1
|
Parkins EV, Gross C. Small Differences and Big Changes: The Many Variables of MicroRNA Expression and Function in the Brain. J Neurosci 2024; 44:e0365242024. [PMID: 39111834 PMCID: PMC11308354 DOI: 10.1523/jneurosci.0365-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 05/15/2024] [Accepted: 05/23/2024] [Indexed: 08/10/2024] Open
Abstract
MicroRNAs are emerging as crucial regulators within the complex, dynamic environment of the synapse, and they offer a promising new avenue for the treatment of neurological disease. These small noncoding RNAs modify gene expression in several ways, including posttranscriptional modulation via binding to complementary and semicomplementary sites on target mRNAs. This rapid, finely tuned regulation of gene expression is essential to meet the dynamic demands of the synapse. Here, we provide a detailed review of the multifaceted world of synaptic microRNA regulation. We discuss the many mechanisms by which microRNAs regulate gene expression at the synapse, particularly in the context of neuronal plasticity. We also describe the various factors, such as age, sex, and neurological disease, that can influence microRNA expression and activity in neurons. In summary, microRNAs play a crucial role in the intricate and quickly changing functional requirements of the synapse, and context is essential in the study of microRNAs and their potential therapeutic applications.
Collapse
Affiliation(s)
- Emma V Parkins
- University of Cincinnati Neuroscience Graduate Program, Cincinnati, Ohio 45229
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
| | - Christina Gross
- University of Cincinnati Neuroscience Graduate Program, Cincinnati, Ohio 45229
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio 45229
| |
Collapse
|
2
|
Martinez B, Peplow PV. MicroRNAs as potential biomarkers for diagnosis of schizophrenia and influence of antipsychotic treatment. Neural Regen Res 2024; 19:1523-1531. [PMID: 38051895 PMCID: PMC10883514 DOI: 10.4103/1673-5374.387966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 09/26/2023] [Indexed: 12/07/2023] Open
Abstract
ABSTRACT Characterized by positive symptoms (such as changes in behavior or thoughts, including delusions and hallucinations), negative symptoms (such as apathy, anhedonia, and social withdrawal), and cognitive impairments, schizophrenia is a chronic, severe, and disabling mental disorder with late adolescence or early adulthood onset. Antipsychotics are the most commonly used drugs to treat schizophrenia, but those currently in use do not fully reverse all three types of symptoms characterizing this condition. Schizophrenia is frequently misdiagnosed, resulting in a delay of or inappropriate treatment. Abnormal expression of microRNAs is connected to brain development and disease and could provide novel biomarkers for the diagnosis and prognosis of schizophrenia. The recent studies reviewed included microRNA profiling in blood- and urine-based materials and nervous tissue materials. From the studies that had validated the preliminary findings, potential candidate biomarkers for schizophrenia in adults could be miR-22-3p, -30e-5p, -92a-3p, -148b-5p, -181a-3p, -181a-5p, -181b-5p, -199b-5p, -137 in whole blood, and miR-130b, -193a-3p in blood plasma. Antipsychotic treatment of schizophrenia patients was found to modulate the expression of certain microRNAs including miR-130b, -193a-3p, -132, -195, -30e, -432 in blood plasma. Further studies are warranted with adolescents and young adults having schizophrenia and consideration should be given to using animal models of the disorder to investigate the effect of suppressing or overexpressing specific microRNAs.
Collapse
Affiliation(s)
- Bridget Martinez
- Department of Pharmacology, University of Nevada-Reno, Reno, NV, USA
- Department of Medicine, University of Nevada-Reno, Reno, NV, USA
| | - Philip V Peplow
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| |
Collapse
|
3
|
Geleta U, Prajapati P, Bachstetter A, Nelson PT, Wang WX. Sex-Biased Expression and Response of microRNAs in Neurological Diseases and Neurotrauma. Int J Mol Sci 2024; 25:2648. [PMID: 38473893 PMCID: PMC10931569 DOI: 10.3390/ijms25052648] [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: 01/27/2024] [Revised: 02/16/2024] [Accepted: 02/21/2024] [Indexed: 03/14/2024] Open
Abstract
Neurological diseases and neurotrauma manifest significant sex differences in prevalence, progression, outcome, and therapeutic responses. Genetic predisposition, sex hormones, inflammation, and environmental exposures are among many physiological and pathological factors that impact the sex disparity in neurological diseases. MicroRNAs (miRNAs) are a powerful class of gene expression regulator that are extensively involved in mediating biological pathways. Emerging evidence demonstrates that miRNAs play a crucial role in the sex dimorphism observed in various human diseases, including neurological diseases. Understanding the sex differences in miRNA expression and response is believed to have important implications for assessing the risk of neurological disease, defining therapeutic intervention strategies, and advancing both basic research and clinical investigations. However, there is limited research exploring the extent to which miRNAs contribute to the sex disparities observed in various neurological diseases. Here, we review the current state of knowledge related to the sexual dimorphism in miRNAs in neurological diseases and neurotrauma research. We also discuss how sex chromosomes may contribute to the miRNA sexual dimorphism phenomenon. We attempt to emphasize the significance of sexual dimorphism in miRNA biology in human diseases and to advocate a gender/sex-balanced science.
Collapse
Affiliation(s)
- Urim Geleta
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY 40536, USA; (U.G.); (P.P.); (A.B.); (P.T.N.)
| | - Paresh Prajapati
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY 40536, USA; (U.G.); (P.P.); (A.B.); (P.T.N.)
| | - Adam Bachstetter
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY 40536, USA; (U.G.); (P.P.); (A.B.); (P.T.N.)
