1
|
Lu W, He J, Wei S, Tang C, Ma X, Li D, Chen H, Zou Y. Circular RNA circRest regulates manganese induced cell apoptosis by targeting the mmu-miR-6914-5p/Ephb3 axis. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 344:123395. [PMID: 38266697 DOI: 10.1016/j.envpol.2024.123395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 11/23/2023] [Accepted: 01/17/2024] [Indexed: 01/26/2024]
Abstract
Overexposure to manganese (Mn) can lead to neurotoxicity, the underlying mechanisms remain incompletely understood. Circular RNAs (circRNAs) have emerged as important regulators in various biological processes. It is plausible that circRNAs may be involved in the biological mechanisms underlying Mn caused neurotoxicity. Here, circRest was downregulated in Mn-exposed mouse neuroblastoma cells (N2a cells) by RNA sequencing and quantitative real-time PCR. When circRest was overexpressed, it led to an increase in cell viability and a decrease in apoptosis following Mn exposure. Conversely, silencing circRest resulted in opposite effects in N2a cells. Further investigation revealed that circRest acts as a mmu-miR-6914-5p sponge, and mmu-miR-6914-5p could bind and inhibit Ephb3, thereby promoting apoptosis in N2a cells. This was confirmed through RNA antisense purification and dual luciferase reporter assays. Additionally, the circRest/mmu-miR-6914-5p/Ephb3 axis may influence memory and learning in mice following Mn exposure. In conclusion, our study uncovers a novel mechanism by which circRest may attenuate Mn caused neurotoxicity via the mmu-miR-6914-5p/Ephb3 axis.
Collapse
Affiliation(s)
- Wenmin Lu
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Jiacheng He
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Shengtao Wei
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Chuanqiao Tang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Xiaoli Ma
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Danni Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Hao Chen
- Department of Occupational and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, 530021, China
| | - Yunfeng Zou
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, Guangxi, China; Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Nanning, 530021, Guangxi, China.
| |
Collapse
|
2
|
Beylerli O, Beilerli A, Ilyasova T, Shumadalova A, Shi H, Sufianov A. CircRNAs in Alzheimer's disease: What are the prospects? Noncoding RNA Res 2024; 9:203-210. [PMID: 38125754 PMCID: PMC10730436 DOI: 10.1016/j.ncrna.2023.11.011] [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: 07/30/2023] [Revised: 11/18/2023] [Accepted: 11/22/2023] [Indexed: 12/23/2023] Open
Abstract
Circular RNAs (circRNAs) is a fascinating covalently closed circular non-coding RNA that is abundantly present in the transcriptome of eukaryotic cells. Its versatile nature allows it to participate in a multitude of pathological and physiological processes within the organism. One of its crucial functions is acting as a microRNA sponge, modulating protein transcription levels, and forming interactions with essential RNA-binding proteins. Remarkably, circRNAs demonstrates a specific enrichment in various vital areas of the brain, including the cortex, hippocampus, white matter, and photoreceptor neurons, particularly in aging organisms. This intriguing characteristic has led scientists to explore its potential as a significant biological marker of neurodegeneration, offering promising insights into neurodegenerative diseases like Alzheimer's disease (AD). In AD, there has been an interesting observation of elevated levels of circRNAs in both peripheral blood and synaptic terminals of affected individuals. This intriguing finding raises the possibility that circRNAs may have a central role in the initiation and progression of AD. Notably, different categories of circRNAs, including HDAC9, HOMER1, Cwc27, Tulp4, and PTK2, have been implicated in driving the pathological changes associated with AD through diverse mechanisms. For instance, these circRNAs have been demonstrated to contribute to the accumulation of beta-amyloid, which is a hallmark characteristic of AD. Additionally, these circRNAs contribute to the excessive phosphorylation of tau protein, a phenomenon associated with neurofibrillary tangles, further exacerbating the disease. Moreover, they are involved in aggravating neuroinflammation, which is known to play a critical role in AD's pathogenesis. Lastly, these circRNAs can cause mitochondrial dysfunction, disrupting cellular energy production and leading to cognitive impairment. As researchers delve deeper into the intricate workings of circRNAs, they hope to unlock its full potential as a diagnostic tool and therapeutic target for neurodegenerative disorders, paving the way for innovative treatments and better management of such devastating conditions.
