1
|
Darbinian N, Hampe M, Martirosyan D, Bajwa A, Darbinyan A, Merabova N, Tatevosian G, Goetzl L, Amini S, Selzer ME. Fetal Brain-Derived Exosomal miRNAs from Maternal Blood: Potential Diagnostic Biomarkers for Fetal Alcohol Spectrum Disorders (FASDs). Int J Mol Sci 2024; 25:5826. [PMID: 38892014 PMCID: PMC11172088 DOI: 10.3390/ijms25115826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/20/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
Fetal alcohol spectrum disorders (FASDs) are leading causes of neurodevelopmental disability but cannot be diagnosed early in utero. Because several microRNAs (miRNAs) are implicated in other neurological and neurodevelopmental disorders, the effects of EtOH exposure on the expression of these miRNAs and their target genes and pathways were assessed. In women who drank alcohol (EtOH) during pregnancy and non-drinking controls, matched individually for fetal sex and gestational age, the levels of miRNAs in fetal brain-derived exosomes (FB-Es) isolated from the mothers' serum correlated well with the contents of the corresponding fetal brain tissues obtained after voluntary pregnancy termination. In six EtOH-exposed cases and six matched controls, the levels of fetal brain and maternal serum miRNAs were quantified on the array by qRT-PCR. In FB-Es from 10 EtOH-exposed cases and 10 controls, selected miRNAs were quantified by ddPCR. Protein levels were quantified by ELISA. There were significant EtOH-associated reductions in the expression of several miRNAs, including miR-9 and its downstream neuronal targets BDNF, REST, Synapsin, and Sonic hedgehog. In 20 paired cases, reductions in FB-E miR-9 levels correlated strongly with reductions in fetal eye diameter, a prominent feature of FASDs. Thus, FB-E miR-9 levels might serve as a biomarker to predict FASDs in at-risk fetuses.
Collapse
Affiliation(s)
- Nune Darbinian
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (M.H.); (D.M.); (A.B.); (N.M.); (G.T.)
| | - Monica Hampe
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (M.H.); (D.M.); (A.B.); (N.M.); (G.T.)
| | - Diana Martirosyan
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (M.H.); (D.M.); (A.B.); (N.M.); (G.T.)
| | - Ahsun Bajwa
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (M.H.); (D.M.); (A.B.); (N.M.); (G.T.)
| | - Armine Darbinyan
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06520, USA;
| | - Nana Merabova
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (M.H.); (D.M.); (A.B.); (N.M.); (G.T.)
- Medical College of Wisconsin-Prevea Health, Green Bay, WI 54304, USA
| | - Gabriel Tatevosian
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (M.H.); (D.M.); (A.B.); (N.M.); (G.T.)
| | - Laura Goetzl
- Department of Obstetrics & Gynecology, University of Texas, Houston, TX 77030, USA;
| | - Shohreh Amini
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA;
| | - Michael E. Selzer
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (M.H.); (D.M.); (A.B.); (N.M.); (G.T.)
- Department of Neurology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
2
|
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
|
3
|
Bake S, Rouzer SK, Mavuri S, Miranda RC, Mahnke AH. The interaction of genetic sex and prenatal alcohol exposure on health across the lifespan. Front Neuroendocrinol 2023; 71:101103. [PMID: 37802472 PMCID: PMC10922031 DOI: 10.1016/j.yfrne.2023.101103] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 09/22/2023] [Accepted: 10/03/2023] [Indexed: 10/10/2023]
Abstract
Prenatal alcohol exposure (PAE) can reprogram the development of cells and tissues, resulting in a spectrum of physical and neurobehavioral teratology. PAE immediately impacts fetal growth, but its effects carry forward post-parturition, into adolescence and adulthood, and can result in a cluster of disabilities, collectively termed Fetal Alcohol Spectrum Disorders. Emerging preclinical and clinical research investigating neurological and behavioral outcomes in exposed offspring point to genetic sex as an important modifier of the effects of PAE. In this review, we discuss the literature on sex differences following PAE, with studies spanning the fetal period through adulthood, and highlight gaps in research where sex differences are likely, but currently under-investigated. Understanding how sex and PAE interact to affect offspring health outcomes across the lifespan is critical for identifying the full complement of PAE-associated secondary conditions, and for refining targeted interventions to improve the quality of life for individuals with PAE.
Collapse
Affiliation(s)
- Shameena Bake
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Medical Research and Education Building I, 8447 Riverside Parkway, Bryan, TX 77807-3620, United States
| | - Siara K Rouzer
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Medical Research and Education Building I, 8447 Riverside Parkway, Bryan, TX 77807-3620, United States
| | - Shruti Mavuri
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Medical Research and Education Building I, 8447 Riverside Parkway, Bryan, TX 77807-3620, United States
| | - Rajesh C Miranda
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Medical Research and Education Building I, 8447 Riverside Parkway, Bryan, TX 77807-3620, United States
| | - Amanda H Mahnke
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Medical Research and Education Building I, 8447 Riverside Parkway, Bryan, TX 77807-3620, United States.
| |
Collapse
|
4
|
Hajiasgharzadeh K, Naghipour B, Shahabi P, Dastmalchi N, Alipour MR. The role of microRNAs in nicotine signaling. EXCLI JOURNAL 2023; 22:433-450. [PMID: 37346805 PMCID: PMC10279964 DOI: 10.17179/excli2023-6096] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 05/15/2023] [Indexed: 06/23/2023]
Abstract
Cigarette smoking is a harmful habit that is widespread around the world. It is among the well-known lifestyle-related risk factors for many diseases. Nicotine, as its principal constituent, has various detrimental, and beneficial functions. Nicotinic acetylcholine receptors (nAChRs), which are present in nearly all body cells, are how nicotine works. Numerous investigations have demonstrated that nicotine causes abnormal microRNA expression (miRNAs). These short sequences of RNAs are known to regulate gene expression post-transcriptionally. A wide range of miRNAs are modulated by nicotine, and nicotine-induced miRNA changes could subsequently mediate nicotine's effect on gene expression regulation. We will focus on the reciprocal interaction between nAChRs and miRNAs and describe the essential targets of these dysregulated miRNAs after nicotine exposure and activation of nAChRs. It appears that crucial subcellular mechanisms implicated in nicotine's effects are miRNA-related pathways. It is crucial to investigate the molecular mechanism underlying the effects of nicotine as well as the dysregulation of miRNA following nAChR activation. The finding about epigenetic mechanisms of nicotine-induced effects may shed light on the establishment of new treatment strategies to prevent the harmful effects of nicotine and perhaps may augment the beneficial effects in diverse smoking-related diseases.
Collapse
Affiliation(s)
| | - Bahman Naghipour
- Department of Anesthesiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parviz Shahabi
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Narges Dastmalchi
- Department of Biology, University College of Nabi Akram, Tabriz, Iran
| | - Mohammad Reza Alipour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
5
|
Chung DD, Mahnke AH, Pinson MR, Salem NA, Lai MS, Collins NP, Hillhouse AE, Miranda RC. Sex differences in the transcriptome of extracellular vesicles secreted by fetal neural stem cells and effects of chronic alcohol exposure. Biol Sex Differ 2023; 14:19. [PMID: 37060018 PMCID: PMC10105449 DOI: 10.1186/s13293-023-00503-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 04/04/2023] [Indexed: 04/16/2023] Open
Abstract
BACKGROUND Prenatal alcohol (ethanol) exposure (PAE) results in brain growth restriction, in part, by reprogramming self-renewal and maturation of fetal neural stem cells (NSCs) during neurogenesis. We recently showed that ethanol resulted in enrichment of both proteins and pro-maturation microRNAs in sub-200-nm-sized extracellular vesicles (EVs) secreted by fetal NSCs. Moreover, EVs secreted by ethanol-exposed NSCs exhibited diminished efficacy in controlling NSC metabolism and maturation. Here we tested the hypothesis that ethanol may also influence the packaging of RNAs into EVs from cell-of-origin NSCs. METHODS Sex-specified fetal murine iso-cortical neuroepithelia from three separate pregnancies were maintained ex vivo, as neurosphere cultures to model the early neurogenic niche. EVs were isolated by ultracentrifugation from NSCs exposed to a dose range of ethanol. RNA from paired EV and cell-of-origin NSC samples was processed for ribosomal RNA-depleted RNA sequencing. Differential expression analysis and exploratory weighted gene co-expression network analysis (WGCNA) identified candidate genes and gene networks that were drivers of alterations to the transcriptome of EVs relative to cells. RESULTS The RNA content of EVs differed significantly from cell-of-origin NSCs. Biological sex contributed to unique transcriptome variance in EV samples, where > 75% of the most variant transcripts were also sex-variant in EVs but not in cell-of-origin NSCs. WGCNA analysis also identified sex-dependent enrichment of pathways, including dopamine receptor binding and ectoderm formation in female EVs and cell-substrate adhesion in male EVs, with the top significant DEGs from differential analysis of overall individual gene expressions, i.e., Arhgap15, enriched in female EVs, and Cenpa, enriched in male EVs, also serving as WCGNA hub genes of sex-biased EV WGCNA clusters. In addition to the baseline RNA content differences, ethanol exposure resulted in a significant dose-dependent change in transcript expression in both EVs and cell-of-origin NSCs that predominantly altered sex-invariant RNAs. Moreover, at the highest dose, ~ 73% of significantly altered RNAs were enriched in EVs, but depleted in NSCs. CONCLUSIONS The EV transcriptome is distinctly different from, and more sex-variant than, the transcriptome of cell-of-origin NSCs. Ethanol, a common teratogen, results in dose-dependent sorting of RNA transcripts from NSCs to EVs which may reprogram the EV-mediated endocrine environment during neurogenesis.
Collapse
Affiliation(s)
- Dae D Chung
- School of Medicine, Department of Neuroscience and Experimental Therapeutics, Medical Research and Education, Texas A&M University Health Science Center, Building 8447 Riverside Parkway, Bryan, TX, 77807-3260, USA
| | - Amanda H Mahnke
- School of Medicine, Department of Neuroscience and Experimental Therapeutics, Medical Research and Education, Texas A&M University Health Science Center, Building 8447 Riverside Parkway, Bryan, TX, 77807-3260, USA
- Women's Health in Neuroscience, Texas A&M University Health Science Center, Bryan, TX, USA
| | - Marisa R Pinson
- School of Medicine, Department of Neuroscience and Experimental Therapeutics, Medical Research and Education, Texas A&M University Health Science Center, Building 8447 Riverside Parkway, Bryan, TX, 77807-3260, USA
| | - Nihal A Salem
- School of Medicine, Department of Neuroscience and Experimental Therapeutics, Medical Research and Education, Texas A&M University Health Science Center, Building 8447 Riverside Parkway, Bryan, TX, 77807-3260, USA
| | - Michael S Lai
- School of Medicine, Department of Neuroscience and Experimental Therapeutics, Medical Research and Education, Texas A&M University Health Science Center, Building 8447 Riverside Parkway, Bryan, TX, 77807-3260, USA
| | - Natalie P Collins
- School of Medicine, Department of Neuroscience and Experimental Therapeutics, Medical Research and Education, Texas A&M University Health Science Center, Building 8447 Riverside Parkway, Bryan, TX, 77807-3260, USA
| | - Andrew E Hillhouse
- Texas A&M Institute for Genome Sciences and Society, Texas A&M University, College Station, TX, 77843, USA
| | - Rajesh C Miranda
- School of Medicine, Department of Neuroscience and Experimental Therapeutics, Medical Research and Education, Texas A&M University Health Science Center, Building 8447 Riverside Parkway, Bryan, TX, 77807-3260, USA.
