branch, Taiwan
| | - Ding-Lieh Liao
- Bali Psychiatric Center, Department of Health, Executive Yuan, New Taipei, Taiwan
| | - Chia-Hsiang Chen
- Department of Psychiatry, Chang Gung Memorial Hospital at Linkou and Chang Gung University School of Medicine, Taoyuan, Taiwan
| | - Tse-Yi Wang
- Department of Medical Informatics, Tzu Chi University, Hualien, Taiwan
| | - Kuang-Chi Chen
- Department of Medical Informatics, Tzu Chi University, Hualien, Taiwan.
| |
Collapse
|
36
|
Patel AA, Ganepola GA, Rutledge JR, Chang DH. The Potential Role of Dysregulated miRNAs in Alzheimer’s Disease Pathogenesis and Progression. J Alzheimers Dis 2019; 67:1123-1145. [DOI: 10.3233/jad-181078] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Ankur A. Patel
- Department of Research, Center for Cancer Research and Genomic Medicine, The Valley Hospital, Paramus, NJ, USA
| | - Ganepola A.P. Ganepola
- Department of Research, Center for Cancer Research and Genomic Medicine, The Valley Hospital, Paramus, NJ, USA
| | - John R. Rutledge
- Department of Oncology Special Program, The Daniel and Gloria Blumenthal Cancer Center, The Valley Hospital, Paramus, NJ, USA
| | - David H. Chang
- Department of Research, Center for Cancer Research and Genomic Medicine, The Valley Hospital, Paramus, NJ, USA
| |
Collapse
|
37
|
Keller RF, Kanlikilicer P, Dragomir A, Fan Y, Akay YM, Akay M. Investigating the Effect of Perinatal Nicotine Exposure on Dopaminergic Neurons in the VTA Using miRNA Expression Profiles. IEEE Trans Nanobioscience 2019; 16:843-849. [PMID: 29364128 DOI: 10.1109/tnb.2017.2776841] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Maternal smoking during pregnancy is associated with developmental, cognitive, and behavioral disorders, including low birth weight, attention deficit hyperactivity disorder, learning disabilities, and drug abuse later in life. Nicotine activates the reward-driven behavior characteristic of drug abuse. Dopaminergic (DA) neurons originating from the ventral tegmental area (VTA) of the brain, which are stimulated by nicotine and other stimuli, are widely implicated in the natural reward pathway that is known to contribute to addiction. In recent years, microRNAs have been implicated in disrupting regulatory mechanisms due to their capability of targeting multiple genes and thus inducing downstream effects along many pathways. In order to investigate miRNA expression of dopaminergic neurons from the VTA, we employed patch clamping to identify and harvest both DA and non-DA neurons from rats perinatally exposed to nicotine for use in single-cell RT-qPCR. Our data indicated that miR-140-5p and miR-140-3p were upregulated in DA neurons; while miR-140-3p and miR-212 were differentially expressed in non-DA neurons. A functional enrichment analysis was also performed on our miRNA-gene prediction network and predicted that our miRNAs target genes involved in drug response and neuroplasticity.
Collapse
|
38
|
Beayno A, El Hayek S, Noufi P, Tarabay Y, Shamseddeen W. The Role of Epigenetics in Addiction: Clinical Overview and Recent Updates. Methods Mol Biol 2019; 2011:609-631. [PMID: 31273724 DOI: 10.1007/978-1-4939-9554-7_35] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Addiction is an international public health problem. It is a polygenic disorder best understood by accounting for the interplay between genetic and environmental factors. A recent way of perceiving this interaction is through epigenetics, which help grasp the neurobiological changes that occur in addiction and explain its relapsing-remitting nature. It is now known that every cell has a different way of expressing its phenotype, despite a universal DNA sequence. This is particularly true in the central nervous system where environmental factors influence this expression. Three major epigenetic processes have been found to participate in the perpetuation of addiction by changing the state of the chromatin and the degree of gene transcription: histone acetylation and methylation, DNA methylation, and noncoding RNAs. In the animal model literature, substantial evidence exists about the role of these epigenetic changes in the different phases of substance use disorders. This book chapter is a non-systematic literature review of the recent publications tackling the topic of epigenetics in addiction. Even though this evidence remains scarce and relatively poorly systematized, it is a promising foundation for future research of molecules that target specific brain regions and their functions to address core behavioral changes seen in addiction.