Collapse
Affiliation(s)
- Ozal Beylerli
- Central Research Laboratory, Bashkir State Medical University, Ufa, Republic of Bashkortostan, 3 Lenin Street, 450008, Russia
| | - Aferin Beilerli
- Department of Obstetrics and Gynecology, Tyumen State Medical University, 54 Odesskaya Street, 625023, Tyumen, Russia
| | - Tatiana Ilyasova
- Department of Internal Diseases, Bashkir State Medical University, Ufa, Republic of Bashkortostan, 450008, Russia
| | - Alina Shumadalova
- Department of General Chemistry, Bashkir State Medical University, Ufa, Republic of Bashkortostan, 3 Lenin Street, 450008, Russia
| | - Huaizhang Shi
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Albert Sufianov
- Educational and Scientific Institute of Neurosurgery, Рeoples’ Friendship University of Russia (RUDN University), Moscow, Russia
- Department of Neurosurgery, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| |
Collapse
|
3
|
Dabrowski KR, Floris G, Gillespie A, Daws SE. Orbitofrontal intronic circular RNA from Nrxn3 mediates reward learning and motivation for reward. Prog Neurobiol 2024; 232:102546. [PMID: 38036039 PMCID: PMC10843848 DOI: 10.1016/j.pneurobio.2023.102546] [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: 07/17/2023] [Revised: 10/27/2023] [Accepted: 11/20/2023] [Indexed: 12/02/2023]
Abstract
The orbitofrontal cortex (OFC) is a vital component of brain reward circuitry that is important for reward seeking behavior. However, OFC-mediated molecular mechanisms underlying rewarding behavior are understudied. Here, we report the first circular RNA (circRNA) profile associated with appetitive reward and identify regulation of 92 OFC circRNAs by sucrose self-administration. Among these changes, we observed downregulation of circNrxn3, a circRNA originating from neurexin 3 (Nrxn3), a gene involved in synaptogenesis, learning, and memory. Transcriptomic profiling via RNA sequencing and qPCR of the OFC following in vivo knock-down of circNrxn3 revealed differential regulation of genes associated with pathways important for learning and memory and altered splicing of Nrxn3. Furthermore, circNrxn3 knock-down enhanced sucrose self-administration and motivation for sucrose. Using RNA-immunoprecipitation, we report binding of circNrxn3 to the known Nrxn3 splicing factor SAM68. circNrxn3 is the first reported circRNA capable of regulating reward behavior and circNrxn3-mediated interactions with SAM68 may impact subsequent downstream processing of RNAs such as the regulation of gene expression and splicing.
Collapse
Affiliation(s)
- Konrad R Dabrowski
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA; Department of Biology, Temple University, Philadelphia, PA, USA
| | - Gabriele Floris
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA; Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Aria Gillespie
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA; Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Stephanie E Daws
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA; Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
4
|
Puri S, Hu J, Sun Z, Lin M, Stein TD, Farrer LA, Wolozin B, Zhang X. Identification of circRNAs linked to Alzheimer's disease and related dementias. Alzheimers Dement 2023; 19:3389-3405. [PMID: 36795937 PMCID: PMC10427739 DOI: 10.1002/alz.12960] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 12/21/2022] [Accepted: 12/27/2022] [Indexed: 02/18/2023]
Abstract
INTRODUCTION Circular RNAs (circRNAs) exhibit selective expression in the brain and differential regulation in Alzheimer's disease (AD). To explore the role of circRNAs in AD, we investigated how circRNA expression varies between brain regions and with AD-related stress in human neuronal precursor cells (NPCs). METHODS Ribosomal RNA-depleted hippocampus RNA-sequencing data were generated. Differentially regulated circRNAs in AD and related dementias were detected using CIRCexplorer3 and limma. circRNA results were validated using quantitative real-time PCR of cDNA from the brain and NPCs. RESULTS We identified 48 circRNAs that were significantly associated with AD. We observed that circRNA expression differed by dementia subtype. Using NPCs, we demonstrated that exposure to oligomeric tau elicits downregulation of circRNA similar to that observed in the AD brain. DISCUSSION Our study shows that differential expression of circRNA can vary by dementia subtype and brain region. We also demonstrated that circRNAs can be regulated by AD-linked neuronal stress independently from their cognate linear messenger RNAs (mRNAs).