- Women's Health in Neuroscience, Texas A&M University Health Science Center, Bryan, TX, USA.
| |
Collapse
|
6
|
The Effects of Transcranial Focused Ultrasound Stimulation of Nucleus Accumbens on Neuronal Gene Expression and Brain Tissue in High Alcohol-Preferring Rats. Mol Neurobiol 2023; 60:1099-1116. [PMID: 36417101 DOI: 10.1007/s12035-022-03130-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 11/09/2022] [Indexed: 11/24/2022]
Abstract
We investigated the effect of low-intensity focused ultrasound (LIFU) on gene expression related to alcohol dependence and histological effects on brain tissue. We also aimed at determining the miRNA-mRNA relationship and their pathways in alcohol dependence-induced expression changes after focused ultrasound therapy. We designed a case-control study for 100 days of observation to investigate differences in gene expression in the short-term stimulation group (STS) and long-term stimulation group (LTS) compared with the control sham group (SG). The study was performed in our Experimental Research Laboratory. 24 male high alcohol-preferring rats 63 to 79 days old, weighing 270 to 300 g, were included in the experiment. LTS received 50-day LIFU and STS received 10-day LIFU and 40-day sham stimulation, while the SG received 50-day sham stimulation. In miRNA expression analysis, it was found that LIFU caused gene expression differences in NAc. Significant differences were found between the groups for gene expression. Compared to the SG, the expression of 454 genes in the NAc region was changed in the STS while the expression of 382 genes was changed in the LTS. In the LTS, the expression of 32 genes was changed in total compared to STS. Our data suggest that LIFU targeted on NAc may assist in the treatment of alcohol dependence, especially in the long term possibly through altering gene expression. Our immunohistochemical studies verified that LIFU does not cause any tissue damage. These findings may lead to new studies in investigating the efficacy of LIFU for the treatment of alcohol dependence and also for other psychiatric disorders.
Collapse
|
7
|
Epigenetics in fetal alcohol spectrum disorder. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 197:211-239. [PMID: 37019593 DOI: 10.1016/bs.pmbts.2023.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
During pregnancy, alcohol abuse and its detrimental effects on developing offspring are major public health, economic and social challenges. The prominent characteristic attributes of alcohol (ethanol) abuse during pregnancy in humans are neurobehavioral impairments in offspring due to damage to the central nervous system (CNS), causing structural and behavioral impairments that are together named fetal alcohol spectrum disorder (FASD). Development-specific alcohol exposure paradigms were established to recapitulate the human FASD phenotypes and establish the underlying mechanisms. These animal studies have offered some critical molecular and cellular underpinnings likely to account for the neurobehavioral impairments associated with prenatal ethanol exposure. Although the pathogenesis of FASD remains unclear, emerging literature proposes that the various genomic and epigenetic components that cause the imbalance in gene expression can significantly contribute to the development of this disease. These studies acknowledged numerous immediate and enduring epigenetic modifications, such as methylation of DNA, post-translational modifications (PTMs) of histone proteins, and regulatory networks related to RNA, using many molecular approaches. Methylated DNA profiles, PTMs of histone proteins, and RNA-regulated expression of genes are essential for synaptic and cognitive behavior. Thus, offering a solution to many neuronal and behavioral impairments reported in FASD. In the current chapter, we review the recent advances in different epigenetic modifications that cause the pathogenesis of FASD. The information discussed can help better explain the pathogenesis of FASD and thereby might provide a basis for finding novel therapeutic targets and innovative treatment strategies.
Collapse
|
8
|
Dose-related shifts in proteome and function of extracellular vesicles secreted by fetal neural stem cells following chronic alcohol exposure. Heliyon 2022; 8:e11348. [PMID: 36387439 PMCID: PMC9649983 DOI: 10.1016/j.heliyon.2022.e11348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 09/07/2022] [Accepted: 10/26/2022] [Indexed: 11/06/2022] Open
Abstract
Accumulating evidence indicates that extracellular vesicles (EVs) mediate endocrine functions and also pathogenic effects of neurodevelopmental perturbagens like ethanol. We performed mass-spectrometry on EVs secreted by fetal murine cerebral cortical neural stem cells (NSCs), cultured ex-vivo as sex-specific neurosphere cultures, to identify overrepresented proteins and signaling pathways in EVs relative to parental NSCs in controls, and following exposure of parental NSCs to a dose range of ethanol. EV proteomes differ substantially from parental NSCs, and though EVs sequester proteins across sub-cellular compartments, they are enriched for distinct morphogenetic signals including the planar cell polarity pathway. Ethanol exposure favored selective protein sequestration in EVs and depletion in parental NSCs, and also resulted in dose-independent overrepresentation of cell-cycle and DNA replication pathways in EVs as well as dose-dependent overrepresentation of rRNA processing and mTor stress pathways. Transfer of untreated EVs to naïve cells resulted in decreased oxidative metabolism and S-phase, while EVs derived from ethanol-treated NSCs exhibited diminished effect. Collectively, these data show that NSCs secrete EVs with a distinct proteome that may have a general growth-inhibitory effect on recipient cells. Moreover, while ethanol results in selective transfer of proteins from NSCs to EVs, the efficacy of these exposure-derived EVs is diminished.
Collapse
|
9
|
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]
|
10
|
Komada M, Nishimura Y. Epigenetics and Neuroinflammation Associated With Neurodevelopmental Disorders: A Microglial Perspective. Front Cell Dev Biol 2022; 10:852752. [PMID: 35646933 PMCID: PMC9133693 DOI: 10.3389/fcell.2022.852752] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 04/22/2022] [Indexed: 12/15/2022] Open
Abstract
Neuroinflammation is a cause of neurodevelopmental disorders such as autism spectrum disorders, fetal alcohol syndrome, and cerebral palsy. Converging lines of evidence from basic and clinical sciences suggest that dysregulation of the epigenetic landscape, including DNA methylation and miRNA expression, is associated with neuroinflammation. Genetic and environmental factors can affect the interaction between epigenetics and neuroinflammation, which may cause neurodevelopmental disorders. In this minireview, we focus on neuroinflammation that might be mediated by epigenetic dysregulation in microglia, and compare studies using mammals and zebrafish.
Collapse
Affiliation(s)
- Munekazu Komada
- Mammalian Embryology, Department of Life Science, Faculty of Science and Engineering, Kindai University, Osaka, Japan
| | - Yuhei Nishimura
- Department of Integrative Pharmacology, Mie University Graduate School of Medicine, Tsu, Japan
- *Correspondence: Yuhei Nishimura,
| |
Collapse
|
11
|
Licheri V, Brigman JL. Altering Cell-Cell Interaction in Prenatal Alcohol Exposure Models: Insight on Cell-Adhesion Molecules During Brain Development. Front Mol Neurosci 2022; 14:753537. [PMID: 34975396 PMCID: PMC8715949 DOI: 10.3389/fnmol.2021.753537] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 11/11/2021] [Indexed: 11/17/2022] Open
Abstract
Alcohol exposure during pregnancy disrupts the development of the brain and produces long lasting behavioral and cognitive impairments collectively known as Fetal Alcohol Spectrum Disorders (FASDs). FASDs are characterized by alterations in learning, working memory, social behavior and executive function. A large body of literature using preclinical prenatal alcohol exposure models reports alcohol-induced changes in architecture and activity in specific brain regions affecting cognition. While multiple putative mechanisms of alcohol’s long-lasting effects on morphology and behavior have been investigated, an area that has received less attention is the effect of alcohol on cell adhesion molecules (CAMs). The embryo/fetal development represents a crucial period for Central Nervous System (CNS) development during which the cell-cell interaction plays an important role. CAMs play a critical role in neuronal migration and differentiation, synaptic organization and function which may be disrupted by alcohol. In this review, we summarize the physiological structure and role of CAMs involved in brain development, review the current literature on prenatal alcohol exposure effects on CAM function in different experimental models and pinpoint areas needed for future study to better understand how CAMs may mediate the morphological, sensory and behavioral outcomes in FASDs.
Collapse
Affiliation(s)
- Valentina Licheri
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Jonathan L Brigman
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, United States.,New Mexico Alcohol Research Center, UNM Health Sciences Center, Albuquerque, NM, United States
| |
Collapse
|
12
|
Jauhari A, Singh T, Yadav S. Neurodevelopmental Disorders and Neurotoxicity: MicroRNA in Focus. J Chem Neuroanat 2022; 120:102072. [DOI: 10.1016/j.jchemneu.2022.102072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 01/16/2022] [Accepted: 01/17/2022] [Indexed: 10/19/2022]
|
13
|
Lussier AA, Bodnar TS, Weinberg J. Intersection of Epigenetic and Immune Alterations: Implications for Fetal Alcohol Spectrum Disorder and Mental Health. Front Neurosci 2021; 15:788630. [PMID: 34924946 PMCID: PMC8680672 DOI: 10.3389/fnins.2021.788630] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 11/02/2021] [Indexed: 01/15/2023] Open
Abstract
Prenatal alcohol exposure can impact virtually all body systems, resulting in a host of structural, neurocognitive, and behavioral abnormalities. Among the adverse impacts associated with prenatal alcohol exposure are alterations in immune function, including an increased incidence of infections and alterations in immune/neuroimmune parameters that last throughout the life-course. Epigenetic patterns are also highly sensitive to prenatal alcohol exposure, with widespread alcohol-related alterations to epigenetic profiles, including changes in DNA methylation, histone modifications, and miRNA expression. Importantly, epigenetic programs are crucial for immune system development, impacting key processes such as immune cell fate, differentiation, and activation. In addition to their role in development, epigenetic mechanisms are emerging as attractive candidates for the biological embedding of environmental factors on immune function and as mediators between early-life exposures and long-term health. Here, following an overview of the impact of prenatal alcohol exposure on immune function and epigenetic patterns, we discuss the potential role for epigenetic mechanisms in reprogramming of immune function and the consequences for health and development. We highlight a range of both clinical and animal studies to provide insights into the array of immune genes impacted by alcohol-related epigenetic reprogramming. Finally, we discuss potential consequences of alcohol-related reprogramming of immune/neuroimmune functions and their effects on the increased susceptibility to mental health disorders. Overall, the collective findings from animal models and clinical studies highlight a compelling relationship between the immune system and epigenetic pathways. These findings have important implications for our understanding of the biological mechanisms underlying the long-term and multisystem effects of prenatal alcohol exposure, laying the groundwork for possible novel interventions and therapeutic strategies to treat individuals prenatally exposed to alcohol.
Collapse
Affiliation(s)
- Alexandre A Lussier
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, United States.,Department of Psychiatry, Harvard Medical School, Boston, MA, United States.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Tamara S Bodnar
- Department of Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Joanne Weinberg
- Department of Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
14
|
Alcohol induced impairment/abnormalities in brain: Role of MicroRNAs. Neurotoxicology 2021; 87:11-23. [PMID: 34478768 DOI: 10.1016/j.neuro.2021.08.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 08/12/2021] [Accepted: 08/28/2021] [Indexed: 12/21/2022]
Abstract
Alcohol is a highly toxic substance and has teratogenic properties that can lead to a wide range of developmental disorders. Excessive use of alcohol can change the structural and functional aspects of developed brain and other organs. Which can further lead to significant health, social and economic implications in many countries of the world. Convincing evidence support the involvement of microRNAs (miRNAs) as important post-transcriptional regulators of gene expression in neurodevelopment and maintenance. They also show differential expression following an injury. MiRNAs are the special class of small non coding RNAs that can modify the gene by targeting the mRNA and fine tune the development of cells to organs. Numerous pieces of evidences have shown the relationship between miRNA, alcohol and brain damage. These studies also show how miRNA controls different cellular mechanisms involved in the development of alcohol use disorder. With the increasing number of research studies, the roles of miRNAs following alcohol-induced injury could help researchers to recognize alternative therapeutic methods to treat/cure alcohol-induced brain damage. The present review summarizes the available data and brings together the important miRNAs, that play a crucial role in alcohol-induced brain damage, which will help in better understanding complex mechanisms. Identifying these miRNAs will not only expand the current knowledge but can lead to the identification of better targets for the development of novel therapeutic interventions.
Collapse
|
15
|
Arzua T, Jiang C, Yan Y, Bai X. The importance of non-coding RNAs in environmental stress-related developmental brain disorders: A systematic review of evidence associated with exposure to alcohol, anesthetic drugs, nicotine, and viral infections. Neurosci Biobehav Rev 2021; 128:633-647. [PMID: 34186153 PMCID: PMC8357057 DOI: 10.1016/j.neubiorev.2021.06.033] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 05/23/2021] [Accepted: 06/23/2021] [Indexed: 12/11/2022]
Abstract
Brain development is a dynamic and lengthy process that includes cell proliferation, migration, neurogenesis, gliogenesis, synaptogenesis, and pruning. Disruption of any of these developmental events can result in long-term outcomes ranging from brain structural changes, to cognitive and behavioral abnormality, with the mechanisms largely unknown. Emerging evidence suggests non-coding RNAs (ncRNAs) as pivotal molecules that participate in normal brain development and neurodevelopmental disorders. NcRNAs such as long non-coding RNAs (lncRNAs) and microRNAs (miRNAs) are transcribed from the genome but not translated into proteins. Many ncRNAs have been implicated as tuners of cell fate. In this review, we started with an introduction of the current knowledge of lncRNAs and miRNAs, and their potential roles in brain development in health and disorders. We then reviewed and discussed the evidence of ncRNA involvement in abnormal brain development resulted from alcohol, anesthetic drugs, nicotine, and viral infections. The complex connections among these ncRNAs were also discussed, along with potential overlapping ncRNA mechanisms, possible pharmacological targets for therapeutic/neuroprotective interventions, and potential biomarkers for brain developmental disorders.