- Spinal Cord and Brain Injury Research Center, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Neuroscience, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Peter T. Nelson
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY 40536, USA; (U.G.); (P.P.); (A.B.); (P.T.N.)
- Spinal Cord and Brain Injury Research Center, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Pathology and Laboratory Medicine, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Wang-Xia Wang
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY 40536, USA; (U.G.); (P.P.); (A.B.); (P.T.N.)
- Spinal Cord and Brain Injury Research Center, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Pathology and Laboratory Medicine, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
4
|
Brown JS. Comparison of Oncogenes, Tumor Suppressors, and MicroRNAs Between Schizophrenia and Glioma: The Balance of Power. Neurosci Biobehav Rev 2023; 151:105206. [PMID: 37178944 DOI: 10.1016/j.neubiorev.2023.105206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 04/25/2023] [Accepted: 04/30/2023] [Indexed: 05/15/2023]
Abstract
The risk of cancer in schizophrenia has been controversial. Confounders of the issue are cigarette smoking in schizophrenia, and antiproliferative effects of antipsychotic medications. The author has previously suggested comparison of a specific cancer like glioma to schizophrenia might help determine a more accurate relationship between cancer and schizophrenia. To accomplish this goal, the author performed three comparisons of data; the first a comparison of conventional tumor suppressors and oncogenes between schizophrenia and cancer including glioma. This comparison determined schizophrenia has both tumor-suppressive and tumor-promoting characteristics. A second, larger comparison between brain-expressed microRNAs in schizophrenia with their expression in glioma was then performed. This identified a core carcinogenic group of miRNAs in schizophrenia offset by a larger group of tumor-suppressive miRNAs. This proposed "balance of power" between oncogenes and tumor suppressors could cause neuroinflammation. This was assessed by a third comparison between schizophrenia, glioma and inflammation in asbestos-related lung cancer and mesothelioma (ALRCM). This revealed that schizophrenia shares more oncogenic similarity to ALRCM than glioma.
Collapse
|
5
|
Singh RP, Hahn BH, Bischoff DS. Identification and Contribution of Inflammation-Induced Novel MicroRNA in the Pathogenesis of Systemic Lupus Erythematosus. Front Immunol 2022; 13:848149. [PMID: 35444657 PMCID: PMC9013931 DOI: 10.3389/fimmu.2022.848149] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 02/24/2022] [Indexed: 02/06/2023] Open
Abstract
Recently microRNAs (miRNAs) have been recognized as powerful regulators of many genes and pathways involved in the pathogenesis of inflammatory diseases including Systemic Lupus Erythematosus (SLE). SLE is an autoimmune disease characterized by production of various autoantibodies, inflammatory immune cells, and dysregulation of epigenetic changes. Several candidate miRNAs regulating inflammation and autoimmunity in SLE are described. In this study, we found significant increases in the expression of miR21, miR25, and miR186 in peripheral blood mononuclear cells (PBMCs) of SLE patients compared to healthy controls. However, miR146a was significantly decreased in SLE patients compared to healthy controls and was negatively correlated with plasma estradiol levels and with SLE disease activity scores (SLEDAI). We also found that protein levels of IL-12 and IL-21 were significantly increased in SLE patients as compared to healthy controls. Further, our data shows that protein levels of IL-12 were positively correlated with miR21 expression and protein levels of IL-21 positively correlated with miR25 and miR186 expression in SLE patients. In addition, we found that levels of miR21, miR25, and miR186 positively correlated with SLEDAI and miR146a was negatively correlated in SLE patients. Thus, our data shows a dynamic interplay between disease pathogenesis and miRNA expression. This study has translational potential and may identify novel therapeutic targets in patients with SLE.
Collapse
Affiliation(s)
- Ram P Singh
- Research Service, Veteran Administration Greater Los Angeles Healthcare System, Los Angeles, CA, United States.,Division of Rheumatology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Bevra H Hahn
- Division of Rheumatology, University of California, Los Angeles, Los Angeles, CA, United States.,Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - David S Bischoff
- Research Service, Veteran Administration Greater Los Angeles Healthcare System, Los Angeles, CA, United States.,Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
6
|
Florijn BW, Bijkerk R, Kruyt ND, van Zonneveld AJ, Wermer MJH. Sex-Specific MicroRNAs in Neurovascular Units in Ischemic Stroke. Int J Mol Sci 2021; 22:11888. [PMID: 34769320 PMCID: PMC8585074 DOI: 10.3390/ijms222111888] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 10/25/2021] [Accepted: 10/27/2021] [Indexed: 12/24/2022] Open
Abstract
Accumulating evidence pinpoints sex differences in stroke incidence, etiology and outcome. Therefore, more understanding of the sex-specific mechanisms that lead to ischemic stroke and aggravation of secondary damage after stroke is needed. Our current mechanistic understanding of cerebral ischemia states that endothelial quiescence in neurovascular units (NVUs) is a major physiological parameter affecting the cellular response to neuron, astrocyte and vascular smooth muscle cell (VSMC) injury. Although a hallmark of the response to injury in these cells is transcriptional activation, noncoding RNAs such as microRNAs exhibit cell-type and context dependent regulation of gene expression at the post-transcriptional level. This review assesses whether sex-specific microRNA expression (either derived from X-chromosome loci following incomplete X-chromosome inactivation or regulated by estrogen in their biogenesis) in these cells controls NVU quiescence, and as such, could differentiate stroke pathophysiology in women compared to men. Their adverse expression was found to decrease tight junction affinity in endothelial cells and activate VSMC proliferation, while their regulation of paracrine astrocyte signaling was shown to neutralize sex-specific apoptotic pathways in neurons. As such, these microRNAs have cell type-specific functions in astrocytes and vascular cells which act on one another, thereby affecting the cell viability of neurons. Furthermore, these microRNAs display actual and potential clinical implications as diagnostic and prognostic biomarkers in ischemic stroke and in predicting therapeutic response to antiplatelet therapy. In conclusion, this review improves the current mechanistic understanding of the molecular mechanisms leading to ischemic stroke in women and highlights the clinical promise of sex-specific microRNAs as novel diagnostic biomarkers for (silent) ischemic stroke.