Collapse
Affiliation(s)
- Sambhavi Puri
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Junming Hu
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA, USA
| | - Zhuorui Sun
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA, USA
| | - Mintao Lin
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA, USA
| | - Thor D. Stein
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
- Alzheimer’s Disease Research Center, Boston University School of Medicine, Boston, MA, USA
- Framingham Heart Study, Boston University School of Medicine, Framingham, MA, USA
- VA Boston Healthcare System, Boston, MA, USA
| | - Lindsay A. Farrer
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA, USA
- Department of Ophthalmology, Boston University School of Medicine, Boston, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
- Alzheimer’s Disease Research Center, Boston University School of Medicine, Boston, MA, USA
- Framingham Heart Study, Boston University School of Medicine, Framingham, MA, USA
| | - Benjamin Wolozin
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Xiaoling Zhang
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- Framingham Heart Study, Boston University School of Medicine, Framingham, MA, USA
| |
Collapse
|
5
|
Daws SE, Gillespie A. Circular RNA regulation and function in drug seeking phenotypes. Mol Cell Neurosci 2023; 125:103841. [PMID: 36935046 PMCID: PMC10247439 DOI: 10.1016/j.mcn.2023.103841] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/27/2023] [Accepted: 03/06/2023] [Indexed: 03/19/2023] Open
Abstract
Drug overdoses have increased dramatically in the United States over the last decade where they are now the leading cause of accidental death. To develop efficient therapeutic options for decreasing drug consumption and overdose risk, it is critical to understand the neurobiological changes induced by drug exposure. Chronic systemic exposure to all drug classes, including opioids, psychostimulants, nicotine, cannabis, and alcohol, induces profound molecular neuroadaptations within the central nervous system that may reveal crucial information about the lasting effects that these substances impart on brain cells. Transcriptome analyses of messenger RNAs (mRNAs) have identified gene patterns in the brain that result from exposure to various classes of drugs. However, mRNAs represent only a small fraction of the RNA within the cell, and drug exposure also impacts other classes of RNA that are largely understudied, especially circular RNAs. Circular RNAs (circRNAs) are a naturally occurring RNA species formed from back-splicing events during mRNA processing and are enriched in the nervous system. circRNAs are a pleiotropic class of RNAs and have a diverse impact on cellular function, with putative functions including regulation of mRNA transcription, protein translation, microRNA sponging, and sequestration of RNA-binding proteins. Recent studies have demonstrated that circRNAs can modulate cognition and are regulated in the brain in response to drug exposure, yet very few studies have explored the contribution of circRNAs to drug seeking phenotypes. In this review, we will provide an overview of the mechanisms of circRNA function in the cell to highlight how drug-induced circRNA dysregulation may impact the molecular substrates that mediate drug seeking behavior and the current studies that have reported drug-induced dysregulation of circRNAs in the brain. Furthermore, we will discuss how principles of circRNA biology can be adapted to study circRNAs in models of drug exposure and seek to provide further insight into the neurobiology of addiction.
Collapse
Affiliation(s)
- Stephanie E Daws
- Center for Substance Abuse Research, Temple University, Philadelphia, PA, USA; Department of Neural Sciences, Temple University, Philadelphia, PA, USA.
| | - Aria Gillespie
- Center for Substance Abuse Research, Temple University, Philadelphia, PA, USA; Department of Neural Sciences, Temple University, Philadelphia, PA, USA
| |
Collapse
|
6
|
Li M, Li J, Ji M, An J, Zhao T, Yang Y, Cai C, Gao P, Cao G, Guo X, Li B. CircHOMER1 inhibits porcine adipogenesis via the miR-23b/SIRT1 axis. FASEB J 2023; 37:e22828. [PMID: 36809667 DOI: 10.1096/fj.202202048rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 02/23/2023]
Abstract
Fat deposition is critical to pork quality. However, the mechanism of fat deposition remains to be elucidated. Circular RNAs (circRNAs) are ideal biomarkers and are involved in adipogenesis. Here, we investigated the effect and mechanism of circHOMER1 on porcine adipogenesis in vitro and in vivo. Western blotting, Oil red O staining, and HE staining were used to assess the function of circHOMER1 in adipogenesis. The results showed that circHOMER1 inhibited adipogenic differentiation of porcine preadipocytes and suppressed adipogenesis in mice. Dual-luciferase reporter gene, RIP, and pull-down assays demonstrated that miR-23b directly bound to circHOMER1 and the 3'-UTR of SIRT1. Rescue experiments further illustrated the regulatory relationship among circHOMER1, miR-23b, and SIRT1. Conclusively, we demonstrate that circHOMER1 plays an inhibitory role in porcine adipogenesis through miR-23b and SIRT1. The present study revealed the mechanism of porcine adipogenesis, which may be helpful to improve pork quality.