Collapse
Affiliation(s)
- Thiago Arzua
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, WI, 53226, USA; Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Congshan Jiang
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Yasheng Yan
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Xiaowen Bai
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, WI, 53226, USA; Center of Systems Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.
| |
Collapse
|
16
|
Wallén E, Auvinen P, Kaminen-Ahola N. The Effects of Early Prenatal Alcohol Exposure on Epigenome and Embryonic Development. Genes (Basel) 2021; 12:genes12071095. [PMID: 34356111 PMCID: PMC8303887 DOI: 10.3390/genes12071095] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/05/2021] [Accepted: 07/15/2021] [Indexed: 12/15/2022] Open
Abstract
Prenatal alcohol exposure is one of the most significant causes of developmental disability in the Western world. Maternal alcohol consumption during pregnancy leads to an increased risk of neurological deficits and developmental abnormalities in the fetus. Over the past decade, several human and animal studies have demonstrated that alcohol causes alterations in epigenetic marks, including DNA methylation, histone modifications, and non-coding RNAs. There is an increasing amount of evidence that early pregnancy is a sensitive period for environmental-induced epigenetic changes. It is a dynamic period of epigenetic reprogramming, cell divisions, and DNA replication and, therefore, a particularly interesting period to study the molecular changes caused by alcohol exposure as well as the etiology of alcohol-induced developmental disorders. This article will review the current knowledge about the in vivo and in vitro effects of alcohol exposure on the epigenome, gene regulation, and the phenotype during the first weeks of pregnancy.
Collapse
|
17
|
Gowen AM, Odegaard KE, Hernandez J, Chand S, Koul S, Pendyala G, Yelamanchili SV. Role of microRNAs in the pathophysiology of addiction. WILEY INTERDISCIPLINARY REVIEWS. RNA 2021; 12:e1637. [PMID: 33336550 PMCID: PMC8026578 DOI: 10.1002/wrna.1637] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 11/12/2020] [Accepted: 11/19/2020] [Indexed: 02/06/2023]
Abstract
Addiction is a chronic and relapsing brain disorder characterized by compulsive seeking despite adverse consequences. There are both heritable and epigenetic mechanisms underlying drug addiction. Emerging evidence suggests that non-coding RNAs (ncRNAs) such as microRNAs (miRNAs), long non-coding RNAs, and circular RNAs regulate synaptic plasticity and related behaviors caused by substances of abuse. These ncRNAs modify gene expression and may contribute to the behavioral phenotypes of addiction. Among the ncRNAs, the most widely researched and impactful are miRNAs. The goal in this systematic review is to provide a detailed account of recent research involving the role of miRNAs in addiction. This article is categorized under: RNA Interactions with Proteins and Other Molecules > Small Molecule-RNA Interactions RNA in Disease and Development > RNA in Disease.
Collapse
Affiliation(s)
- Austin M Gowen
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Katherine E Odegaard
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Jordan Hernandez
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Subhash Chand
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Sneh Koul
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Gurudutt Pendyala
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Sowmya V Yelamanchili
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
18
|
Mahnke AH, Sideridis GD, Salem NA, Tseng AM, Carter RC, Dodge NC, Rathod AB, Molteno CD, Meintjes EM, Jacobson SW, Miranda RC, Jacobson JL. Infant circulating MicroRNAs as biomarkers of effect in fetal alcohol spectrum disorders. Sci Rep 2021; 11:1429. [PMID: 33446819 PMCID: PMC7809131 DOI: 10.1038/s41598-020-80734-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 12/10/2020] [Indexed: 12/19/2022] Open
Abstract
Prenatal alcohol exposure (PAE) can result in cognitive and behavioral disabilities and growth deficits. Because alcohol-related neurobehavioral deficits may occur in the absence of overt dysmorphic features or growth deficits, there is a need to identify biomarkers of PAE that can predict neurobehavioral impairment. In this study, we assessed infant plasma extracellular, circulating miRNAs (exmiRNAs) obtained from a heavily exposed Cape Town cohort to determine whether these can be used to predict PAE-related growth restriction and cognitive impairment. PAE, controlling for smoking as a covariate, altered 27% of expressed exmiRNAs with clinically-relevant effect sizes (Cohen's d ≥ 0.4). Moreover, at 2 weeks, PAE increased correlated expression of exmiRNAs across chromosomes, suggesting potential co-regulation. In confirmatory factor analysis, the variance in expression for PAE-altered exmiRNAs at 2 weeks and 6.5 months was best described by three-factor models. Pathway analysis found that factors at 2 weeks were associated with (F1) cell maturation, cell cycle inhibition, and somatic growth, (F2) cell survival, apoptosis, cardiac development, and metabolism, and (F3) cell proliferation, skeletal development, hematopoiesis, and inflammation, and at 6.5 months with (F1) neurodevelopment, neural crest/mesoderm-derivative development and growth, (F2) immune system and inflammation, and (F3) somatic growth and cardiovascular development. Factors F3 at 2 weeks and F2 at 6.5 months partially mediated PAE-induced growth deficits, and factor F3 at 2 weeks partially mediated effects of PAE on infant recognition memory at 6.5 months. These findings indicate that infant exmiRNAs can help identify infants who will exhibit PAE-related deficits in growth and cognition.
Collapse
Affiliation(s)
- Amanda H Mahnke
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, TX, 77807, USA.
- Women's Health in Neuroscience Program, Texas A&M University Health Science Center, Bryan, TX, 77807, USA.
| | - Georgios D Sideridis
- Harvard Medical School, Boston Children's Hospital, Institutional Centers for Clinical and Translational Research, Boston, MA, 02115, USA
| | - Nihal A Salem
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, TX, 77807, USA
| | - Alexander M Tseng
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, TX, 77807, USA
| | - R Colin Carter
- Departments of Pediatrics and Emergency Medicine, Institute of Human Nutrition, Columbia University Medical Center, New York, NY, 10032, USA
| | - Neil C Dodge
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Aniruddha B Rathod
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, TX, 77807, USA
| | - Christopher D Molteno
- Departments of Human Biology and of Psychiatry and Mental Health, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Ernesta M Meintjes
- Division of Biomedical Engineering, Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Sandra W Jacobson
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, 48201, USA
- Departments of Human Biology and of Psychiatry and Mental Health, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Rajesh C Miranda
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, TX, 77807, USA
- Women's Health in Neuroscience Program, Texas A&M University Health Science Center, Bryan, TX, 77807, USA
| | - Joseph L Jacobson
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, 48201, USA.
- Departments of Human Biology and of Psychiatry and Mental Health, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
19
|
MicroRNAs Dysregulation and Mitochondrial Dysfunction in Neurodegenerative Diseases. Int J Mol Sci 2020; 21:ijms21175986. [PMID: 32825273 PMCID: PMC7504116 DOI: 10.3390/ijms21175986] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/12/2020] [Accepted: 08/18/2020] [Indexed: 02/06/2023] Open
Abstract
Neurodegenerative diseases are debilitating and currently incurable conditions causing severe cognitive and motor impairments, defined by the progressive deterioration of neuronal structure and function, eventually causing neuronal loss. Understand the molecular and cellular mechanisms underlying these disorders are essential to develop therapeutic approaches. MicroRNAs (miRNAs) are short non-coding RNAs implicated in gene expression regulation at the post-transcriptional level. Moreover, miRNAs are crucial for different processes, including cell growth, signal transmission, apoptosis, cancer and aging-related neurodegenerative diseases. Altered miRNAs levels have been associated with the formation of reactive oxygen species (ROS) and mitochondrial dysfunction. Mitochondrial dysfunction and ROS formation occur in many neurodegenerative diseases such as Alzheimer's, Parkinson's and Huntington's diseases. The crosstalk existing among oxidative stress, mitochondrial dysfunction and miRNAs dysregulation plays a pivotal role in the onset and progression of neurodegenerative diseases. Based on this evidence, in this review, with a focus on miRNAs and their role in mitochondrial dysfunction in aging-related neurodegenerative diseases, with a focus on their potential as diagnostic biomarkers and therapeutic targets.
Collapse
|
20
|
Elsonbaty SM, Ismail AFM. Nicotine encourages oxidative stress and impairment of rats' brain mitigated by Spirulina platensis lipopolysaccharides and low-dose ionizing radiation. Arch Biochem Biophys 2020; 689:108382. [PMID: 32343976 DOI: 10.1016/j.abb.2020.108382] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 04/04/2020] [Accepted: 04/17/2020] [Indexed: 12/20/2022]
Abstract
Nicotine is a psychoactive alkaloid of tobacco, which is ingested during cigarettes or electronic cigarette smoking. Extensive consumption of nicotine induced oxidative stress. Accordingly, it is implicated in many pathophysiology brain disorders and triggers neurodegeneration. In this study, we investigated the protective role of Spirulina platensis-lipopolysaccharides (S.LPS) and the low dose-ionizing radiation (LD-IR) against the induced neurotoxicity in the rats' brain due to the prolonged administration of high nicotine levels. Rats treated with nicotine for two months showed alterations in the oxidative stress markers (malondialdehyde (MDA), reduced glutathione (GSH) and oxidized glutathione disulfide (GSSG)), antioxidant enzymes (superoxide dismutase (SOD), catalase (Cat), glutathione enzymes (GPx and GST)) as well as several pro-inflammatory markers (Tumor Necrosis Factor-alpha (TNF-α), Interleukin-17 (IL-17), and Nuclear Factor-kappa B (NF-κB)), and induced apoptosis through Caspase-3 activity. Nicotine also upregulated the mRNA gene expression of cytochrome P450 enzymes (CYP2B1 and CYP2E1), Cyclin-dependent kinase 5 (CDK5), Toll-Like Receptor 4 (TLR4), and phospho-Tau (p-Tau) protein expression. Besides, it downregulated the alpha-7 nicotinic receptor (α7nAChR) mRNA gene expression accompanied by a decline in the calcium (Ca2+) level. S.LPS exhibited antioxidant, anti-inflammatory, anti-apoptotic and neuroprotective activities, which counteracting the detrimental effects of chronic nicotine administration. LD-IR demonstrated comparable effects to S.LPS. Exposure of rats to LD-IR enhanced the neuroprotective effects of S.LPS against nicotine toxicity. The light microscopic examination of the brain tissues was in agreement with the biochemical investigations. These findings display that S.LPS and LD-IR mitigated the oxidative stress and the impairment of rats' brain induced by nicotine, due to regulation of the mRNA gene expression of cytochrome P450 enzymes (CYP2B1 and CYP2E1) and the signaling pathway of Tau protein phosphorylation.
Collapse
Affiliation(s)
- Sawsan M Elsonbaty
- Radiation Microbiology Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Ahmed El-Zomor St. 3, El-Zohoor Dist., Nasr City, 11787, Cairo, Egypt
| | - Amel F M Ismail
- Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Ahmed El-Zomor St. 3, El-Zohoor Dist., Nasr City, 11787, Cairo, Egypt.
| |
Collapse
|
21
|
Almeida L, Andreu-Fernández V, Navarro-Tapia E, Aras-López R, Serra-Delgado M, Martínez L, García-Algar O, Gómez-Roig MD. Murine Models for the Study of Fetal Alcohol Spectrum Disorders: An Overview. Front Pediatr 2020; 8:359. [PMID: 32760684 PMCID: PMC7373736 DOI: 10.3389/fped.2020.00359] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 05/29/2020] [Indexed: 12/15/2022] Open
Abstract
Prenatal alcohol exposure is associated to different physical, behavioral, cognitive, and neurological impairments collectively known as fetal alcohol spectrum disorder. The underlying mechanisms of ethanol toxicity are not completely understood. Experimental studies during human pregnancy to identify new diagnostic biomarkers are difficult to carry out beyond genetic or epigenetic analyses in biological matrices. Therefore, animal models are a useful tool to study the teratogenic effects of alcohol on the central nervous system and analyze the benefits of promising therapies. Animal models of alcohol spectrum disorder allow the analysis of key variables such as amount, timing and frequency of ethanol consumption to describe the harmful effects of prenatal alcohol exposure. In this review, we aim to synthetize neurodevelopmental disabilities in rodent fetal alcohol spectrum disorder phenotypes, considering facial dysmorphology and fetal growth restriction. We examine the different neurodevelopmental stages based on the most consistently implicated epigenetic mechanisms, cell types and molecular pathways, and assess the advantages and disadvantages of murine models in the study of fetal alcohol spectrum disorder, the different routes of alcohol administration, and alcohol consumption patterns applied to rodents. Finally, we analyze a wide range of phenotypic features to identify fetal alcohol spectrum disorder phenotypes in murine models, exploring facial dysmorphology, neurodevelopmental deficits, and growth restriction, as well as the methodologies used to evaluate behavioral and anatomical alterations produced by prenatal alcohol exposure in rodents.