Collapse
Affiliation(s)
- Barend W. Florijn
- Department of Neurology, Leiden University Medical Center, 2333 ZR Leiden, The Netherlands; (N.D.K.); (M.J.H.W.)
- Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (R.B.); (A.J.v.Z.)
| | - Roel Bijkerk
- Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (R.B.); (A.J.v.Z.)
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Nyika D. Kruyt
- Department of Neurology, Leiden University Medical Center, 2333 ZR Leiden, The Netherlands; (N.D.K.); (M.J.H.W.)
| | - Anton Jan van Zonneveld
- Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (R.B.); (A.J.v.Z.)
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Marieke J. H. Wermer
- Department of Neurology, Leiden University Medical Center, 2333 ZR Leiden, The Netherlands; (N.D.K.); (M.J.H.W.)
| |
Collapse
|
7
|
Neuroepigenetics of psychiatric disorders: Focus on lncRNA. Neurochem Int 2021; 149:105140. [PMID: 34298078 DOI: 10.1016/j.neuint.2021.105140] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 07/16/2021] [Accepted: 07/18/2021] [Indexed: 01/01/2023]
Abstract
Understanding the pathology of psychiatric disorders is challenging due to their complexity and multifactorial origin. However, development of high-throughput technologies has allowed for better insight into their molecular signatures. Advancement of sequencing methodologies have made it possible to study not only the protein-coding but also the noncoding genome. It is now clear that besides the genetic component, different epigenetic mechanisms play major roles in the onset and development of psychiatric disorders. Among them, examining the role of long noncoding RNAs (lncRNAs) is a relatively new field. Here, we present an overview of what is currently known about the involvement of lncRNAs in schizophrenia, major depressive and bipolar disorders, as well as suicide. The diagnosis of psychiatric disorders mainly relies on clinical evaluation without using measurable biomarkers. In this regard, lncRNA may open new opportunities for development of molecular tests. However, so far only a small set of known lncRNAs have been characterized at molecular level, which means they have a long way to go before clinical implementation. Understanding how changes in lncRNAs affect the appearance and development of psychiatric disorders may lead to a more classified and objective diagnostic system, but also open up new therapeutic targets for these patients.
Collapse
|
8
|
Segaran RC, Chan LY, Wang H, Sethi G, Tang FR. Neuronal Development-Related miRNAs as Biomarkers for Alzheimer's Disease, Depression, Schizophrenia and Ionizing Radiation Exposure. Curr Med Chem 2021; 28:19-52. [PMID: 31965936 DOI: 10.2174/0929867327666200121122910] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 09/30/2019] [Accepted: 10/22/2019] [Indexed: 11/22/2022]
Abstract
Radiation exposure may induce Alzheimer's disease (AD), depression or schizophrenia. A number of experimental and clinical studies suggest the involvement of miRNA in the development of these diseases, and also in the neuropathological changes after brain radiation exposure. The current literature review indicated the involvement of 65 miRNAs in neuronal development in the brain. In the brain tissue, blood, or cerebral spinal fluid (CSF), 11, 55, or 28 miRNAs are involved in the development of AD respectively, 89, 50, 19 miRNAs in depression, and 102, 35, 8 miRNAs in schizophrenia. We compared miRNAs regulating neuronal development to those involved in the genesis of AD, depression and schizophrenia and also those driving radiation-induced brain neuropathological changes by reviewing the available data. We found that 3, 11, or 8 neuronal developmentrelated miRNAs from the brain tissue, 13, 16 or 14 miRNAs from the blood of patient with AD, depression and schizophrenia respectively were also involved in radiation-induced brain pathological changes, suggesting a possibly specific involvement of these miRNAs in radiation-induced development of AD, depression and schizophrenia respectively. On the other hand, we noted that radiationinduced changes of two miRNAs, i.e., miR-132, miR-29 in the brain tissue, three miRNAs, i.e., miR- 29c-5p, miR-106b-5p, miR-34a-5p in the blood were also involved in the development of AD, depression and schizophrenia, thereby suggesting that these miRNAs may be involved in the common brain neuropathological changes, such as impairment of neurogenesis and reduced learning memory ability observed in these three diseases and also after radiation exposure.