Collapse
Affiliation(s)
- Meng Li
- College of Animal Science, Shanxi Agricultural University, Taigu, China
| | - Jiao Li
- College of Animal Science, Shanxi Agricultural University, Taigu, China
| | - Mengting Ji
- College of Animal Science, Shanxi Agricultural University, Taigu, China
| | - Jiaqi An
- College of Animal Science, Shanxi Agricultural University, Taigu, China
| | - Tianzhi Zhao
- College of Animal Science, Shanxi Agricultural University, Taigu, China
| | - Yang Yang
- College of Animal Science, Shanxi Agricultural University, Taigu, China
| | - Chunbo Cai
- College of Animal Science, Shanxi Agricultural University, Taigu, China
| | - Pengfei Gao
- College of Animal Science, Shanxi Agricultural University, Taigu, China
| | - Guoqing Cao
- College of Animal Science, Shanxi Agricultural University, Taigu, China
| | - Xiaohong Guo
- College of Animal Science, Shanxi Agricultural University, Taigu, China
| | - Bugao Li
- College of Animal Science, Shanxi Agricultural University, Taigu, China
| |
Collapse
|
7
|
Seyednejad SA, Sartor GC. Noncoding RNA therapeutics for substance use disorder. ADVANCES IN DRUG AND ALCOHOL RESEARCH 2022; 2:10807. [PMID: 36601439 PMCID: PMC9808746 DOI: 10.3389/adar.2022.10807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Although noncoding RNAs (ncRNAs) have been shown to regulate maladaptive neuroadaptations that drive compulsive drug use, ncRNA-targeting therapeutics for substance use disorder (SUD) have yet to be clinically tested. Recent advances in RNA-based drugs have improved many therapeutic issues related to immune response, specificity, and delivery, leading to multiple successful clinical trials for other diseases. As the need for safe and effective treatments for SUD continues to grow, novel nucleic acid-based therapeutics represent an appealing approach to target ncRNA mechanisms in SUD. Here, we review ncRNA processes implicated in SUD, discuss recent therapeutic approaches for targeting ncRNAs, and highlight potential opportunities and challenges of ncRNA-targeting therapeutics for SUD.
Collapse
Affiliation(s)
- Seyed Afshin Seyednejad
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, United States
- Connecticut Institute for the Brain and Cognitive Sciences (CT IBACS), Storrs, CT, United States
| | - Gregory C. Sartor
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, United States
- Connecticut Institute for the Brain and Cognitive Sciences (CT IBACS), Storrs, CT, United States
| |
Collapse
|
8
|
Methamphetamine induced neurotoxic diseases, molecular mechanism, and current treatment strategies. Biomed Pharmacother 2022; 154:113591. [PMID: 36007276 DOI: 10.1016/j.biopha.2022.113591] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/18/2022] [Accepted: 08/18/2022] [Indexed: 11/21/2022] Open
Abstract
Methamphetamine (MA) is a extremely addictive psychostimulant drug with a significant abuse potential. Long-term MA exposure can induce neurotoxic effects through oxidative stress, mitochondrial functional impairment, endoplasmic reticulum stress, the activation of astrocytes and microglial cells, axonal transport barriers, autophagy, and apoptosis. However, the molecular and cellular mechanisms underlying MA-induced neurotoxicity remain unclear. MA abuse increases the chances of developing neurotoxic conditions such as Parkinson's disease (PD), Alzheimer's disease (AD) and other neurotoxic diseases. MA increases the risk of PD by increasing the expression of alpha-synuclein (ASYN). Furthermore, MA abuse is linked to high chances of developing AD and subsequent neurodegeneration due to biological variations in the brain region or genetic and epigenetic variations. To date, there is no Food and Drug Administration (FDA)-approved therapy for MA-induced neurotoxicity, although many studies are being conducted to develop effective therapeutic strategies. Most current studies are now focused on developing therapies to diminish the neurotoxic effects of MA, based on the underlying mechanism of neurotoxicity. This review article highlights current research on several therapeutic techniques targeting multiple pathways to reduce the neurotoxic effects of MA in the brain, as well as the putative mechanism of MA-induced neurotoxicity.