Collapse
Affiliation(s)
- Antoine Beayno
- Department of Psychiatry, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Samer El Hayek
- Department of Psychiatry, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Paul Noufi
- Department of Psychiatry, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Yara Tarabay
- Faculty of Pedagogy, Lebanese University, New Rawda, Lebanon.,Faculty of Natural and Applied Sciences, Notre Dame University, Louaize, Lebanon
| | - Wael Shamseddeen
- Department of Psychiatry, Faculty of Medicine, American University of Beirut, Beirut, Lebanon. .,Department of Psychiatry, University of Michigan, Ann Arbor, Michigan, USA.
| |
Collapse
|
39
|
Armañanzas R. Revealing post-transcriptional microRNA-mRNA regulations in Alzheimer's disease through ensemble graphs. BMC Genomics 2018; 19:668. [PMID: 30255799 PMCID: PMC6157163 DOI: 10.1186/s12864-018-5025-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND In silico investigations on the integration of multiple datasets are in need of higher statistical power methods to unveil secondary findings that were hidden from the initial analyses. We present here a novel method for the network analysis of messenger RNA post-translational regulation by microRNA molecules. The method integrates expression data and sequence binding predictions through a set of sound machine learning techniques, forwarding all results to an ensemble graph of regulations. RESULTS Bayesian network classifiers are induced based on a pool of ensemble graphs with ascending order of complexity. Individual goodness-of-fit and classification performances are evaluated for each learned model. As a testbed, four Alzheimer's disease datasets are integrated using the new approach, achieving top values of 0.9794 ± 0.01 for the area under the receiver operating characteristic curve and 0.9439 ± 0.0234 for the prediction accuracy. CONCLUSIONS Post-transcriptional regulations found by the optimal network classifier concur with previous literature findings. Furthermore, additional network structures suggest previously unreported regulations in the state of the art of Alzheimer's research. The quantitative performance as well as sound biological findings provide confidence in the ensemble approach and encourage similar integrative analyses for other conditions.
Collapse
Affiliation(s)
- Rubén Armañanzas
- Department of Bioengineering, Krasnow Institute for Advanced Study, George Mason University, 4400 University Dr, MS2A1, Fairfax, 22030, VA, USA.
| |
Collapse
|
40
|
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
|
41
|
Ji Y, Yang X, Su H. Overexpression of microRNA-375 impedes platelet-derived growth factor-induced proliferation and migration of human fetal airway smooth muscle cells by targeting Janus kinase 2. Biomed Pharmacother 2017; 98:69-75. [PMID: 29245068 DOI: 10.1016/j.biopha.2017.12.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 11/20/2017] [Accepted: 12/04/2017] [Indexed: 12/21/2022] Open
Abstract
The abnormal proliferation and migration of airway smooth muscle (ASM) cells play a critical role in airway remodeling during the development of asthma. MicroRNAs (miRNAs) have emerged as critical regulators of ASM cell proliferation and migration in airway remodeling. In this study, we aimed to investigate the potential role of miR-375 in the regulation of platelet-derived growth factor (PDGF)-induced fetal ASM cell proliferation and migration. Our results showed that miR-375 expression was significantly decreased in fetal ASM cells that were treated with PDGF. Functional data showed that overexpression of miR-375 inhibited the proliferation and migration of fetal ASM cells, whereas inhibition of miR-375 enhanced the proliferation and migration of fetal ASM cells. The results of bioinformatics analysis and a dual-luciferase reporter assay showed that miR-375 binds directly to the 3'-untranslated region of Janus kinase 2 (JAK2). Further data confirmed that miR-375 negatively regulates the expression of JAK2 in fetal ASM cells. Moreover, miR-375 also impeded the PDGF-induced activation of signal transducer and activator of transcription 3 (STAT3) in fetal ASM cells. However, restoration of JAK2 expression partially reversed the inhibitory effect of miR-375 on fetal ASM cell proliferation and migration. Overall, our results demonstrate that miR-375 inhibits fetal ASM cell proliferation and migration by targeting JAK2/STAT3 signaling. Our study provides a potential therapeutic target for the development of novel treatment strategies for pediatric asthma.