Collapse
Affiliation(s)
- Renu Chandra Segaran
- Radiation Physiology Lab, Singapore Nuclear Research and Safety Initiative, National University of Singapore, CREATE Tower, Singapore 138602, Singapore
| | - Li Yun Chan
- Radiation Physiology Lab, Singapore Nuclear Research and Safety Initiative, National University of Singapore, CREATE Tower, Singapore 138602, Singapore
| | - Hong Wang
- Radiation Physiology Lab, Singapore Nuclear Research and Safety Initiative, National University of Singapore, CREATE Tower, Singapore 138602, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Feng Ru Tang
- Radiation Physiology Lab, Singapore Nuclear Research and Safety Initiative, National University of Singapore, CREATE Tower, Singapore 138602, Singapore
| |
Collapse
|
9
|
Ermakov EA, Dmitrieva EM, Parshukova DA, Kazantseva DV, Vasilieva AR, Smirnova LP. Oxidative Stress-Related Mechanisms in Schizophrenia Pathogenesis and New Treatment Perspectives. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8881770. [PMID: 33552387 PMCID: PMC7847339 DOI: 10.1155/2021/8881770] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 12/15/2020] [Accepted: 01/02/2021] [Indexed: 02/07/2023]
Abstract
Schizophrenia is recognized to be a highly heterogeneous disease at various levels, from genetics to clinical manifestations and treatment sensitivity. This heterogeneity is also reflected in the variety of oxidative stress-related mechanisms contributing to the phenotypic realization and manifestation of schizophrenia. At the molecular level, these mechanisms are supposed to include genetic causes that increase the susceptibility of individuals to oxidative stress and lead to gene expression dysregulation caused by abnormal regulation of redox-sensitive transcriptional factors, noncoding RNAs, and epigenetic mechanisms favored by environmental insults. These changes form the basis of the prooxidant state and lead to altered redox signaling related to glutathione deficiency and impaired expression and function of redox-sensitive transcriptional factors (Nrf2, NF-κB, FoxO, etc.). At the cellular level, these changes lead to mitochondrial dysfunction and metabolic abnormalities that contribute to aberrant neuronal development, abnormal myelination, neurotransmitter anomalies, and dysfunction of parvalbumin-positive interneurons. Immune dysfunction also contributes to redox imbalance. At the whole-organism level, all these mechanisms ultimately contribute to the manifestation and development of schizophrenia. In this review, we consider oxidative stress-related mechanisms and new treatment perspectives associated with the correction of redox imbalance in schizophrenia. We suggest that not only antioxidants but also redox-regulated transcription factor-targeting drugs (including Nrf2 and FoxO activators or NF-κB inhibitors) have great promise in schizophrenia. But it is necessary to develop the stratification criteria of schizophrenia patients based on oxidative stress-related markers for the administration of redox-correcting treatment.
Collapse
Affiliation(s)
- Evgeny A. Ermakov
- Laboratory of Repair Enzymes, Institute of Chemical Biology and Fundamental Medicine, Siberian Division of Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Elena M. Dmitrieva
- Laboratory of Molecular Genetics and Biochemistry, Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk 634014, Russia
| | - Daria A. Parshukova
- Laboratory of Molecular Genetics and Biochemistry, Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk 634014, Russia
| | | | | | - Liudmila P. Smirnova
- Laboratory of Molecular Genetics and Biochemistry, Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk 634014, Russia
| |
Collapse
|
10
|
Wang W, Jia L. Regulatory Mechanism of MicroRNA-30b on Neonatal Hypoxic-Ischemic Encephalopathy (HIE). J Stroke Cerebrovasc Dis 2020; 30:105553. [PMID: 33360521 DOI: 10.1016/j.jstrokecerebrovasdis.2020.105553] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 12/02/2020] [Accepted: 12/12/2020] [Indexed: 01/24/2023] Open
Abstract
OBJECTIVE This study is to investigate the role of microRNA (miR)-30b in the pathogenesis of hypoxic-ischemic encephalopathy (HIE) in neonates. METHODS Totally 26 cases of neonatal HIE were included in this study. The protein expression levels of CD26P and PAI-1 were detected with ELISA. Serum levels of miR-30b and PAI-1 mRNA was measured by quantitative real-time PCR. Human brain microvascular endothelial cells (HBMECs) were cultured under hypoxic condition, and the intracellular expression levels of miR-30b and PAI-1 were evaluated. Dual-luciferase reporter assay was performed to confirm the interaction between miR-30b and PAI-1. RESULTS Compared with the control group, both the mRNA and protein expression levels of PAI-1 in the serum were up-regulated in the neonates with HIE, together with up-regulated serum CD26P levels. However, the serum expression level of miR-30b was down-regulated in neonatal HIE. In hypoxia-induced HBMECs, the mRNA and protein expression levels of PAI-1 were significantly up-regulated, while the miR-30b expression level was significantly down-regulated. Dual-luciferase reporter assay showed that PAI-1 was the direct target of miR-30b. CONCLUSION Neonatal HIE is accompanied with abnormal platelet activation, significantly up-regulated serum PAI-1 expression levels, and down-regulated miR-30b expression. MiR-30b might regulate the disease pathogenesis and immune responses via modulating PAI-1.