Collapse
Affiliation(s)
- Laura Almeida
- Maternal and Child Health and Development Network II (SAMID II), Instituto de Salud Carlos III (ISCIII), Barcelona, Spain
- Fundació Sant Joan de Déu, Barcelona, Spain
- BCNatal Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Sant Joan de Déu and Hospital Clínic, Barcelona, Spain
| | - Vicente Andreu-Fernández
- Maternal and Child Health and Development Network II (SAMID II), Instituto de Salud Carlos III (ISCIII), Barcelona, Spain
- Nutrition and Health Deparment, Valencian International University (VIU), Valencia, Spain
- Grup de Recerca Infancia i Entorn (GRIE), Institut D'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Elisabet Navarro-Tapia
- Maternal and Child Health and Development Network II (SAMID II), Instituto de Salud Carlos III (ISCIII), Barcelona, Spain
- BCNatal Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Sant Joan de Déu and Hospital Clínic, Barcelona, Spain
- Grup de Recerca Infancia i Entorn (GRIE), Institut D'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Rosa Aras-López
- Maternal and Child Health and Development Network II (SAMID II), Instituto de Salud Carlos III (ISCIII), Barcelona, Spain
- Congenital Malformations Lab, Institute of Medicine and Molecular Genetic (INGEMM), Institute for Health Research of La Paz Universitary Hospital (IdiPAZ), Madrid, Spain
| | - Mariona Serra-Delgado
- BCNatal Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Sant Joan de Déu and Hospital Clínic, Barcelona, Spain
| | - Leopoldo Martínez
- Maternal and Child Health and Development Network II (SAMID II), Instituto de Salud Carlos III (ISCIII), Barcelona, Spain
- Congenital Malformations Lab, Institute of Medicine and Molecular Genetic (INGEMM), Institute for Health Research of La Paz Universitary Hospital (IdiPAZ), Madrid, Spain
- Department of Pediatric Surgery, Hospital Universitario La Paz, Madrid, Spain
| | - Oscar García-Algar
- Maternal and Child Health and Development Network II (SAMID II), Instituto de Salud Carlos III (ISCIII), Barcelona, Spain
- Grup de Recerca Infancia i Entorn (GRIE), Institut D'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Department of Neonatology, Hospital Clínic-Maternitat, ICGON, IDIBAPS, BCNatal, Barcelona, Spain
| | - María Dolores Gómez-Roig
- Maternal and Child Health and Development Network II (SAMID II), Instituto de Salud Carlos III (ISCIII), Barcelona, Spain
- Fundació Sant Joan de Déu, Barcelona, Spain
- BCNatal Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Sant Joan de Déu and Hospital Clínic, Barcelona, Spain
| |
Collapse
|
22
|
Singh T, Yadav S. Role of microRNAs in neurodegeneration induced by environmental neurotoxicants and aging. Ageing Res Rev 2020; 60:101068. [PMID: 32283224 DOI: 10.1016/j.arr.2020.101068] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 03/02/2020] [Accepted: 04/04/2020] [Indexed: 02/06/2023]
Abstract
The progressive loss of neuronal structure and functions resulting in the death of neurons is considered as neurodegeneration. Environmental toxicants induced degeneration of neurons is accelerated with aging. In adult brains, most of the neurons are post-mitotic, and their loss results in the development of diseases like amyotrophic lateral sclerosis (ALS), Parkinson's disease (PD), Alzheimer's disease (AD), and Huntington's disease (HD). Neurodegenerative diseases have several similarities at the sub-cellular and molecular levels, such as synaptic degeneration, oxidative stress, inflammation, and cognitive decline, which are also known in brain aging. Identification of these similarities at the molecular level offers hope for the development of new therapeutics to ameliorate all neurodegenerative diseases simultaneously. Aging is known as the most strongly associated additive factor in the pathogenesis of neurodegenerative diseases. Studies carried out so far identified several genes, which are responsible for selective degeneration of neurons in different neurodegenerative diseases. Countless efforts have been made in identifying therapeutics for neurodegenerative diseases; however, the discovery of effective therapy remains elusive. Findings made in the last two decades identified microRNAs (miRNAs) as the most potent post-transcription regulatory RNA molecule, which can condition protein levels in the cell and tissue-specific manner. Identification of miRNAs, which regulate both neurotoxicant and aging-associated degeneration of brain cells, raises the possibility that roads leading to aging and neurotoxicant induced neurodegeneration cross at some point. Identification of miRNAs, which are common to aging and neurotoxicant induced neurodegeneration, will help in understanding the complex mechanism of neurodegenerative disease development. In the future, the use of natural miRNAs in vivo in therapy will be able to tackle several issues of aging and neurodegeneration. In the present review, we have provided a summary of findings made on the role of miRNAs in neurodegeneration and explored the common link made by miRNAs between aging and neurotoxicants induced neurodegeneration.
Collapse
Affiliation(s)
- Tanisha Singh
- Developmental Toxicology Division, Systems Toxicology and Health Risk Assessment Group, CSIR- Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan,31 Mahatma Gandhi Marg, Lucknow-226001, Uttar Pradesh, India; Department of Neurological Surgery, School of Medicine, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, Pennsylvania-15213, USA.
| | - Sanjay Yadav
- Developmental Toxicology Division, Systems Toxicology and Health Risk Assessment Group, CSIR- Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan,31 Mahatma Gandhi Marg, Lucknow-226001, Uttar Pradesh, India; Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Raebareli, Munsiganj, Raebareli 229405, UP, India.
| |
Collapse
|
23
|
Janeczek P, Colson N, Dodd PR, Lewohl JM. Sex Differences in the Expression of the α5 Subunit of the GABA A Receptor in Alcoholics with and without Cirrhosis of the Liver. Alcohol Clin Exp Res 2020; 44:423-434. [PMID: 31840824 DOI: 10.1111/acer.14266] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 12/02/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND Alcohol exposure alters the expression of a large number of genes, resulting in neuronal adaptions and neuronal loss, but the underlying mechanisms are largely unknown. miRNAs are gene repressors that are abundant in the brain. A recent study identified ~ 35 miRNAs that are up-regulated in the prefrontal cortex of human alcoholics and predicted to target genes that are down-regulated in the same region. Although interactions between alcohol-responsive miRNAs and their target genes have been predicted, few studies have validated these predictions. METHODS We measured the expression of GABAA α5 mRNA in the prefrontal and motor cortices of human alcoholics and matched controls using real-time PCR. The expression of miR-203 was measured in a subset of these cases. The predicted interaction of miR-203 and GABRA5 was validated for miR-203 using a luciferase reporter assay. RESULTS In both frontal and motor cortices, the expression of GABAA α5 was significantly lower in cirrhotic alcoholics compared with controls. Further, the pattern of expression between the groups was significantly different between males and females. The expression of miR-203 was higher in the prefrontal cortex of cirrhotic alcoholics compared with controls and uncomplicated alcoholics. These differences were particularly marked in female cases. Cotransfection of GABRA5 with miR-203 in HEK293T cells reduced luciferase reporter activity. CONCLUSION There are sex differences in the expression of GABAA α5 and miR-203 in the brain of human alcoholics which are particularly marked in alcoholics with cirrhosis of the liver. Further, miR-203 may mediate the changes in expression of this GABAA receptor isoform that is brought about by alcohol exposure.
Collapse
Affiliation(s)
- Paulina Janeczek
- From the, School of Medical Science, (PJ, NC, JML), Griffith University Gold Coast campus, Southport, Queensland, Australia
| | - Natalie Colson
- From the, School of Medical Science, (PJ, NC, JML), Griffith University Gold Coast campus, Southport, Queensland, Australia
| | - Peter R Dodd
- School of Chemistry and Molecular Biosciences, (PRD), The University of Queensland St Lucia campus, Brisbane, Queensland, Australia
| | - Joanne M Lewohl
- From the, School of Medical Science, (PJ, NC, JML), Griffith University Gold Coast campus, Southport, Queensland, Australia
| |
Collapse
|
24
|
Pucci M, Micioni Di Bonaventura MV, Wille-Bille A, Fernández MS, Maccarrone M, Pautassi RM, Cifani C, D’Addario C. Environmental stressors and alcoholism development: Focus on molecular targets and their epigenetic regulation. Neurosci Biobehav Rev 2019; 106:165-181. [DOI: 10.1016/j.neubiorev.2018.07.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 06/13/2018] [Accepted: 07/09/2018] [Indexed: 01/17/2023]
|
25
|
Pinson MR, Miranda RC. Noncoding RNAs in development and teratology, with focus on effects of cannabis, cocaine, nicotine, and ethanol. Birth Defects Res 2019; 111:1308-1319. [PMID: 31356004 DOI: 10.1002/bdr2.1559] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 07/11/2019] [Accepted: 07/12/2019] [Indexed: 02/06/2023]
Abstract
Completion of the Human Genome Project has led to the identification of a large number of transcription start sites that are not paired with protein-coding genes, supporting the growing recognition of the abundance of encoded nonprotein-coding RNAs (ncRNAs) and their importance for speciation and species-specific development. Present in both plants and animals, ncRNAs vary in size, function, primary sequence, and secondary structure. While microRNAs (miRNAs) are the best known, there are a number of other ncRNAs (long[er] nonprotein-coding RNA, pseudogenes, circular RNAs, and so on) that have been shown to play an important role in the development either directly or via networks of proteins and other ncRNAs, including modulating the impact of miRNAs. Furthermore, these ncRNAs and their developmental regulatory networks are sensitive to teratogens such as ethanol, cannabis, cocaine, and nicotine. A better understanding of the developmental role of ncRNAs and their capacity to mediate teratogenesis is a necessary step in efforts to minimize the long-term consequences of developmental exposures to drugs-of-abuse. Moreover, with increasing awareness of the prevalence of polydrug use, experimental models will need to incorporate more complex drug exposure paradigms into meaningful assessments of developmental ncRNA function.
Collapse
Affiliation(s)
- Marisa R Pinson
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, 8447 Riverside Pkwy Suite 1005 MREB, Bryan, Texas
| | - Rajesh C Miranda
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, 8447 Riverside Pkwy Suite 1005 MREB, Bryan, Texas
| |
Collapse
|
26
|
Tseng AM, Chung DD, Pinson MR, Salem NA, Eaves SE, Miranda RC. Ethanol Exposure Increases miR-140 in Extracellular Vesicles: Implications for Fetal Neural Stem Cell Proliferation and Maturation. Alcohol Clin Exp Res 2019; 43:1414-1426. [PMID: 31009095 DOI: 10.1111/acer.14066] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Accepted: 04/12/2019] [Indexed: 12/23/2022]
Abstract
BACKGROUND Neural stem cells (NSCs) generate most of the neurons of the adult brain in humans, during the mid-first through second-trimester period. This critical neurogenic window is particularly vulnerable to prenatal alcohol exposure, which can result in diminished brain growth. Previous studies showed that ethanol (EtOH) exposure does not kill NSCs, but, rather, results in their depletion by influencing cell cycle kinetics and promoting aberrant maturation, in part, by altering NSC expression of key neurogenic miRNAs. NSCs reside in a complex microenvironment rich in extracellular vesicles, shown to traffic miRNA cargo between cells. METHODS We profiled the miRNA content of extracellular vesicles from control and EtOH-exposed ex vivo neurosphere cultures of fetal NSCs. We subsequently examined the effects of one EtOH-sensitive miRNA, miR-140-3p, on NSC growth, survival, and maturation. RESULTS EtOH exposure significantly elevates levels of a subset of miRNAs in secreted extracellular vesicles. Overexpression of one of these elevated miRNAs, miR-140-3p, and its passenger strand relative, miR-140-5p, significantly increased the proportion of S-phase cells while decreasing the proportion of G0 /G1 cells compared to controls. In contrast, while miR-140-3p knockdown had minimal effects on the proportion of cells in each phase of the cell cycle, knockdown of miR-140-5p significantly decreased the proportion of cells in G2 /M phase. Furthermore, miR-140-3p overexpression, during mitogen-withdrawal-induced NSC differentiation, favors astroglial maturation at the expense of neural and oligodendrocyte differentiation. CONCLUSIONS Collectively, the dysregulated miRNA content of extracellular vesicles following EtOH exposure may result in aberrant neural progenitor cell growth and maturation, explaining brain growth deficits associated with prenatal alcohol exposure.