Collapse
Affiliation(s)
- Yamei Ji
- Department of Paediatrics, Yulin Xingyuan Hospital, Yulin, Shaanxi 719000, China
| | - Xin Yang
- Second Department of Paediatric Internal Medicine, Yulin Children's Hospital, Yulin, Shaanxi 719000, China
| | - Huixia Su
- Second Department of Paediatric Internal Medicine, Yulin Children's Hospital, Yulin, Shaanxi 719000, China.
| |
Collapse
|
42
|
Huang B, Jiang XC, Zhang TY, Hu YL, Tabata Y, Chen Z, Pluchino S, Gao JQ. Peptide modified mesenchymal stem cells as targeting delivery system transfected with miR-133b for the treatment of cerebral ischemia. Int J Pharm 2017; 531:90-100. [PMID: 28827201 DOI: 10.1016/j.ijpharm.2017.08.073] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 07/12/2017] [Accepted: 08/12/2017] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSCs) have been regarded as potential targeting vehicles and demonstrated to exert therapeutic benefits for brain diseases. Direct homing to diseased tissue is crucial for stem cell-based therapy. In this study, a peptide-based targeting approach was established to enhance cell homing to cerebral ischemic lesion. Palmitic acid-peptide painted onto the cell membrane was able to direct MSCs to ischemic tissues without any observed cell cytotoxicity and influence on differentiation, thus reducing accumulation of cells in peripheral organs and increasing engraftment of cells in the targeted tissues. With enhanced cell homing, MSCs were used to deliver miR-133b to increase the expression level of miR-133b in an ischemic lesion and further improve therapeutic effects. This study is the first to develop MSCs co-modified with targeting peptide and microRNAs as potential targeting therapeutic agents. This targeting delivery system is expected to be applicable to other cell types and other diseases aside from stroke.
Collapse
Affiliation(s)
- Bing Huang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Xin-Chi Jiang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Tian-Yuan Zhang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Yu-Lan Hu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Yasuhiko Tabata
- Department of Biomaterials, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Zhong Chen
- Department of Pharmacology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Stefano Pluchino
- Department of Clinical Neurosciences, Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridgeshire, UK
| | - Jian-Qing Gao
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China.
| |
Collapse
|
43
|
Mohamed RMP, Kumar J, Yap E, Mohamed IN, Sidi H, Adam RL, Das S. Try to Remember: Interplay between Memory and Substance Use Disorder. Curr Drug Targets 2017. [PMID: 28641520 DOI: 10.2174/1389450118666170622092824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Memories associated with substance use disorders, or substance-associated cues increase the likelihood of craving and relapse during abstinence. There is a growing consensus that manipulation of synaptic plasticity may reduce the strength of substance abuse-related memories. On the biological front, there are new insights that suggest memories associated with substance use disorder may follow unique neurobiological pathways that render them more accessible to pharmacological intervention. In parallel to this, research in neurochemistry has identified several potential candidate molecules that could influence the formation and maintenance of long-term memory. Drugs that target these molecules (blebbistatin, isradipine and zeta inhibitory peptide) have shown promise at the preclinical stage. In this review, we shall provide an overview of the evolving understanding on the biochemical mechanisms involved in memory formation and expound on the premise that substance use disorder is a learning disorder.
Collapse
Affiliation(s)
- Rashidi Mohamed Pakri Mohamed
- Department of Psychological Medicine, Faculty of Medicine, University of Malaya, Lembah Pantai, 59100 Kuala Lumpur, Malaysia
| | - Jaya Kumar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Ernie Yap
- Department of Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Isa Naina Mohamed
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Hatta Sidi
- Department of Psychiatry, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Raja Lope Adam
- Department of Psychiatry, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Srijit Das
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, 56000 Cheras, Kuala Lumpur, Malaysia
| |
Collapse
|
44
|
Xia X, Fan L, Cheng C, Eickhoff SB, Chen J, Li H, Jiang T. Multimodal connectivity-based parcellation reveals a shell-core dichotomy of the human nucleus accumbens. Hum Brain Mapp 2017; 38:3878-3898. [PMID: 28548226 DOI: 10.1002/hbm.23636] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 04/14/2017] [Accepted: 04/21/2017] [Indexed: 12/21/2022] Open
Abstract
The subdifferentiation of the nucleus accumbens (NAc) has been extensively studied using neuroanatomy and histochemistry, yielding a well-accepted dichotomic shell/core architecture that reflects dissociable roles, such as in reward and aversion, respectively. However, in vivo parcellation of these structures in humans has been rare, potentially impairing future research into the structural and functional characteristics and alterations of putative NAc subregions. Here, we used three complementary parcellation schemes based on tractography, task-independent functional connectivity, and task-dependent co-activation to investigate the regional differentiation within the NAc. We found that a 2-cluster solution with shell-like and core-like subdivisions provided the best description of the data and was consistent with the earlier anatomical shell/core architecture. The consensus clusters from this optimal solution, which was based on the three schemes, were used as the final parcels for the subsequent connection analyses. The resulting connectivity patterns presented inter-hemispheric symmetry, convergence and divergence across the modalities, and, most importantly, clearly distinct patterns between the two subregions. This convergent connectivity patterns also confirmed the connections in animal models, supporting views that the two subregions could have antagonistic roles in some circumstances. Finally, the identified parcels should be helpful in further neuroimaging studies of the NAc. Hum Brain Mapp 38:3878-3898, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Xiaoluan Xia