Collapse
Affiliation(s)
- Weiwei Wang
- Department of Pediatrics, Tianjin hospital, Tianjin 300211, China.
| | - Lifang Jia
- Department of Pediatrics, Tianjin hospital, Tianjin hospital of Tianjin University, Tianjin 300211, China.
| |
Collapse
|
11
|
Mahmoudi E, Atkins JR, Quidé Y, Reay WR, Cairns HM, Fitzsimmons C, Carr VJ, Green MJ, Cairns MJ. The MIR137 VNTR rs58335419 Is Associated With Cognitive Impairment in Schizophrenia and Altered Cortical Morphology. Schizophr Bull 2020; 47:495-504. [PMID: 32910167 PMCID: PMC8370045 DOI: 10.1093/schbul/sbaa123] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Genome-wide association studies (GWAS) of schizophrenia have strongly implicated a risk locus in close proximity to the gene for miR-137. While there are candidate single-nucleotide polymorphisms (SNPs) with functional implications for the microRNA's expression encompassed by the common haplotype tagged by rs1625579, there are likely to be others, such as the variable number tandem repeat (VNTR) variant rs58335419, that have no proxy on the SNP genotyping platforms used in GWAS to date. Using whole-genome sequencing data from schizophrenia patients (n = 299) and healthy controls (n = 131), we observed that the MIR137 4-repeats VNTR (VNTR4) variant was enriched in a cognitive deficit subtype of schizophrenia and associated with altered brain morphology, including thicker left inferior temporal gyrus and deeper right postcentral sulcus. These findings suggest that the MIR137 VNTR4 may impact neuroanatomical development that may, in turn, influence the expression of more severe cognitive symptoms in patients with schizophrenia.
Collapse
Affiliation(s)
- Ebrahim Mahmoudi
- School of Biomedical Sciences and Pharmacy, University of
Newcastle, Callaghan, New South Wales, Australia,Centre for Brain and Mental Health Research, University of
Newcastle, Callaghan, New South Wales, Australia,Hunter Medical Research Institute, New South Wales, New Lambton,
Australia
| | - Joshua R Atkins
- School of Biomedical Sciences and Pharmacy, University of
Newcastle, Callaghan, New South Wales, Australia,Centre for Brain and Mental Health Research, University of
Newcastle, Callaghan, New South Wales, Australia,Hunter Medical Research Institute, New South Wales, New Lambton,
Australia
| | - Yann Quidé
- School of Psychiatry, University of New South Wales, Sydney, New
South Wales, Australia,Neuroscience Research Australia, Randwick, New South Wales,
Australia
| | - William R Reay
- School of Biomedical Sciences and Pharmacy, University of
Newcastle, Callaghan, New South Wales, Australia,Centre for Brain and Mental Health Research, University of
Newcastle, Callaghan, New South Wales, Australia,Hunter Medical Research Institute, New South Wales, New Lambton,
Australia
| | - Heath M Cairns
- School of Biomedical Sciences and Pharmacy, University of
Newcastle, Callaghan, New South Wales, Australia,Centre for Brain and Mental Health Research, University of
Newcastle, Callaghan, New South Wales, Australia,Hunter Medical Research Institute, New South Wales, New Lambton,
Australia
| | - Chantel Fitzsimmons
- School of Biomedical Sciences and Pharmacy, University of
Newcastle, Callaghan, New South Wales, Australia,Centre for Brain and Mental Health Research, University of
Newcastle, Callaghan, New South Wales, Australia,Hunter Medical Research Institute, New South Wales, New Lambton,
Australia
| | - Vaughan J Carr
- School of Psychiatry, University of New South Wales, Sydney, New
South Wales, Australia,Neuroscience Research Australia, Randwick, New South Wales,
Australia,Department of Psychiatry, School of Clinical Sciences, Monash
University, Clayton, Victoria, Australia
| | - Melissa J Green
- School of Psychiatry, University of New South Wales, Sydney, New
South Wales, Australia,Neuroscience Research Australia, Randwick, New South Wales,
Australia
| | - Murray J Cairns
- School of Biomedical Sciences and Pharmacy, University of
Newcastle, Callaghan, New South Wales, Australia,Centre for Brain and Mental Health Research, University of
Newcastle, Callaghan, New South Wales, Australia,Hunter Medical Research Institute, New South Wales, New Lambton,
Australia,To whom correspondence should be addressed; tel: +61 (02) 4921 8670, fax:
+61 (02) 4921 7903, e-mail:
| |
Collapse
|
12
|
Epigenomic Dysregulation in Schizophrenia: In Search of Disease Etiology and Biomarkers. Cells 2020; 9:cells9081837. [PMID: 32764320 PMCID: PMC7463953 DOI: 10.3390/cells9081837] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/27/2020] [Accepted: 07/31/2020] [Indexed: 12/13/2022] Open
Abstract
Schizophrenia is a severe psychiatric disorder with a complex array of signs and symptoms that causes very significant disability in young people. While schizophrenia has a strong genetic component, with heritability around 80%, there is also a very significant range of environmental exposures and stressors that have been implicated in disease development and neuropathology, such as maternal immune infection, obstetric complications, childhood trauma and cannabis exposure. It is postulated that epigenetic factors, as well as regulatory non-coding RNAs, mediate the effects of these environmental stressors. In this review, we explore the most well-known epigenetic marks, including DNA methylation and histone modification, along with emerging RNA mediators of epigenomic state, including miRNAs and lncRNAs, and discuss their collective potential for involvement in the pathophysiology of schizophrenia implicated through the postmortem analysis of brain tissue. Given that peripheral tissues, such as blood, saliva, and olfactory epithelium have the same genetic composition and are exposed to many of the same environmental exposures, we also examine some studies supporting the application of peripheral tissues for epigenomic biomarker discovery in schizophrenia. Finally, we provide some perspective on how these biomarkers may be utilized to capture a signature of past events that informs future treatment.