Collapse
Affiliation(s)
- Alexander M Tseng
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, Texas
| | - Dae D Chung
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, Texas
| | - Marisa R Pinson
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, Texas
| | - Nihal A Salem
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, Texas
| | - Sarah E Eaves
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, Texas
| | - Rajesh C Miranda
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, Texas
| |
Collapse
|
27
|
Montgomery KS, Bancroft EA, Fincher AS, Migut EA, Provasek V, Murchison D, DuBois DW. Effects of ethanol and varenicline on female Sprague-Dawley rats in a third trimester model of fetal alcohol syndrome. Alcohol 2018; 71:75-87. [PMID: 30059955 PMCID: PMC6223131 DOI: 10.1016/j.alcohol.2018.02.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 02/03/2018] [Accepted: 02/26/2018] [Indexed: 12/17/2022]
Abstract
Perinatal ethanol exposure disrupts a variety of developmental processes in neurons important for establishing a healthy brain. These ethanol-induced impairments known as fetal alcohol spectrum disorder (FASD) are not fully understood, and currently, there is no effective treatment. Further, growing evidence suggests that adult females are more susceptible to ethanol, with the effects of perinatal ethanol exposure also being sexually divergent. Female models have been historically underutilized in neurophysiological investigations, but here, we used a third-trimester binge-ethanol model of FASD to examine changes to basal forebrain (BF) physiology and behavior in female Sprague-Dawley rats. We also tested varenicline as a potential cholinomimetic therapeutic. Rat pups were gavage-treated with binge-like ethanol, varenicline and ethanol, and varenicline alone. Using patch-clamp electrophysiology in BF slices, we observed that binge-ethanol exposure increased spontaneous post-synaptic current (sPSC) frequency. Varenicline exposure alone also enhanced sPSC frequency. Varenicline plus ethanol co-treatment prevented the sPSC frequency increase. Changes in BF synaptic transmission persisted into adolescence after binge-ethanol treatment. Behaviorally, binge-ethanol treated females displayed increased anxiety (thigmotaxis) and demonstrated learning deficits in the water maze. Varenicline/ethanol co-treatment was effective at reducing these behavioral deficits. In the open field, ethanol-treated rats displayed longer distances traveled and spent less time in the center of the open field box. Co-treated rats displayed less anxiety, demonstrating a possible effect of varenicline on this measure. In conclusion, ethanol-induced changes in both BF synaptic transmission and behavior were reduced by varenicline in female rats, supporting a role for cholinergic therapeutics in FASD treatment.
Collapse
Affiliation(s)
- Karienn S Montgomery
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, TX, United States
| | - Eric A Bancroft
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, TX, United States
| | - Annette S Fincher
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, TX, United States
| | - Ewelina A Migut
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, TX, United States
| | - Vincent Provasek
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, TX, United States
| | - David Murchison
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, TX, United States
| | - Dustin W DuBois
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, TX, United States.
| |
Collapse
|
28
|
Ranjit S, Patters BJ, Gerth KA, Haque S, Choudhary S, Kumar S. Potential neuroprotective role of astroglial exosomes against smoking-induced oxidative stress and HIV-1 replication in the central nervous system. Expert Opin Ther Targets 2018; 22:703-714. [PMID: 30015535 DOI: 10.1080/14728222.2018.1501473] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
INTRODUCTION HIV-1-infected smokers are at risk of oxidative damage to neuronal cells in the central nervous system by both HIV-1 and cigarette smoke. Since neurons have a weak antioxidant defense system, they mostly depend on glial cells, particularly astrocytes, for protection against oxidative damage and neurotoxicity. Astrocytes augment the neuronal antioxidant system by supplying cysteine-containing products for glutathione synthesis, antioxidant enzymes such as SOD and catalase, glucose for antioxidant regeneration via the pentose-phosphate pathway, and by recycling of ascorbic acid. Areas covered: The transport of antioxidants and energy substrates from astrocytes to neurons could possibly occur via extracellular nanovesicles called exosomes. This review highlights the neuroprotective potential of exosomes derived from astrocytes against smoking-induced oxidative stress, HIV-1 replication, and subsequent neurotoxicity observed in HIV-1-positive smokers. Expert opinion: During stress conditions, the antioxidants released from astrocytes either via extracellular fluid or exosomes to neurons may not be sufficient to provide neuroprotection. Therefore, we put forward a novel strategy to combat oxidative stress in the central nervous system, using synthetically developed exosomes loaded with antioxidants such as glutathione and the anti-aging protein Klotho.
Collapse
Affiliation(s)
- Sabina Ranjit
- a Department of Pharmaceutical Sciences , University of Tennessee Health Science Center , Memphis , TN , United States
| | - Benjamin J Patters
- a Department of Pharmaceutical Sciences , University of Tennessee Health Science Center , Memphis , TN , United States
| | - Kelli A Gerth
- a Department of Pharmaceutical Sciences , University of Tennessee Health Science Center , Memphis , TN , United States
| | - Sanjana Haque
- a Department of Pharmaceutical Sciences , University of Tennessee Health Science Center , Memphis , TN , United States
| | - Sanjeev Choudhary
- b Department of Internal Medicine , University of Texas Medical Branch , Galveston , TX , United States
| | - Santosh Kumar
- a Department of Pharmaceutical Sciences , University of Tennessee Health Science Center , Memphis , TN , United States
| |
Collapse
|
29
|
Nonprotein-coding RNAs in Fetal Alcohol Spectrum Disorders. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 157:299-342. [PMID: 29933954 DOI: 10.1016/bs.pmbts.2017.11.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Early developmental exposure to ethanol, a known teratogen, can result in a range of neurodevelopmental disorders, collectively referred to as Fetal Alcohol Spectrum Disorders (FASDs). Changes in the environment, including exposure to teratogens, can result in long term alterations to the epigenetic landscape of a cell, thereby altering gene expression. Noncoding RNAs (ncRNAs) can affect transcription and translation of networks of genes. ncRNAs are dynamically expressed during development and have been identified as a target of alcohol. ncRNAs therefore make for attractive targets for novel therapeutics to address the developmental deficits associated with FASDs.
Collapse
|
30
|
Boschen KE, Keller SM, Roth TL, Klintsova AY. Epigenetic mechanisms in alcohol- and adversity-induced developmental origins of neurobehavioral functioning. Neurotoxicol Teratol 2018; 66:63-79. [PMID: 29305195 DOI: 10.1016/j.ntt.2017.12.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 12/11/2017] [Accepted: 12/26/2017] [Indexed: 12/13/2022]
Abstract
The long-term effects of developmental alcohol and stress exposure are well documented in both humans and non-human animal models. Damage to the brain and attendant life-long impairments in cognition and increased risk for psychiatric disorders are debilitating consequences of developmental exposure to alcohol and/or psychological stress. Here we discuss evidence for a role of epigenetic mechanisms in mediating these consequences. While we highlight some of the common ways in which stress or alcohol impact the epigenome, we point out that little is understood of the epigenome's response to experiencing both stress and alcohol exposure, though stress is a contributing factor as to why women drink during pregnancy. Advancing our understanding of this relationship is of critical concern not just for the health and well-being of individuals directly exposed to these teratogens, but for generations to come.
Collapse
Affiliation(s)
- K E Boschen
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, NC 27599, United States
| | - S M Keller
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, United States
| | - T L Roth
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, United States.
| | - A Y Klintsova
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, United States.
| |
Collapse
|
31
|
Lopez MS, Dempsey RJ, Vemuganti R. The microRNA miR-21 conditions the brain to protect against ischemic and traumatic injuries. CONDITIONING MEDICINE 2017; 1:35-46. [PMID: 34268484 PMCID: PMC8279043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Ischemic and traumatic injuries to CNS remain leading causes of death and disability worldwide, despite decades of research into risk factors, therapies, and preventative measures. Recent studies showed that CNS injuries significantly alter the cerebral microRNAome that impact the secondary brain damage as well as plasticity and recovery. Many microRNA based therapies are currently in various clinical trials for different pathologic conditions indicating their therapeutic potential. In the present review, we discuss the role of miR-21 in acute CNS injuries which is currently thought to be a potent neuroprotective microRNA. We emphasize on the potential of miR-21 in promoting cell and tissue survival and preventing inflammation and apoptosis. We also discussed the role of miR-21 in conditioning the brain to promote ischemic tolerance. Finally, we discussed some of the challenges and difficulties to develop miR-21 as a neuroprotective therapy in humans.
Collapse
Affiliation(s)
- Mary S Lopez
- Cellular and Molecular Pathology Program, University of Wisconsin, Madison, WI, USA
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Robert J Dempsey
- Cellular and Molecular Pathology Program, University of Wisconsin, Madison, WI, USA
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Raghu Vemuganti
- Cellular and Molecular Pathology Program, University of Wisconsin, Madison, WI, USA
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
- William S. Middleton Veteran's Administration Hospital, Madison, WI, USA
| |
Collapse
|
32
|
Tarren JR, Lester HA, Belmer A, Bartlett SE. Acute Ethanol Administration Upregulates Synaptic α4-Subunit of Neuronal Nicotinic Acetylcholine Receptors within the Nucleus Accumbens and Amygdala. Front Mol Neurosci 2017; 10:338. [PMID: 29114204 PMCID: PMC5660714 DOI: 10.3389/fnmol.2017.00338] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 10/05/2017] [Indexed: 12/26/2022] Open
Abstract
Alcohol and nicotine are two of the most frequently abused drugs, with their comorbidity well described. Previous data show that chronic exposure to nicotine upregulates high-affinity nicotinic acetylcholine receptors (nAChRs) in several brain areas. Effects of ethanol on specific brain nAChR subtypes within the mesolimbic dopaminergic (DA) pathway may be a key element in the comorbidity of ethanol and nicotine. However, it is unknown how alcohol affects the abundance of these receptor proteins. In the present study, we measured the effect of acute binge ethanol on nAChR α4 subunit levels in the prefrontal cortex (PFC), nucleus accumbens (NAc), ventral tegmental area (VTA), and amygdala (Amg) by western blot analysis using a knock-in mouse line, generated with a normally functioning α4 nAChR subunit tagged with yellow fluorescent protein (YFP). We observed a robust increase in α4-YFP subunit levels in the NAc and the Amg following acute ethanol, with no changes in the PFC and VTA. To further investigate whether this upregulation was mediated by increased local mRNA transcription, we quantified mRNA levels of the Chrna4 gene using qRT-PCR. We found no effect of ethanol on α4 mRNA expression, suggesting that the upregulation of α4 protein rather occurs post-translationally. The quantitative counting of YFP immunoreactive puncta further revealed that α4-YFP protein is upregulated in presynaptic boutons of the dopaminergic axons projecting to the shell and the core regions of the NAc as well as to the basolateral amygdala (BLA), but not to the central or lateral Amg. Together, our results demonstrate that a single exposure to binge ethanol upregulates level of synaptic α4∗ nAChRs in dopaminergic inputs to the NAc and BLA. This upregulation could be linked to the functional dysregulation of dopaminergic signalling observed during the development of alcohol dependence.