- College of Computer Science and Technology, Taiyuan University of Technology, Taiyuan, 030600, China
| | - Lingzhong Fan
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Chen Cheng
- College of Computer Science and Technology, Taiyuan University of Technology, Taiyuan, 030600, China
| | - Simon B Eickhoff
- Institute of Neuroscience and Medicine (INM-1), Research Centre Juelich, 52425 Juelich, Germany.,Institute for Clinical Neuroscience and Medical Psychology, Heinrich-Heine-University Düsseldorf, Düsseldorf, 40225, Germany
| | - Junjie Chen
- College of Computer Science and Technology, Taiyuan University of Technology, Taiyuan, 030600, China
| | - Haifang Li
- College of Computer Science and Technology, Taiyuan University of Technology, Taiyuan, 030600, China
| | - Tianzi Jiang
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China.,National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China.,The Queensland Brain Institute, University of Queensland, Brisbane, QLD, 4072, Australia
| |
Collapse
|
45
|
Crews FT, Walter TJ, Coleman LG, Vetreno RP. Toll-like receptor signaling and stages of addiction. Psychopharmacology (Berl) 2017; 234:1483-1498. [PMID: 28210782 PMCID: PMC5420377 DOI: 10.1007/s00213-017-4560-6] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 02/03/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND Athina Markou and her colleagues discovered persistent changes in adult behavior following adolescent exposure to ethanol or nicotine consistent with increased risk for developing addiction. Building on Dr. Markou's important work and that of others in the field, researchers at the Bowles Center for Alcohol Studies have found that persistent changes in behavior following adolescent stress or alcohol exposure may be linked to induction of immune signaling in brain. AIM This study aims to illuminate the critical interrelationship of the innate immune system (e.g., toll-like receptors [TLRs], high-mobility group box 1 [HMGB1]) in the neurobiology of addiction. METHOD This study reviews the relevant research regarding the relationship between the innate immune system and addiction. CONCLUSION Emerging evidence indicates that TLRs in brain, particularly those on microglia, respond to endogenous innate immune agonists such as HMGB1 and microRNAs (miRNAs). Multiple TLRs, HMGB1, and miRNAs are induced in the brain by stress, alcohol, and other drugs of abuse and are increased in the postmortem human alcoholic brain. Enhanced TLR-innate immune signaling in brain leads to epigenetic modifications, alterations in synaptic plasticity, and loss of neuronal cell populations, which contribute to cognitive and emotive dysfunctions. Addiction involves progressive stages of drug binges and intoxication, withdrawal-negative affect, and ultimately compulsive drug use and abuse. Toll-like receptor signaling within cortical-limbic circuits is modified by alcohol and stress in a manner consistent with promoting progression through the stages of addiction.
Collapse
Affiliation(s)
- Fulton T Crews
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| | - T Jordan Walter
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Leon G Coleman
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Ryan P Vetreno
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| |
Collapse
|
46
|
Blum K, Febo M, Badgaiyan RD, Demetrovics Z, Simpatico T, Fahlke C, Oscar-Berman M, Li M, Dushaj K, Gold MS. Common Neurogenetic Diagnosis and Meso-Limbic Manipulation of Hypodopaminergic Function in Reward Deficiency Syndrome (RDS): Changing the Recovery Landscape. Curr Neuropharmacol 2017; 15:184-194. [PMID: 27174576 PMCID: PMC5327445 DOI: 10.2174/1570159x13666160512150918] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 04/11/2016] [Accepted: 04/21/2016] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND In 1990, Blum and associates provided the first confirmed genetic link between the DRD2 polymorphisms and alcoholism. This finding was based on an earlier conceptual framework, which served as a blueprint for their seminal genetic association discovery they termed "Brain Reward Cascade." These findings were followed by a new way of understanding all addictive behaviors (substance and non-substance) termed "Reward Deficiency Syndrome" (RDS). RDS incorporates a complex multifaceted array of inheritable behaviors that are polygenic. OBJECTIVE In this review article, we attempt to clarify these terms and provide a working model to accurately diagnose and treat these unwanted behaviors. METHOD We are hereby proposing the development of a translational model we term "Reward Deficiency Solution System™" that incorporates neurogenetic testing and meso-limbic manipulation of a "hypodopaminergic" trait/state, which provides dopamine agonistic therapy (DAT) as well as reduced "dopamine resistance," while embracing "dopamine homeostasis." RESULT The result is better recovery and relapse prevention, despite DNA antecedents, which could impact the recovery process and relapse. Understanding the commonality of mental illness will transform erroneous labeling based on symptomatology, into a genetic and anatomical etiology. WC: 184.