Collapse
|
13
|
Smigielski L, Jagannath V, Rössler W, Walitza S, Grünblatt E. Epigenetic mechanisms in schizophrenia and other psychotic disorders: a systematic review of empirical human findings. Mol Psychiatry 2020; 25:1718-1748. [PMID: 31907379 DOI: 10.1038/s41380-019-0601-3] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 11/05/2019] [Accepted: 11/07/2019] [Indexed: 12/26/2022]
Abstract
Schizophrenia and other psychotic disorders are highly debilitating psychiatric conditions that lack a clear etiology and exhibit polygenic inheritance underlain by pleiotropic genes. The prevailing explanation points to the interplay between predisposing genes and environmental exposure. Accumulated evidence suggests that epigenetic regulation of the genome may mediate dynamic gene-environment interactions at the molecular level by modulating the expression of psychiatric phenotypes through transcription factors. This systematic review summarizes the current knowledge linking schizophrenia and other psychotic disorders to epigenetics, based on PubMed and Web of Science database searches conducted according to the PRISMA guidelines. Three groups of mechanisms in case-control studies of human tissue (i.e., postmortem brain and bio-fluids) were considered: DNA methylation, histone modifications, and non-coding miRNAs. From the initial pool of 3,204 records, 152 studies met our inclusion criteria (11,815/11,528, 233/219, and 2,091/1,827 cases/controls for each group, respectively). Many of the findings revealed associations with epigenetic modulations of genes regulating neurotransmission, neurodevelopment, and immune function, as well as differential miRNA expression (e.g., upregulated miR-34a, miR-7, and miR-181b). Overall, actual evidence moderately supports an association between epigenetics and schizophrenia and other psychotic disorders. However, heterogeneous results and cross-tissue extrapolations call for future work. Integrating epigenetics into systems biology may critically enhance research on psychosis and thus our understanding of the disorder. This may have implications for psychiatry in risk stratification, early recognition, diagnostics, precision medicine, and other interventional approaches targeting epigenetic fingerprints.
Collapse
Affiliation(s)
- Lukasz Smigielski
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital of Psychiatry Zurich, University of Zurich, Zurich, Switzerland. .,The Zurich Program for Sustainable Development of Mental Health Services (ZInEP), University Hospital of Psychiatry Zurich, Zurich, Switzerland.
| | - Vinita Jagannath
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital of Psychiatry Zurich, University of Zurich, Zurich, Switzerland.,Merck Sharp & Dohme (MSD) R&D Innovation Centre, London, UK
| | - Wulf Rössler
- The Zurich Program for Sustainable Development of Mental Health Services (ZInEP), University Hospital of Psychiatry Zurich, Zurich, Switzerland.,Department of Psychiatry, Psychotherapy and Psychosomatics, University Hospital of Psychiatry, University of Zurich, Zurich, Switzerland.,Department of Psychiatry and Psychotherapy, Charité Universitätsmedizin, Berlin, Germany.,Laboratory of Neuroscience, Institute of Psychiatry, Universidade de São Paulo, São Paulo, Brazil
| | - Susanne Walitza
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital of Psychiatry Zurich, University of Zurich, Zurich, Switzerland.,The Zurich Program for Sustainable Development of Mental Health Services (ZInEP), University Hospital of Psychiatry Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Edna Grünblatt
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital of Psychiatry Zurich, University of Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
14
|
Catechol-O-methyltransferase gene promoter methylation as a peripheral biomarker in male schizophrenia. Eur Psychiatry 2020; 44:39-46. [DOI: 10.1016/j.eurpsy.2017.03.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Revised: 03/06/2017] [Accepted: 03/06/2017] [Indexed: 02/03/2023] Open
Abstract
AbstractAs an epigenetic modification, DNA methylation may reflect the interaction between genetic and environmental factors in the development of schizophrenia (SCZ). Catechol-O-methyltransferase (COMT) gene is a promising candidate gene of SCZ. In the present study, we investigate the association of COMT methylation with the risk of SCZ using bisulfite pyrosequencing technology. Significant association between DNA methylation of COMT and the risk of SCZ is identified (P = 1.618e−007). A breakdown analysis by gender shows that the significance is driven by males (P = 3.310e−009), but not by females. DNA methylation of COMT is not significantly associated with SCZ clinical phenotypes, including p300 and cysteine level. No interaction is found between COMT genotypes and the percent methylation of this gene. Receiver operating characteristic (ROC) curve shows that DNA methylation of COMT is able to predict the SCZ risk in males (area under curve [AUC] = 0.802, P = 1.91e−007). The current study indicates the clinical value of COMT methylation as a potential male-specific biomarker in SCZ diagnosis.