Collapse
Affiliation(s)
- Josephine R Tarren
- Translational Research Institute, Institute for Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
| | - Henry A Lester
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, United States
| | - Arnauld Belmer
- Translational Research Institute, Institute for Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
| | - Selena E Bartlett
- Translational Research Institute, Institute for Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
| |
Collapse
|
33
|
Miguel-Hidalgo JJ, Hall KO, Bonner H, Roller AM, Syed M, Park CJ, Ball JP, Rothenberg ME, Stockmeier CA, Romero DG. MicroRNA-21: Expression in oligodendrocytes and correlation with low myelin mRNAs in depression and alcoholism. Prog Neuropsychopharmacol Biol Psychiatry 2017; 79:503-514. [PMID: 28802862 PMCID: PMC5610939 DOI: 10.1016/j.pnpbp.2017.08.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 07/21/2017] [Accepted: 08/07/2017] [Indexed: 02/08/2023]
Abstract
MiR-21 is a microRNA implicated in cancer, development, and cardiovascular diseases and expressed in the central nervous system (CNS), especially after injury. However, the cellular expression of miR-21 in the adult CNS has not been clearly established either in mice or human subjects, while its alteration in psychiatric disorders is unknown. MiR-21 expression was characterized in reporter mice expressing β-galactosidase (LacZ) under the endogenous miR-21 promoter (miR-21/LacZ). Brain co-localization of miR-21/LacZ with specific neural markers was examined by double immunofluorescence in reporter mice, while extent of immunostaining for myelin basic protein and PDGFRα was determined in miR-21 knockout and wild-type mice. Levels of miR-21, and mRNAs of selected miR-21 targets, miR-21 regulator STAT3 and myelin-related proteins were measured by qRT-PCR in the white matter (WM) adjacent to the left postmortem orbitofrontal cortex (OFC) of human subjects with major depressive disorder (MDD), alcoholism, comorbid MDD plus alcoholism (MDA) and non-psychiatric control subjects. MiR-21/LacZ was highly expressed in cell bodies of WM and myelinated portions of gray matter (GM). Labeled cell bodies were identified as oligodendrocytes, while miR-21/LacZ was barely detectable in other cell types. MiR-21, as well as the mRNAs of several myelin-related proteins, were reduced in the WM of subjects with MDD and alcoholism. MiR-21 positively correlated with mRNA of myelin-related proteins and astrocytic GFAP. High expression of miR-21 in adult oligodendrocytes and the correlation of miR-21 decrease with mRNA of some myelin proteins, regulator STAT3, and oligodendrocyte-related transcription factors suggest an involvement of miR-21 in WM alterations in depression and alcoholism.
Collapse
Affiliation(s)
- José Javier Miguel-Hidalgo
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA.
| | - Katherine O. Hall
- Department of Psychiatry and Human Behavior at the University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Hannah Bonner
- Department of Psychiatry and Human Behavior at the University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Anna M. Roller
- Department of Psychiatry and Human Behavior at the University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Maryam Syed
- Department of Biochemistry at the University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Casey J. Park
- Department of Biochemistry at the University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Jana P. Ball
- Department of Biochemistry at the University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Marc E. Rothenberg
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Craig A. Stockmeier
- Department of Psychiatry and Human Behavior at the University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Damian G. Romero
- Department of Biochemistry at the University of Mississippi Medical Center, Jackson, Mississippi, USA
| |
Collapse
|
34
|
Cirnigliaro M, Barbagallo C, Gulisano M, Domini CN, Barone R, Barbagallo D, Ragusa M, Di Pietro C, Rizzo R, Purrello M. Expression and Regulatory Network Analysis of miR-140-3p, a New Potential Serum Biomarker for Autism Spectrum Disorder. Front Mol Neurosci 2017; 10:250. [PMID: 28848387 PMCID: PMC5554380 DOI: 10.3389/fnmol.2017.00250] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 07/25/2017] [Indexed: 12/21/2022] Open
Abstract
Given its prevalence and social impact, Autism Spectrum Disorder (ASD) is drawing much interest. Molecular basis of ASD is heterogeneous and only partially known. Many factors, including disorders comorbid with ASD, like TS (Tourette Syndrome), complicate ASD behavior-based diagnosis and make it vulnerable to bias. To further investigate ASD etiology and to identify potential biomarkers to support its precise diagnosis, we used TaqMan Low Density Array technology to profile serum miRNAs from ASD, TS, and TS+ASD patients, and unaffected controls (NCs). Through validation assays in 30 ASD, 24 TS, and 25 TS+ASD patients and 25 NCs, we demonstrated that miR-140-3p is upregulated in ASD vs.: NC, TS, and TS+ASD (Tukey's test, p-values = 0.03, = 0.01, < 0.0001, respectively). ΔCt values for miR-140-3p and YGTSS (Yale Global Tic Severity Scale) scores are positively correlated (Spearman r = 0.33; Benjamini-Hochberg p = 0.008) and show a linear relationship (p = 0.002). Network functional analysis showed that nodes controlled by miR-140-3p, especially CD38 and NRIP1 which are its validated targets, are involved in processes convergingly dysregulated in ASD, such as synaptic plasticity, immune response, and chromatin binding. Biomarker analysis proved that serum miR-140-3p can discriminate among: (1) ASD and NC (Area under the ROC curve, AUC: 0.70; sensitivity: 63.33%; specificity: 68%); (2) ASD and TS (AUC: 0.72; sensitivity: 66.66%; specificity: 70.83%); (3) ASD and TS+ASD (AUC: 0.78; sensitivity: 73.33%; specificity: 76%). Characterization of miR-140-3p network would contribute to further clarify ASD etiology. Serum miR-140-3p could represent a potential non-invasive biomarker for ASD, easy to test through liquid biopsy.
Collapse
Affiliation(s)
- Matilde Cirnigliaro
- Section of Biology and Genetics Giovanni Sichel, Department of Biomedical and Biotechnological Sciences, University of CataniaCatania, Italy
| | - Cristina Barbagallo
- Section of Biology and Genetics Giovanni Sichel, Department of Biomedical and Biotechnological Sciences, University of CataniaCatania, Italy
| | - Mariangela Gulisano
- Section of Child and Adolescent Psychiatry, Department of Clinical and Experimental Medicine, University of CataniaCatania, Italy
| | - Carla N Domini
- Section of Child and Adolescent Psychiatry, Department of Clinical and Experimental Medicine, University of CataniaCatania, Italy
| | - Rita Barone
- Section of Child and Adolescent Psychiatry, Department of Clinical and Experimental Medicine, University of CataniaCatania, Italy
| | - Davide Barbagallo
- Section of Biology and Genetics Giovanni Sichel, Department of Biomedical and Biotechnological Sciences, University of CataniaCatania, Italy
| | - Marco Ragusa
- Section of Biology and Genetics Giovanni Sichel, Department of Biomedical and Biotechnological Sciences, University of CataniaCatania, Italy.,Associazione Oasi Maria SS. Onlus (IRCCS), Institute for Research on Mental Retardation and Brain AgingTroina, Italy
| | - Cinzia Di Pietro
- Section of Biology and Genetics Giovanni Sichel, Department of Biomedical and Biotechnological Sciences, University of CataniaCatania, Italy
| | - Renata Rizzo
- Section of Child and Adolescent Psychiatry, Department of Clinical and Experimental Medicine, University of CataniaCatania, Italy
| | - Michele Purrello
- Section of Biology and Genetics Giovanni Sichel, Department of Biomedical and Biotechnological Sciences, University of CataniaCatania, Italy
| |
Collapse
|
35
|
Mandal C, Halder D, Jung KH, Chai YG. Maternal alcohol consumption and altered miRNAs in the developing fetus: Context and future perspectives. J Appl Toxicol 2017; 38:100-107. [PMID: 28677831 DOI: 10.1002/jat.3504] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 05/29/2017] [Accepted: 05/29/2017] [Indexed: 12/11/2022]
Abstract
Alcohol is a teratogenic agent that can cause a wide range of developmental disorders, and sometimes, the effects persist throughout an individual's lifetime. Researchers have shown the involvement of epigenetic mechanisms in alcohol-mediated disorders. Non-coding RNAs are one of the major sources of epigenetic modifications, especially microRNAs. The association of microRNAs with alcohol consumption leads to a new focus on finding the molecular mechanisms of alcohol toxicity. It has been suggested that alcohol alters the relative expression of microRNAs and regulates target mRNA expression in both in vitro and in vivo models. Currently, we lack information regarding the relationship between altered microRNA expression and disease phenotypes in alcohol-mediated disorders. In this review, we tried to gather all of the available information about the alcohol-mediated dysregulation of microRNA expression in utero. We hope that our efforts will help future researchers identify major microRNAs in the field of prenatal alcohol toxicity and related therapeutics.
Collapse
Affiliation(s)
- Chanchal Mandal
- Department of Molecular and Life Science, Hanyang University, Ansan, Republic of Korea
| | - Debasish Halder
- Department of Molecular and Life Science, Hanyang University, Ansan, Republic of Korea
| | - Kyoung Hwa Jung
- Department of Molecular and Life Science, Hanyang University, Ansan, Republic of Korea.,Institute of Natural Science and Technology, Hanyang University, Ansan, Republic of Korea
| | - Young Gyu Chai
- Department of Molecular and Life Science, Hanyang University, Ansan, Republic of Korea.,Department of Bionanotechnology, Hanyang University, Seoul, Republic of Korea
| |
Collapse
|
36
|
Zheng H, Zou AE, Saad MA, Wang XQ, Kwok JG, Korrapati A, Li P, Kisseleva T, Wang-Rodriguez J, Ongkeko WM. Alcohol-dysregulated microRNAs in hepatitis B virus-related hepatocellular carcinoma. PLoS One 2017; 12:e0178547. [PMID: 28562643 PMCID: PMC5451132 DOI: 10.1371/journal.pone.0178547] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 05/15/2017] [Indexed: 12/17/2022] Open
Abstract
Alcohol consumption and chronic hepatitis B virus (HBV) infection are two well-established risk factors for Hepatocellular carcinoma (HCC); however, there remains a limited understanding of the molecular pathway behind the pathogenesis and progression behind HCC, and how alcohol promotes carcinogenesis in the context of HBV+ HCC. Using next-generation sequencing data from 130 HCC patients and 50 normal liver tissues, we identified a panel of microRNAs that are significantly dysregulated by alcohol consumption in HBV+ patients. In particular, two microRNAs, miR-944 and miR-223-3p, showed remarkable correlation with clinical indication and genomic alterations. We confirmed the dysregulation of these two microRNAs in liver cell lines treated by alcohol and acetaldehyde, and showed that manipulation of miR-223-3p and miR-944 expression induces significant changes in cellular proliferation, sensitivity to doxorubicin, and the expression of both direct-binding and downstream mRNA targets. Together, the results of this study suggest that alcohol consumption in HBV+ HCCs regulates microRNAs that likely play previously uncharacterized roles in the alcohol-associated carcinogenesis of HCC, and future studies of these microRNAs may be valuable for furthering the understanding and treatment of alcohol and HBV-associated HCC.
Collapse
Affiliation(s)
- Hao Zheng
- Department of Surgery, University of California, San Diego, La Jolla, California, United States of America
| | - Angela E. Zou
- Department of Surgery, University of California, San Diego, La Jolla, California, United States of America
| | - Maarouf A. Saad
- School of medicine, Yale University, New Haven, Connecticut, United States of America
| | - Xiao Qi Wang
- Department of Surgery, The University of Hong Kong, Hong Kong, China
| | - James G. Kwok
- Department of Surgery, University of California, San Diego, La Jolla, California, United States of America
| | - Avinaash Korrapati
- Department of Surgery, University of California, San Diego, La Jolla, California, United States of America
| | - Pinxue Li
- Department of Surgery, University of California, San Diego, La Jolla, California, United States of America
| | - Tatiana Kisseleva
- Department of Surgery, University of California, San Diego, La Jolla, California, United States of America
| | - Jessica Wang-Rodriguez
- Department of Pathology, Veterans Administration Medical Center, San Diego, La Jolla, California, United States of America
- Department of Pathology, University of California, San Diego, La Jolla, California, United States of America
| | - Weg M. Ongkeko
- Department of Surgery, University of California, San Diego, La Jolla, California, United States of America
| |
Collapse
|
37
|
Burrowes SG, Salem NA, Tseng AM, Balaraman S, Pinson MR, Garcia C, Miranda RC. The BAF (BRG1/BRM-Associated Factor) chromatin-remodeling complex exhibits ethanol sensitivity in fetal neural progenitor cells and regulates transcription at the miR-9-2 encoding gene locus. Alcohol 2017; 60:149-158. [PMID: 28438527 DOI: 10.1016/j.alcohol.2017.01.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 12/15/2016] [Accepted: 01/03/2017] [Indexed: 12/25/2022]
Abstract
Fetal alcohol spectrum disorders are a leading cause of intellectual disability worldwide. Previous studies have shown that developmental ethanol exposure results in loss of microRNAs (miRNAs), including miR-9, and loss of these miRNAs, in turn, mediates some of ethanol's teratogenic effects in the developing brain. We previously found that ethanol increased methylation at the miR-9-2 encoding gene locus in mouse fetal neural stem cells (NSC), advancing a mechanism for epigenetic silencing of this locus and consequently, miR-9 loss in NSCs. Therefore, we assessed the role of the BAF (BRG1/BRM-Associated Factor) complex, which disassembles nucleosomes to facilitate access to chromatin, as an epigenetic mediator of ethanol's effects on miR-9. Chromatin immunoprecipitation and DNAse I-hypersensitivity analyses showed that the BAF complex was associated with both transcriptionally accessible and heterochromatic regions of the miR-9-2 locus, and that disintegration of the BAF complex by combined knockdown of BAF170 and BAF155 resulted in a significant decrease in miR-9. We hypothesized that ethanol exposure would result in loss of BAF-complex function at the miR-9-2 locus. However, ethanol exposure significantly increased mRNA transcripts for maturation-associated BAF-complex members BAF170, SS18, ARID2, BAF60a, BRM/BAF190b, and BAF53b. Ethanol also significantly increased BAF-complex binding within an intron containing a CpG island and in the terminal exon encoding precursor (pre)-miR-9-2. These data suggest that the BAF complex may adaptively respond to ethanol exposure to protect against a complete loss of miR-9-2 in fetal NSCs. Chromatin remodeling factors may adapt to the presence of a teratogen, to maintain transcription of critical miRNA regulatory pathways.