Collapse
Affiliation(s)
- Kenneth Blum
- Department of Psychiatry, University of Florida College of Medicine and McKnight Brain Institute, Gainesville, FL, USA
- Department of Nutrigenomics, RDSolutions, Inc., Salt Lake City, UT, USA
- Department of Psychology, Eotvos Lorand University, Budapest, Hungary
- PATH Foundation NY, New York, NY, USA
- Division of Neuroscience Research and Addiction Therapy, The Shores Treatment and Recovery, Port Saint Lucie, FL, USA
| | - Marcelo Febo
- Department of Psychiatry, University of Florida College of Medicine and McKnight Brain Institute, Gainesville, FL, USA
| | - Rajendra D. Badgaiyan
- Division of Neuroimaging, Department of Psychiatry, University of Minnesota College of Medicine, Minneapolis, MN, USA;
| | - Zsolt Demetrovics
- Department of Psychology, Eotvos Lorand University, Budapest, Hungary
| | - Thomas Simpatico
- Department of Psychiatry, University of Vermont, Burlington, VT, USA
| | - Claudia Fahlke
- Department of Psychology, University of Gothenburg, Göteborg, Sweden;
| | - Oscar-Berman M
- Departments of Psychiatry and Anatomy & Neurobiology, Boston University School of Medicine and Boston VA Healthcare System, Boston, MA, USA
| | - Mona Li
- PATH Foundation NY, New York, NY, USA
| | | | | |
Collapse
|
47
|
Iurescia S, Seripa D, Rinaldi M. Looking Beyond the 5-HTTLPR Polymorphism: Genetic and Epigenetic Layers of Regulation Affecting the Serotonin Transporter Gene Expression. Mol Neurobiol 2016; 54:8386-8403. [DOI: 10.1007/s12035-016-0304-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 11/16/2016] [Indexed: 01/01/2023]
|
48
|
Mueller M, Wolfs TGA, Schoeberlein A, Gavilanes AWD, Surbek D, Kramer BW. Mesenchymal stem/stromal cells-a key mediator for regeneration after perinatal morbidity? Mol Cell Pediatr 2016; 3:6. [PMID: 26869264 PMCID: PMC4751100 DOI: 10.1186/s40348-016-0034-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 01/19/2016] [Indexed: 02/07/2023] Open
Abstract
Perinatal complications in both term- and preterm-born infants are a leading cause of neonatal morbidities and mortality. Infants face different challenges in the neonatal intensive care unit with long-term morbidities such as perinatal brain injury and bronchopulmonary dysplasia being particularly devastating. While advances in perinatal medicine have improved our understanding of the pathogenesis, effective therapies to prevent and/or reduce the severity of these disorders are still lacking. The potential of mesenchymal stem/stromal cell (MSC) therapy has emerged during the last two decades, and an increasing effort is conducted to address brain- and lung-related morbidities in neonates at risk. Various studies support the notion that MSCs have protective effects. MSCs are an easy source and may be readily available after birth in a clinical setting. MSCs' mechanisms of action are diverse, including migration and homing, release of growth factors and immunomodulation, and the potential to replace injured cells. Here, we review the pathophysiology of perinatally acquired brain and lung injuries and focus on MSCs as potential candidates for therapeutic strategies summarizing preclinical and clinical evidence.
Collapse
Affiliation(s)
- Martin Mueller
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA.