Collapse
|
15
|
Amoah SK, Rodriguez BA, Logothetis CN, Chander P, Sellgren CM, Weick JP, Sheridan SD, Jantzie LL, Webster MJ, Mellios N. Exosomal secretion of a psychosis-altered miRNA that regulates glutamate receptor expression is affected by antipsychotics. Neuropsychopharmacology 2020; 45:656-665. [PMID: 31775160 PMCID: PMC7021900 DOI: 10.1038/s41386-019-0579-1] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 10/24/2019] [Accepted: 11/19/2019] [Indexed: 12/19/2022]
Abstract
The ability of small secretory microvesicles known as exosomes to influence neuronal and glial function via their microRNA (miRNA) cargo has positioned them as a novel and effective method of cell-to-cell communication. However, little is known about the role of exosome-secreted miRNAs in the regulation of glutamate receptor gene expression and their relevance for schizophrenia (SCZ) and bipolar disorder (BD). Using mature miRNA profiling and quantitative real-time PCR (qRT-PCR) in the orbitofrontal cortex (OFC) of SCZ (N = 29; 20 male and 9 female), BD (N = 26; 12 male and 14 female), and unaffected control (N = 25; 21 male and 4 female) subjects, we uncovered that miR-223, an exosome-secreted miRNA that targets glutamate receptors, was increased at the mature miRNA level in the OFC of SCZ and BD patients with positive history of psychosis at the time of death and was inversely associated with deficits in the expression of its targets glutamate ionotropic receptor NMDA-type subunit 2B (GRIN2B) and glutamate ionotropic receptor AMPA-type subunit 2 (GRIA2). Furthermore, changes in miR-223 levels in the OFC were positively and negatively correlated with inflammatory and GABAergic gene expression, respectively. Moreover, miR-223 was found to be enriched in astrocytes and secreted via exosomes, and antipsychotics were shown to control its cellular and exosomal localization in a cell-specific manner. Furthermore, addition of astrocytic exosomes in neuronal cultures resulted in a significant increase in miR-223 expression and a notable reduction in Grin2b and Gria2 mRNA levels, which was strongly inversely associated with miR-223 expression. Lastly, inhibition of astrocytic miR-223 abrogated the exosomal-mediated reduction in neuronal Grin2b expression. Taken together, our results demonstrate that the exosomal secretion of a psychosis-altered and glial-enriched miRNA that controls neuronal gene expression is regulated by antipsychotics.
Collapse
Affiliation(s)
- Stephen K Amoah
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, USA
- Autophagy inflammation and metabolism (AIM) center, Albuquerque, NM, USA
| | - Brian A Rodriguez
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | | | - Praveen Chander
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Carl M Sellgren
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Jason P Weick
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Steven D Sheridan
- Center for Genomic Medicine, Chemical Neurobiology Laboratory, Departments of Neurology and Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Center for Experimental Drugs and Diagnostics, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Lauren L Jantzie
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Maree J Webster
- Laboratory of Brain Research, Stanley Medical Research Institute, Chevy Chase, MD, USA
| | - Nikolaos Mellios
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, USA.
- Autophagy inflammation and metabolism (AIM) center, Albuquerque, NM, USA.
| |
Collapse
|
16
|
Abstract
PURPOSE OF REVIEW To better understand the shared basis of language and mental health, this review examines the behavioral and neurobiological features of aberrant language in five major neuropsychiatric conditions. Special attention is paid to genes implicated in both language and neuropsychiatric disorders, as they reveal biological domains likely to underpin the processes controlling both. RECENT FINDINGS Abnormal language and communication are common manifestations of neuropsychiatric conditions, and children with impaired language are more likely to develop psychiatric disorders than their peers. Major themes in the genetics of both language and psychiatry include master transcriptional regulators, like FOXP2; key developmental regulators, like AUTS2; and mediators of neurotransmission, like GRIN2A and CACNA1C.
Collapse
|
17
|
MicroRNAs and Child Neuropsychiatric Disorders: A Brief Review. Neurochem Res 2019; 45:232-240. [DOI: 10.1007/s11064-019-02917-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 10/23/2019] [Accepted: 11/21/2019] [Indexed: 12/12/2022]
|
18
|
Zhao Z, Jinde S, Koike S, Tada M, Satomura Y, Yoshikawa A, Nishimura Y, Takizawa R, Kinoshita A, Sakakibara E, Sakurada H, Yamagishi M, Nishimura F, Inai A, Nishioka M, Eriguchi Y, Araki T, Takaya A, Kan C, Umeda M, Shimazu A, Hashimoto H, Bundo M, Iwamoto K, Kakiuchi C, Kasai K. Altered expression of microRNA-223 in the plasma of patients with first-episode schizophrenia and its possible relation to neuronal migration-related genes. Transl Psychiatry 2019; 9:289. [PMID: 31712567 PMCID: PMC6848172 DOI: 10.1038/s41398-019-0609-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 09/10/2019] [Accepted: 09/30/2019] [Indexed: 12/26/2022] Open
Abstract
Recent studies have shown that microRNAs (miRNAs) play a role as regulators of neurodevelopment by modulating gene expression. Altered miRNA expression has been reported in various psychiatric disorders, including schizophrenia. However, the changes in the miRNA expression profile that occur during the initial stage of schizophrenia have not been fully investigated. To explore the global alterations in miRNA expression profiles that may be associated with the onset of schizophrenia, we first profiled miRNA expression in plasma from 17 patients with first-episode schizophrenia and 17 healthy controls using microarray analysis. Among the miRNAs that showed robust changes, the elevated expression of has-miR-223-3p (miR-223) was validated via quantitative reverse transcription-polymerase chain reaction (qRT-PCR) using another independent sample set of 21 schizophrenia patients and 21 controls. To identify the putative targets of miR-223, we conducted a genome-wide gene expression analysis in neuronally differentiated SK-N-SH cells with stable miR-223 overexpression and an in silico analysis. We found that the mRNA expression levels of four genes related to the cytoskeleton or cell migration were significantly downregulated in miR-223-overexpressing cells, possibly due to interactions with miR-223. The in silico analysis suggested the presence of miR-223 target sites in these four genes. Lastly, a luciferase assay confirmed that miR-223 directly interacted with the 3' untranslated regions (UTRs) of all four genes. Our results reveal an increase in miR-223 in plasma during both the first episode and the later stage of schizophrenia, which may affect the expression of cell migration-related genes targeted by miR-223.