Collapse
|
38
|
Balaraman S, Idrus NM, Miranda RC, Thomas JD. Postnatal choline supplementation selectively attenuates hippocampal microRNA alterations associated with developmental alcohol exposure. Alcohol 2017; 60:159-167. [PMID: 28433422 PMCID: PMC5559286 DOI: 10.1016/j.alcohol.2016.12.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Revised: 12/28/2016] [Accepted: 12/29/2016] [Indexed: 11/25/2022]
Abstract
Prenatal alcohol exposure can result in a range of physical, neuropathological, and behavioral alterations, collectively termed fetal alcohol spectrum disorders (FASD). We have shown that supplementation with the nutrient choline reduces the severity of developmental alcohol-associated deficits in hippocampal-dependent behaviors and normalizes some aspects of hippocampal cholinergic development and DNA methylation patterns. Alcohol's developmental effects may also be mediated, in part, by altering microRNAs (miRNAs) that serve as negative regulators of gene translation. To determine whether choline supplementation alters ethanol's long-lasting effects on miRNAs, Sprague-Dawley rats were exposed to 5.25 g/kg/day ethanol from postnatal days (PD) 4-9 via intubation; controls received sham intubations. Subjects were treated with choline chloride (100 mg/kg/day) or saline vehicle subcutaneously (s.c.) from PD 4-21. On PD 22, subjects were sacrificed, and RNA was isolated from the hippocampus. MiRNA expression was assessed with TaqMan Human MicroRNA Panel Low-Density Arrays. Ethanol significantly increased miRNA expression variance, an effect that was attenuated with choline supplementation. Cluster analysis of stably expressed miRNAs that exceeded an ANOVA p < 0.05 criterion indicated that for both male and female offspring, control and ethanol-exposed groups were most dissimilar from each other, with choline-supplemented groups in between. MiRNAs that expressed an average 2-fold change due to ethanol exposure were further analyzed to identify which ethanol-sensitive miRNAs were protected by choline supplementation. We found that at a false discovery rate (FDR)-adjusted criterion of p < 0.05, miR-200c was induced by ethanol exposure and that choline prevented this effect. Collectively, our data show that choline supplementation can normalize disturbances in miRNA expression following developmental alcohol exposure and can protect specific miRNAs from induction by ethanol. These findings have important implications for the mechanisms by which choline may serve as a potential treatment for FASD.
Collapse
Affiliation(s)
- Sridevi Balaraman
- Department of Neuroscience and Experimental Therapeutics, Women's Health in Neuroscience Program, College of Medicine, Texas A&M Health Science Center, College Station, TX 77843, USA
| | - Nirelia M Idrus
- Center for Behavioral Teratology, Department of Psychology, San Diego State University, San Diego, CA 92120, USA
| | - Rajesh C Miranda
- Department of Neuroscience and Experimental Therapeutics, Women's Health in Neuroscience Program, College of Medicine, Texas A&M Health Science Center, College Station, TX 77843, USA
| | - Jennifer D Thomas
- Center for Behavioral Teratology, Department of Psychology, San Diego State University, San Diego, CA 92120, USA.
| |
Collapse
|
39
|
Puhakka N, Bot AM, Vuokila N, Debski KJ, Lukasiuk K, Pitkänen A. Chronically dysregulated NOTCH1 interactome in the dentate gyrus after traumatic brain injury. PLoS One 2017; 12:e0172521. [PMID: 28273100 PMCID: PMC5342204 DOI: 10.1371/journal.pone.0172521] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 02/06/2017] [Indexed: 12/13/2022] Open
Abstract
Traumatic brain injury (TBI) can result in several dentate gyrus-regulated disabilities. Almost nothing is known about the chronic molecular changes after TBI, and their potential as treatment targets. We hypothesized that chronic transcriptional alterations after TBI are under microRNA (miRNA) control. Expression of miRNAs and their targets in the dentate gyrus was analyzed using microarrays at 3 months after experimental TBI. Of 305 miRNAs present on the miRNA-array, 12 were downregulated (p<0.05). In parallel, 75 of their target genes were upregulated (p<0.05). A bioinformatics analysis of miRNA targets highlighted the dysregulation of the transcription factor NOTCH1 and 39 of its target genes (NOTCH1 interactome). Validation assays confirmed downregulation of miR-139-5p, upregulation of Notch1 and its activated protein, and positive enrichment of NOTCH1 target gene expression. These findings demonstrate that miRNA-based transcriptional regulation can be present at chronic time points after TBI, and highlight the NOTCH1 interactome as one of the mechanisms behind the dentate gyrus pathology-related morbidities.
Collapse
Affiliation(s)
- Noora Puhakka
- Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Anna Maria Bot
- The Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Niina Vuokila
- Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Konrad Jozef Debski
- The Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Katarzyna Lukasiuk
- The Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Asla Pitkänen
- Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- * E-mail:
| |
Collapse
|
40
|
Lussier AA, Weinberg J, Kobor MS. Epigenetics studies of fetal alcohol spectrum disorder: where are we now? Epigenomics 2017; 9:291-311. [PMID: 28234026 PMCID: PMC5549650 DOI: 10.2217/epi-2016-0163] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Adverse in utero events can alter the development and function of numerous physiological systems, giving rise to lasting neurodevelopmental deficits. In particular, data have shown that prenatal alcohol exposure can reprogram neurobiological systems, altering developmental trajectories and resulting in increased vulnerability to adverse neurobiological, behavioral and health outcomes. Increasing evidence suggests that epigenetic mechanisms are potential mediators for the reprogramming of neurobiological systems, as they may provide a link between the genome, environmental conditions and neurodevelopmental outcomes. This review outlines the current state of epigenetic research in fetal alcohol spectrum disorder, highlighting the role of epigenetic mechanisms in the reprogramming of neurobiological systems by alcohol and as potential diagnostic tools for fetal alcohol spectrum disorder. We also present an assessment of the current limitations in studies of prenatal alcohol exposure, and highlight the future steps needed in the field.
Collapse
Affiliation(s)
- Alexandre A Lussier
- Department of Medical Genetics, Centre for Molecular Medicine & Therapeutics, British Columbia Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Cellular & Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Joanne Weinberg
- Department of Cellular & Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michael S Kobor
- Department of Medical Genetics, Centre for Molecular Medicine & Therapeutics, British Columbia Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada.,Human Early Learning Partnership, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
41
|
Gardiner AS, Gutierrez HL, Luo L, Davies S, Savage DD, Bakhireva LN, Perrone-Bizzozero NI. Alcohol Use During Pregnancy is Associated with Specific Alterations in MicroRNA Levels in Maternal Serum. Alcohol Clin Exp Res 2016; 40:826-37. [PMID: 27038596 PMCID: PMC4825401 DOI: 10.1111/acer.13026] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 01/25/2016] [Indexed: 12/12/2022]
Abstract
Background Given the challenges of confirming prenatal alcohol exposure (PAE) during pregnancy using currently established biomarkers of alcohol consumption, we examined whether serum microRNAs (miRNAs) may serve as stable biomarkers for PAE. Alterations in the levels of specific circulating miRNAs have been associated with various disease states and in animal models of fetal alcohol spectrum disorder. Methods Pregnant women in this prospective study were recruited from substance abuse and general maternity clinics affiliated with the University of New Mexico. Serum was collected at the time of admission for delivery from 14 subjects who reported ≥1 binge‐drinking episode or ≥3 drinks/wk during pregnancy and 16 subjects who reported abstinence during pregnancy and tested negative for 5 ethanol biomarkers. Total RNA was isolated from serum and used for microarray analysis. Results False discovery rate‐corrected analyses of covariance revealed that 55 miRNAs were significantly altered between the 2 groups. Hierarchical clustering using only the significantly altered miRNAs grouped samples into alcohol‐consuming and non‐alcohol‐consuming individuals. Discriminant analysis then identified miRs‐122*, ‐126, ‐216b, ‐221*, ‐3119, ‐3942‐5p, ‐4704‐3p, ‐4743, ‐514‐5p, and ‐602 as the top 10 discriminators between the 2 groups. Ingenuity Pathway Analysis of putative miRNA targets illustrated that miRNAs identified in this study are involved in biological pathways that mediate the effects of alcohol, such as brain‐derived neurotrophic factor, ERK1/2, and PI3K/AKT signaling. Conclusions This is the first report of alterations in serum miRNA expression that are associated with alcohol use during human pregnancy. These results suggest that serum miRNAs could be useful as biomarkers of alcohol exposure.
Collapse
Affiliation(s)
- Amy S Gardiner
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Hilda L Gutierrez
- Department of Pharmacy Practice and Administrative Sciences, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Li Luo
- Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Suzy Davies
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Daniel D Savage
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, New Mexico.,Department of Pediatrics, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Ludmila N Bakhireva
- Department of Pharmacy Practice and Administrative Sciences, University of New Mexico Health Sciences Center, Albuquerque, New Mexico.,Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico.,Department of Family and Community Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Nora I Perrone-Bizzozero
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, New Mexico.,Department of Psychiatry and Behavioral Sciences, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| |
Collapse
|
42
|
Plasma miRNA Profiles in Pregnant Women Predict Infant Outcomes following Prenatal Alcohol Exposure. PLoS One 2016; 11:e0165081. [PMID: 27828986 PMCID: PMC5102408 DOI: 10.1371/journal.pone.0165081] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Accepted: 10/05/2016] [Indexed: 12/04/2022] Open
Abstract
Fetal alcohol spectrum disorders (FASD) are difficult to diagnose since many heavily exposed infants, at risk for intellectual disability, do not exhibit craniofacial dysmorphology or growth deficits. Consequently, there is a need for biomarkers that predict disability. In both animal models and human studies, alcohol exposure during pregnancy resulted in significant alterations in circulating microRNAs (miRNAs) in maternal blood. In the current study, we asked if changes in plasma miRNAs in alcohol-exposed pregnant mothers, either alone or in conjunction with other clinical variables, could predict infant outcomes. Sixty-eight pregnant women at two perinatal care clinics in western Ukraine were recruited into the study. Detailed health and alcohol consumption histories, and 2nd and 3rd trimester blood samples were obtained. Birth cohort infants were assessed by a geneticist and classified as unexposed (UE), heavily prenatally exposed and affected (HEa) or heavily exposed but apparently unaffected (HEua). MiRNAs were assessed in plasma samples using qRT-PCR arrays. ANOVA models identified 11 miRNAs that were all significantly elevated in maternal plasma from the HEa group relative to HEua and UE groups. In a random forest analysis classification model, a combination of high variance miRNAs, smoking history and socioeconomic status classified membership in HEa and UE groups, with a misclassification rate of 13%. The RFA model also classified 17% of the HEua group as UE-like, whereas 83% were HEa-like, at least at one stage of pregnancy. Collectively our data indicate that maternal plasma miRNAs predict infant outcomes, and may be useful to classify difficult-to-diagnose FASD subpopulations.