- Department of Obstetrics and Gynecology, University Hospital Bern and Department of Clinical Research, University of Bern, Bern, Switzerland.
| | - Tim G A Wolfs
- Department of Pediatrics, Maastricht University Medical Center (MUMC), Maastricht, The Netherlands.
- School of Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, The Netherlands.
| | - Andreina Schoeberlein
- Department of Obstetrics and Gynecology, University Hospital Bern and Department of Clinical Research, University of Bern, Bern, Switzerland.
| | - Antonio W D Gavilanes
- Department of Pediatrics, Maastricht University Medical Center (MUMC), Maastricht, The Netherlands.
- Institute of Biomedicine, Facultad de Ciencias Médicas, Universidad Católica de Santiago de Guayaquil, Guayaquil, Ecuador.
- Department of Neuropsychology, Division Neuroscience, School of Mental Health and neuroscience (MHeNS), Maastricht University, Maastricht, The Netherlands.
| | - Daniel Surbek
- Department of Obstetrics and Gynecology, University Hospital Bern and Department of Clinical Research, University of Bern, Bern, Switzerland.
| | - Boris W Kramer
- Department of Pediatrics, Maastricht University Medical Center (MUMC), Maastricht, The Netherlands.
- School of Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, The Netherlands.
- Department of Neuropsychology, Division Neuroscience, School of Mental Health and neuroscience (MHeNS), Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
49
|
Weng HL, Wang MJ. Effects of microRNA‑338‑3p on morphine‑induced apoptosis and its underlying mechanisms. Mol Med Rep 2016; 14:2085-92. [PMID: 27432229 DOI: 10.3892/mmr.2016.5506] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 05/25/2016] [Indexed: 11/05/2022] Open
Abstract
The aim of the present study was to investigate the effects of microRNA-338-3p (miR-338-3p) on morphine (MP)-induced apoptosis, and its underlying mechanisms. Freshly‑isolated mouse peritoneal macrophages were cultured in vitro and treated with MP following transfection with miR‑338‑3p mimic, inhibitor or controls. miR‑338‑3p expression levels increased significantly following MP treatment (P<0.01). This increase was enhanced following transfection with miR‑338‑3p mimic (P<0.05) and abrogated following transfection with miR‑338‑3p inhibitor (P<0.05). The apoptotic rate increased significantly in groups treated with MP (P<0.05); however, this increase was abrogated by transfection with miR‑338‑3p inhibitor (P<0.05). Bioinformatics software predicted that sex determining region Y‑box 4 (SOX4) was the target gene of miR‑338‑3p and this was verified using a dual‑luciferase reporter gene system. SOX4 mRNA and protein expression levels decreased significantly following MP treatment (P<0.05); however, this decrease was abrogated following transfection with miR‑338‑3p inhibitor (P<0.05). Caspase‑3 protein expression levels increased markedly following MP treatment (P<0.05); however, this increase was inhibited by transfection with miR‑338‑3p inhibitor (P<0.05). Therefore, decreased expression of miR‑338‑3p may suppress MP‑induced apoptosis, potentially via the upregulation of SOX4 expression and the caspase‑3‑dependent apoptotic signaling pathway.
Collapse
Affiliation(s)
- Hong-Liang Weng
- Department of Anesthesia, Linyi Yishui Central Hospital, Linyi, Shandong 276400, P.R. China
| | - Ming-Jing Wang
- Department of Clinical Laboratory, Linyi Yishui Central Hospital, Linyi, Shandong 276400, P.R. China
| |
Collapse
|
50
|
Yang Y, Zhong N, Imamura K, Lu S, Li M, Zhou H, Li H, Yang X, Wan Z, Wang G, Hu B, Li K. Task and Resting-State fMRI Reveal Altered Salience Responses to Positive Stimuli in Patients with Major Depressive Disorder. PLoS One 2016; 11:e0155092. [PMID: 27192082 PMCID: PMC4871416 DOI: 10.1371/journal.pone.0155092] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 04/25/2016] [Indexed: 02/06/2023] Open
Abstract
Altered brain function in patients with major depressive disorder (MDD) has been repeatedly demonstrated by task-based and resting-state studies, respectively. However, less is known concerning whether overlapped abnormalities in functional activities across modalities exist in MDD patients. To find out the answer, we implemented an fMRI experiment and collected both task and resting-state data from 19 MDD patients and 19 matched, healthy, controls. A distraction paradigm involving emotionally valenced pictures was applied to induce affective responses in subjects. As a result, concurrent deficits were found in arousing activation during a positive task in both the reward circuit and salience network (SN) that is composed of the dorsal part of anterior cingulate cortex (dACC) and bilateral anterior insulae (AI) in only the MDD group. Subsequent amplitude of low frequency fluctuations (ALFF) and functional connectivity analyses based on resting-state data exhibited consistent alterations in the bilateral AI of MDD patients, and indicated patients' difficulties in regulating the balance between central executive network (CEN) and default mode network (DMN) due to altered connectivity among the CEN, DMN, and SN. Our findings provide new evidence demonstrating impaired salience processing and resulting alterations in responses to positive stimuli in MDD patients. Furthermore, brain abnormalities synchronized across functional states in MDD patients can be evidenced by a combination of task and resting-state fMRI analyses.