Collapse
|
9
|
Inactivation of the Lateral Hypothalamus Attenuates Methamphetamine-Induced Conditioned Place Preference through Regulation of Kcnq3 Expression. Int J Mol Sci 2022; 23:ijms23137305. [PMID: 35806315 PMCID: PMC9266452 DOI: 10.3390/ijms23137305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/25/2022] [Accepted: 06/26/2022] [Indexed: 12/03/2022] Open
Abstract
Repeated administration of methylamphetamine (MA) induces MA addiction, which is featured by awfully unpleasant physical and emotional experiences after drug use is terminated. Neurophysiological studies show that the lateral hypothalamus (LH) is involved in reward development and addictive behaviors. Here, we show that repeated administration of MA activates the expression of c-Fos in LH neurons responding to conditioned place preference (CPP). Chemogenetic inhibition of the LH can disrupt the addiction behavior, demonstrating that the LH plays an important role in MA-induced reward processing. Critically, MA remodels the neurons of LH synaptic plasticity, increases intracellular calcium level, and enhances spontaneous current and evoked potentials of neurons compared to the saline group. Furthermore, overexpression of the potassium voltage-gated channel subfamily Q member 3 (Kcnq3) expression can reverse the CPP score and alleviate the occurrence of addictive behaviors. Together, these results unravel a new neurobiological mechanism underlying the MA-induced addiction in the lateral hypothalamus, which could pave the way toward new and effective interventions for this addiction disease.
Collapse
|
10
|
Chen Y, Li X, Meng S, Huang S, Chang S, Shi J. Identification of Functional CircRNA–miRNA–mRNA Regulatory Network in Dorsolateral Prefrontal Cortex Neurons of Patients With Cocaine Use Disorder. Front Mol Neurosci 2022; 15:839233. [PMID: 35493321 PMCID: PMC9048414 DOI: 10.3389/fnmol.2022.839233] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 03/01/2022] [Indexed: 11/25/2022] Open
Abstract
Increasing evidence has indicated that circular RNAs (circRNAs) act as competing endogenous RNAs (ceRNAs) regulatory network to regulate the expression of target genes by sponging microRNAs (miRNAs), and therefore play an essential role in many neuropsychiatric disorders, including cocaine use disorder. However, the functional roles and regulatory mechanisms of circRNAs as ceRNAs in dorsolateral prefrontal cortex (dlPFC) of patients with cocaine use disorder remain to be determined. In this study, an expression profiling for dlPFC in 19 patients with cocaine use disorder and 17 controls from Gene Expression Omnibus datasets was used for the differentially expressed circRNAs analysis and the differentially expressed mRNAs analysis. Several tools were used to predict the miRNAs targeted by the circRNAs and the miRNAs targeted mRNAs, which then overlapped with the cocaine-associated differentially expressed mRNAs to determine the functional roles of circRNAs. Functional analysis for the obtained mRNAs was performed via Gene Ontology (GO) in Metascape database. Integrated bioinformatics analysis was conducted to further characterize the circRNA–miRNA–mRNA regulatory network and identify the functions of distinct circRNAs. We found a total of 41 differentially expressed circRNAs, and 98 miRNAs were targeted by these circRNAs. The overlapped mRNAs targeted by the miRNAs and the differentially expressed mRNAs constructed a circRNA–miRNA–mRNA regulation network including 24 circRNAs, 43 miRNAs, and 82 mRNAs in the dlPFC of patients with cocaine use disorder. Functional analysis indicated the regulation network mainly participated in cell response-related, receptor signaling-related, protein modification-related and axonogenesis-related pathways, which might be involved with cocaine use disorder. Additionally, we determined four hub genes (HSP90AA1, HSPA1B, YWHAG, and RAB8A) from the protein–protein interaction network and constructed a circRNA–miRNA-hub gene subnetwork based on the four hub genes. In conclusion, our findings provide a deeper understanding of the circRNAs-related ceRNAs regulatory mechanisms in the pathogenesis of cocaine use disorder.