Collapse
|
43
|
Epigenetic Mechanisms in Developmental Alcohol-Induced Neurobehavioral Deficits. Brain Sci 2016; 6:brainsci6020012. [PMID: 27070644 PMCID: PMC4931489 DOI: 10.3390/brainsci6020012] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 03/17/2016] [Accepted: 04/05/2016] [Indexed: 12/22/2022] Open
Abstract
Alcohol consumption during pregnancy and its damaging consequences on the developing infant brain are significant public health, social, and economic issues. The major distinctive features of prenatal alcohol exposure in humans are cognitive and behavioral dysfunction due to damage to the central nervous system (CNS), which results in a continuum of disarray that is collectively called fetal alcohol spectrum disorder (FASD). Many rodent models have been developed to understand the mechanisms of and to reproduce the human FASD phenotypes. These animal FASD studies have provided several molecular pathways that are likely responsible for the neurobehavioral abnormalities that are associated with prenatal alcohol exposure of the developing CNS. Recently, many laboratories have identified several immediate, as well as long-lasting, epigenetic modifications of DNA methylation, DNA-associated histone proteins and microRNA (miRNA) biogenesis by using a variety of epigenetic approaches in rodent FASD models. Because DNA methylation patterns, DNA-associated histone protein modifications and miRNA-regulated gene expression are crucial for synaptic plasticity and learning and memory, they can therefore offer an answer to many of the neurobehavioral abnormalities that are found in FASD. In this review, we briefly discuss the current literature of DNA methylation, DNA-associated histone proteins modification and miRNA and review recent developments concerning epigenetic changes in FASD.
Collapse
|
44
|
Liu R, Shi P, Nie Z, Liang H, Zhou Z, Chen W, Chen H, Dong C, Yang R, Liu S, Chen C. Mifepristone Suppresses Basal Triple-Negative Breast Cancer Stem Cells by Down-regulating KLF5 Expression. Theranostics 2016; 6:533-44. [PMID: 26941846 PMCID: PMC4775863 DOI: 10.7150/thno.14315] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 01/09/2016] [Indexed: 12/31/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is currently the most malignant subtype of breast cancers without effective targeted therapies. Mifepristone (MIF), a drug regularly used for abortion, has been reported to have anti-tumor activity in multiple hormone-dependent cancers, including luminal type breast cancers. In this study, we showed that MIF suppressed tumor growth of the TNBC cell lines and patient-derived xenografts in NOD-SCID mice. Furthermore, MIF reduced the TNBC cancer stem cell (CSC) population through down-regulating KLF5 expression, a stem cell transcription factor over-expressed in basal type TNBC and promoting cell proliferation, survival and stemness. Interestingly, MIF suppresses the expression of KLF5 through inducing the expression of miR-153. Consistently, miR-153 decreases CSC and miR-153 inhibitor rescued MIF-induced down-regulation of the KLF5 protein level and CSC ratio. Taken together, our findings suggest that MIF inhibits basal TNBC via the miR-153/KLF5 axis and MIF may be used for the treatment of TNBC.
Collapse
|
45
|
M. Carballosa C, M. Greenberg J, S. Cheung H. Expression and function of nicotinic acetylcholine receptors in stem cells. AIMS BIOENGINEERING 2016. [DOI: 10.3934/bioeng.2016.3.245] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
|
46
|
Codocedo JF, Inestrosa NC. Environmental control of microRNAs in the nervous system: Implications in plasticity and behavior. Neurosci Biobehav Rev 2015; 60:121-38. [PMID: 26593111 DOI: 10.1016/j.neubiorev.2015.10.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 10/24/2015] [Accepted: 10/26/2015] [Indexed: 02/07/2023]
Abstract
The discovery of microRNAs (miRNAs) a little over 20 years ago was revolutionary given that miRNAs are essential to numerous physiological and physiopathological processes. Currently, several aspects of the biogenic process of miRNAs and of the translational repression mechanism exerted on their targets mRNAs are known in detail. In fact, the development of bioinformatics tools for predicting miRNA targets has established that miRNAs have the potential to regulate almost all known biological processes. Therefore, the identification of the signals and molecular mechanisms that regulate miRNA function is relevant to understanding the role of miRNAs in both pathological and adaptive processes. Recently, a series of studies has focused on miRNA expression in the brain, establishing that their levels are altered in response to various environmental factors (EFs), such as light, sound, odorants, nutrients, drugs and stress. In this review, we discuss how exposure to various EFs modulates the expression and function of several miRNAs in the nervous system and how this control determines adaptation to their environment, behavior and disease state.
Collapse
Affiliation(s)
- Juan F Codocedo
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nibaldo C Inestrosa
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile; Centre for Healthy Brain Ageing, School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, Australia; Centro UC Síndrome de Down, Pontificia Universidad Católica de Chile, Santiago, Chile; Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile.
| |
Collapse
|
47
|
Bosch PJ, Benton MC, Macartney-Coxson D, Kivell BM. mRNA and microRNA analysis reveals modulation of biochemical pathways related to addiction in the ventral tegmental area of methamphetamine self-administering rats. BMC Neurosci 2015; 16:43. [PMID: 26188473 PMCID: PMC4506769 DOI: 10.1186/s12868-015-0186-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 07/14/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Methamphetamine is a highly addictive central nervous system stimulant with increasing levels of abuse worldwide. Alterations to mRNA and miRNA expression within the mesolimbic system can affect addiction-like behaviors and thus play a role in the development of drug addiction. While many studies have investigated the effects of high-dose methamphetamine, and identified neurotoxic effects, few have looked at the role that persistent changes in gene regulation play following methamphetamine self-administration. Therefore, the aim of this study was to identify RNA changes in the ventral tegmental area following methamphetamine self-administration. We performed microarray analyses on RNA extracted from the ventral tegmental area of Sprague-Dawley rats following methamphetamine self-administration training (2 h/day) and 14 days of abstinence. RESULTS We identified 78 miRNA and 150 mRNA transcripts that were differentially expressed (fdr adjusted p < 0.05, absolute log2 fold change >0.5); these included genes not previously associated with addiction (miR-125a-5p, miR-145 and Foxa1), loci encoding receptors related to drug addiction behaviors and genes with previously recognized roles in addiction such as miR-124, miR-181a, DAT and Ret. CONCLUSION This study provides insight into the effects of methamphetamine on RNA expression in a key brain region associated with addiction, highlighting the possibility that persistent changes in the expression of genes with both known and previously unknown roles in addiction occur.
Collapse
Affiliation(s)
- P J Bosch
- Centre for Biodiscovery, School of Biological Sciences, Victoria University of Wellington, Kelburn Parade, PO Box 600, Wellington, 6140, New Zealand.
| | - M C Benton
- Institute of Environmental Science and Research, Wellington, New Zealand.
- Genomics Research Centre, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia.
| | - D Macartney-Coxson
- Institute of Environmental Science and Research, Wellington, New Zealand.
| | - B M Kivell
- Centre for Biodiscovery, School of Biological Sciences, Victoria University of Wellington, Kelburn Parade, PO Box 600, Wellington, 6140, New Zealand.
| |
Collapse
|
48
|
Chen K, He H, Xie Y, Zhao L, Zhao S, Wan X, Yang W, Mo Z. miR-125a-3p and miR-483-5p promote adipogenesis via suppressing the RhoA/ROCK1/ERK1/2 pathway in multiple symmetric lipomatosis. Sci Rep 2015; 5:11909. [PMID: 26148871 PMCID: PMC4493643 DOI: 10.1038/srep11909] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 04/22/2015] [Indexed: 01/08/2023] Open
Abstract
Multiple symmetric lipomatosis (MSL) is a rare disease characterized by symmetric and abnormal distribution of subcutaneous adipose tissue (SAT); however, the etiology is largely unknown. We report here that miR-125a-3p and miR-483-5p are upregulated in the SAT of MSL patients, promoting adipogenesis through suppressing the RhoA/ROCK1/ERK1/2 pathway. TaqMan microRNA (miR) array analysis revealed that 18 miRs were upregulated in the SAT of MSL patients. Transfection of human adipose-derived mesenchymal stem cells (hADSCs) with the individual agomirs of these 18 miRs showed that miR-125a-3p and miR-483-5p significantly promoted adipogenesis. A dual-luciferase assay showed that RhoA and ERK1 were the targets of miR-125a-3p and miR-483-5p, respectively. Moreover, transfection of hADSCs with mimics of miR-125a-3p and miR-483-5p resulted in a pronounced decrease of ERK1/2 phosphorylation in the nucleus; conversely, transfection of hADSCs with inhibitors of miR-125a-3p and miR-483-5p led to a significant increase of ERK1/2 phosphorylation in the nucleus. Most importantly, we found that miR-125a-3p and miR-483-5p promoted de novo adipose tissue formation in nude mice. These results demonstrated that miR-125a-3p and miR-483-5p coordinately promoted adipogenesis through suppressing the RhoA/ROCK1/ERK1/2 pathway. Our findings may provide novel strategies for the management and treatment of MSL or obesity.
Collapse
Affiliation(s)
- Ke Chen
- Department of Endocrinology, Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, China
| | - Honghui He
- Department of Endocrinology, Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, China
| | - Yanhong Xie
- Department of Endocrinology, Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, China
| | - Liling Zhao
- Department of Endocrinology, Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, China
| | - Shaoli Zhao
- Department of Endocrinology, Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, China
| | - Xinxing Wan
- Department of Endocrinology, Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, China
| | - Wenjun Yang
- Department of Endocrinology, Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, China
| | - Zhaohui Mo
- Department of Endocrinology, Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, China
| |
Collapse
|
49
|
Taki FA, Pan X, Zhang B. Revisiting Chaos Theorem to Understand the Nature of miRNAs in Response to Drugs of Abuse. J Cell Physiol 2015; 230:2857-68. [PMID: 25966899 DOI: 10.1002/jcp.25037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 05/05/2015] [Indexed: 11/08/2022]
Abstract
Just like Matryoshka dolls, biological systems follow a hierarchical order that is based on dynamic bidirectional communication among its components. In addition to the convoluted inter-relationships, the complexity of each component spans several folds. Therefore, it becomes rather challenging to investigate phenotypes resulting from these networks as it requires the integration of reductionistic and holistic approaches. One dynamic system is the transcriptome which comprises a variety of RNA species. Some, like microRNAs, have recently received a lot of attention. miRNAs are very pleiotropic and have been considered as therapeutic and diagnostic candidates in the biomedical fields. In this review, we survey miRNA profiles in response to drugs of abuse (DA) using 118 studies. After providing a summary of miRNAs related to substance use disorders (SUD), general patterns of miRNA signatures are compared among studies for single or multiple drugs of abuse. Then, current challenges and drawbacks in the field are discussed. Finally, we provide support for considering miRNAs as a chaotic system in normal versus disrupted states particularly in SUD and propose an integrative approach for studying and analyzing miRNA data.
Collapse
Affiliation(s)
- Faten A Taki
- Department of Biology, East Carolina University, Greenville, North Carolina
| | - Xiaoping Pan
- Department of Biology, East Carolina University, Greenville, North Carolina
| | - Baohong Zhang
- Department of Biology, East Carolina University, Greenville, North Carolina
| |
Collapse
|
50
|
Taki FA, Pan X, Lee MH, Zhang B. Nicotine exposure and transgenerational impact: a prospective study on small regulatory microRNAs. Sci Rep 2014; 4:7513. [PMID: 25515333 PMCID: PMC4894410 DOI: 10.1038/srep07513] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 11/24/2014] [Indexed: 12/22/2022] Open
Abstract
Early developmental stages are highly sensitive to stress and it has been reported that pre-conditioning with tobacco smoking during adolescence predisposes those youngsters to become smokers as adults. However, the molecular mechanisms of nicotine-induced transgenerational consequences are unknown. In this study, we genome-widely investigated the impact of nicotine exposure on small regulatory microRNAs (miRNAs) and its implication on health disorders at a transgenerational aspect. Our results demonstrate that nicotine exposure, even at the low dose, affected the global expression profiles of miRNAs not only in the treated worms (F0 parent generation) but also in two subsequent generations (F1 and F2, children and grandchildren). Some miRNAs were commonly affected by nicotine across two or more generations while others were specific to one. The general miRNA patterns followed a “two-hit” model as a function of nicotine exposure and abstinence. Target prediction and pathway enrichment analyses showed daf-4, daf-1, fos-1, cmk-1, and unc-30 to be potential effectors of nicotine addiction. These genes are involved in physiological states and phenotypes that paralleled previously published nicotine induced behavior. Our study offered new insights and further awareness on the transgenerational effects of nicotine exposed during the vulnerable post-embryonic stages, and identified new biomarkers for nicotine addiction.
Collapse
Affiliation(s)
- Faten A Taki
- Department of Biology, East Carolina University, Greenville, NC 27858, USA
| | - Xiaoping Pan
- Department of Biology, East Carolina University, Greenville, NC 27858, USA
| | - Myon-Hee Lee
- Department of Medicine, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Baohong Zhang
- Department of Biology, East Carolina University, Greenville, NC 27858, USA
| |
Collapse
|