Collapse
Affiliation(s)
- Yang Yang
- Beijing Advanced Innovation Center for Future Internet Technology, Beijing University of Technology, Beijing, China
- Department of Life Science and Informatics, Maebashi Institute of Technology, Maebashi, Gunma, Japan
- Beijing International Collaboration Base on Brain Informatics and Wisdom Services, Beijing, China
- Beijing Key Laboratory of MRI and Brain Informatics, Beijing, China
| | - Ning Zhong
- Beijing Advanced Innovation Center for Future Internet Technology, Beijing University of Technology, Beijing, China
- Department of Life Science and Informatics, Maebashi Institute of Technology, Maebashi, Gunma, Japan
- International WIC Institute, Beijing University of Technology, Beijing, China
- Beijing International Collaboration Base on Brain Informatics and Wisdom Services, Beijing, China
- Beijing Key Laboratory of MRI and Brain Informatics, Beijing, China
- * E-mail:
| | - Kazuyuki Imamura
- Department of Systems Life Engineering, Maebashi Institute of Technology, Maebashi, Gunma, Japan
| | - Shengfu Lu
- Beijing Advanced Innovation Center for Future Internet Technology, Beijing University of Technology, Beijing, China
- International WIC Institute, Beijing University of Technology, Beijing, China
- Beijing International Collaboration Base on Brain Informatics and Wisdom Services, Beijing, China
- Beijing Key Laboratory of MRI and Brain Informatics, Beijing, China
| | - Mi Li
- Beijing Advanced Innovation Center for Future Internet Technology, Beijing University of Technology, Beijing, China
- International WIC Institute, Beijing University of Technology, Beijing, China
- Beijing International Collaboration Base on Brain Informatics and Wisdom Services, Beijing, China
- Beijing Key Laboratory of MRI and Brain Informatics, Beijing, China
| | - Haiyan Zhou
- Beijing Advanced Innovation Center for Future Internet Technology, Beijing University of Technology, Beijing, China
- International WIC Institute, Beijing University of Technology, Beijing, China
- Beijing International Collaboration Base on Brain Informatics and Wisdom Services, Beijing, China
- Beijing Key Laboratory of MRI and Brain Informatics, Beijing, China
| | - Huaizhou Li
- International WIC Institute, Beijing University of Technology, Beijing, China
- Beijing International Collaboration Base on Brain Informatics and Wisdom Services, Beijing, China
- Beijing Key Laboratory of MRI and Brain Informatics, Beijing, China
| | - Xiaojing Yang
- International WIC Institute, Beijing University of Technology, Beijing, China
- Beijing International Collaboration Base on Brain Informatics and Wisdom Services, Beijing, China
- Beijing Key Laboratory of MRI and Brain Informatics, Beijing, China
| | - Zhijiang Wan
- Beijing Advanced Innovation Center for Future Internet Technology, Beijing University of Technology, Beijing, China
- Department of Life Science and Informatics, Maebashi Institute of Technology, Maebashi, Gunma, Japan
- Beijing International Collaboration Base on Brain Informatics and Wisdom Services, Beijing, China
- Beijing Key Laboratory of MRI and Brain Informatics, Beijing, China
| | - Gang Wang
- Beijing Advanced Innovation Center for Future Internet Technology, Beijing University of Technology, Beijing, China
- Mood Disorders Center, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Bin Hu
- Ubiquitous Awareness and Intelligent Solutions Lab, Lanzhou University, Lanzhou, Gansu, China
| | - Kuncheng Li
- International WIC Institute, Beijing University of Technology, Beijing, China
- Beijing Key Laboratory of MRI and Brain Informatics, Beijing, China
- Department of Radiology, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|