Collapse
Affiliation(s)
- Yun Chen
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Beijing Key Laboratory on Drug Dependence Research, National Institute on Drug Dependence, Peking University, Beijing, China
| | - Xianfeng Li
- Department of Gastroenterology of Dapping Hospital, Third Military Medical University, Chongqing, China
| | - Shiqiu Meng
- Beijing Key Laboratory on Drug Dependence Research, National Institute on Drug Dependence, Peking University, Beijing, China
| | - Shihao Huang
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Suhua Chang
- Institute of Mental Health, National Clinical Research Center for Mental Disorders, Key Laboratory of Mental Health and Peking University Sixth Hospital, Peking University, Beijing, China
- Suhua Chang,
| | - Jie Shi
- Beijing Key Laboratory on Drug Dependence Research, National Institute on Drug Dependence, Peking University, Beijing, China
- Peking University, Shenzhen Hospital, Shenzhen, China
- *Correspondence: Jie Shi,
| |
Collapse
|
11
|
Li J, Sun C, Cui H, Sun J, Zhou P. Role of circRNAs in neurodevelopment and neurodegenerative diseases. J Mol Neurosci 2021; 71:1743-1751. [PMID: 34378140 DOI: 10.1007/s12031-021-01882-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 06/01/2021] [Indexed: 12/14/2022]
Abstract
With the rapid development of sequencing technology, scientists have been able to study and acquire a better understanding of non-coding RNAs (ncRNAs). Circular RNAs (circRNAs), a unique class of ncRNAs with a special loop structure, have been found to possess modulatory properties with respect to various biological processes, such as interacting with nucleic acids or proteins. In addition to their tissue-specific expression and high conservation across species, circRNAs are abundant and dynamically expressed in the nervous system, especially in nerve synapse, indicating their potential regulation in synaptic plasticity or neuronal disorders. In this review, we discuss the characteristics of circRNAs and their common biological functions, as well as their significant role in neurodevelopment, drug addiction and neurodegenerative diseases, aiming to guide further disease diagnoses and efficient therapy.
Collapse
Affiliation(s)
- Junwei Li
- Department of Anatomy, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.,Institute of Neuroscience, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chenyou Sun
- Department of Anatomy, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.,Institute of Neuroscience, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Huairui Cui
- Department of Anatomy, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jinhao Sun
- Department of Anatomy, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Peng Zhou
- Department of Anatomy, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China. .,Institute of Neuroscience, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
12
|
Zhu Z, Shen S, Zhao S, Wang Z. Hsa_circ_0006916 Knockdown Represses the Development of Hepatocellular Carcinoma via Modulating miR-599/SRSF2 Axis. Onco Targets Ther 2020; 13:11301-11313. [PMID: 33177838 PMCID: PMC7649248 DOI: 10.2147/ott.s267471] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 09/22/2020] [Indexed: 12/12/2022] Open
Abstract
Background The aberrantly expressed circular RNAs (circRNAs) are implicated in the progression of hepatocellular carcinoma (HCC). CircRNA hsa_circ_0006916 (circ_0006916) is dysregulated in HCC, but the function and mechanism of this circRNA in HCC development remain uncertain. Methods Thirty paired HCC and normal tissues were collected. circ_0006916, microRNA (miR)-599 and serine/arginine rich splicing factor 2 (SRSF2) abundances were examined via quantitative reverse transcription polymerase chain reaction or Western blot. Cell viability, colony ability, migration, invasion, cell cycle and apoptosis were tested via cell counting kit-8, colony formation, wound healing analysis, transwell analysis, and flow cytometry. The interaction between miR-599 and circ_0006916 or SRSF2 was analyzed via dual-luciferase reporter and RNA immunoprecipitation analyses. The function of circ_0006916 on cell growth in vivo was analyzed via xenograft model. Results circ_0006916 expression was increased in HCC tissues and cell lines. circ_0006916 knockdown reduced cell viability, colony formation, migration and invasion and caused cell cycle arrest and apoptosis. miR-599 was targeted via circ_0006916, and miR-599 knockdown reversed the influence of circ_0006916 silence on HCC progression. SRSF2 was targeted via miR-599, and miR-599 overexpression suppressed cell viability, colony formation, migration and invasion and promoted cell cycle arrest and apoptosis via decreasing SRSF2. circ_0006916 could regulate SRSF2 expression via miR-599. circ_0006916 knockdown decreased HCC cell growth in the xenograft model. Conclusion circ_0006916 knockdown represses the progression of HCC via regulating miR-599 and SRSF2.
Collapse
Affiliation(s)
- Zhixiang Zhu
- Image Center, The First Affiliated Hospital of Henan University, Kaifeng, Henan 475001, People's Republic of China
| | - Songhe Shen
- Image Center, The First Affiliated Hospital of Henan University, Kaifeng, Henan 475001, People's Republic of China
| | - Sen Zhao
- Image Center, The First Affiliated Hospital of Henan University, Kaifeng, Henan 475001, People's Republic of China
| | - Zhixue Wang
- Image Center, The First Affiliated Hospital of Henan University, Kaifeng, Henan 475001, People's Republic of China
| |
Collapse
|