Collapse
Affiliation(s)
- Zhilei Zhao
- 0000 0001 2151 536Xgrid.26999.3dDepartment of Neuropsychiatry, Graduate School of Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, 113-8655 Japan ,0000 0001 2151 536Xgrid.26999.3dInternational Research Center for Neurointelligence, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033 Japan
| | - Seiichiro Jinde
- Department of Neuropsychiatry, Graduate School of Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Shinsuke Koike
- 0000 0001 2151 536Xgrid.26999.3dDepartment of Neuropsychiatry, Graduate School of Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, 113-8655 Japan
| | - Mariko Tada
- 0000 0001 2151 536Xgrid.26999.3dDepartment of Neuropsychiatry, Graduate School of Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, 113-8655 Japan
| | - Yoshihiro Satomura
- 0000 0001 2151 536Xgrid.26999.3dDepartment of Neuropsychiatry, Graduate School of Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, 113-8655 Japan
| | - Akane Yoshikawa
- 0000 0001 2151 536Xgrid.26999.3dDepartment of Neuropsychiatry, Graduate School of Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, 113-8655 Japan
| | - Yukika Nishimura
- 0000 0001 2151 536Xgrid.26999.3dDepartment of Neuropsychiatry, Graduate School of Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, 113-8655 Japan
| | - Ryu Takizawa
- 0000 0001 2151 536Xgrid.26999.3dDepartment of Neuropsychiatry, Graduate School of Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, 113-8655 Japan
| | - Akihide Kinoshita
- 0000 0001 2151 536Xgrid.26999.3dDepartment of Neuropsychiatry, Graduate School of Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, 113-8655 Japan
| | - Eisuke Sakakibara
- 0000 0001 2151 536Xgrid.26999.3dDepartment of Neuropsychiatry, Graduate School of Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, 113-8655 Japan
| | - Hanako Sakurada
- 0000 0001 2151 536Xgrid.26999.3dDepartment of Neuropsychiatry, Graduate School of Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, 113-8655 Japan
| | - Mika Yamagishi
- 0000 0001 2151 536Xgrid.26999.3dDepartment of Neuropsychiatry, Graduate School of Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, 113-8655 Japan
| | - Fumichika Nishimura
- 0000 0001 2151 536Xgrid.26999.3dDepartment of Neuropsychiatry, Graduate School of Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, 113-8655 Japan
| | - Aya Inai
- 0000 0001 2151 536Xgrid.26999.3dDepartment of Child Neuropsychiatry, Graduate School of Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, 113-8655 Japan
| | - Masaki Nishioka
- 0000 0001 2151 536Xgrid.26999.3dDepartment of Neuropsychiatry, Graduate School of Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, 113-8655 Japan
| | - Yosuke Eriguchi
- 0000 0001 2151 536Xgrid.26999.3dDepartment of Child Neuropsychiatry, Graduate School of Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, 113-8655 Japan
| | - Tsuyoshi Araki
- 0000 0001 2151 536Xgrid.26999.3dDepartment of Neuropsychiatry, Graduate School of Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, 113-8655 Japan
| | - Atsuhiko Takaya
- Department of Psychiatry, Fukui Kinen Hospital, Miura City, Kanagawa 238-0115 Japan
| | - Chiemi Kan
- 0000 0001 2151 536Xgrid.26999.3dDepartment of Mental Health, Graduate School of Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, 113-0033 Japan
| | - Maki Umeda
- 0000 0001 2151 536Xgrid.26999.3dDepartment of Mental Health, Graduate School of Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, 113-0033 Japan ,0000 0001 0318 6320grid.419588.9Department of Public Health Nursing, Graduate School of Nursing Science, St. Luke’s International University, Chuo-ku, Tokyo, 104-0044 Japan
| | - Akihito Shimazu
- 0000 0000 9206 2938grid.410786.cCenter for Human and Social Sciences, College of Liberal Arts and Sciences, Kitasato University, Sagamihara City, Kanagawa 252-0373 Japan
| | - Hideki Hashimoto
- 0000 0001 2151 536Xgrid.26999.3dDepartment of Health Economics and Epidemiology Research, School of Public Health, the University of Tokyo, Bunkyo-ku, Tokyo, 113-0033 Japan
| | - Miki Bundo
- 0000 0001 0660 6749grid.274841.cDepartment of Molecular Brain Science, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto City, Kumamoto, 860-8556 Japan
| | - Kazuya Iwamoto
- 0000 0001 0660 6749grid.274841.cDepartment of Molecular Brain Science, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto City, Kumamoto, 860-8556 Japan
| | - Chihiro Kakiuchi
- 0000 0001 2151 536Xgrid.26999.3dDepartment of Neuropsychiatry, Graduate School of Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, 113-8655 Japan
| | - Kiyoto Kasai
- 0000 0001 2151 536Xgrid.26999.3dDepartment of Neuropsychiatry, Graduate School of Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, 113-8655 Japan ,0000 0001 2151 536Xgrid.26999.3dInternational Research Center for Neurointelligence, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033 Japan
| |
Collapse
|
19
|
Piscopo P, Grasso M, Puopolo M, D'Acunto E, Talarico G, Crestini A, Gasparini M, Campopiano R, Gambardella S, Castellano AE, Bruno G, Denti MA, Confaloni A. Circulating miR-127-3p as a Potential Biomarker for Differential Diagnosis in Frontotemporal Dementia. J Alzheimers Dis 2019; 65:455-464. [PMID: 30056425 DOI: 10.3233/jad-180364] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Given the heterogeneous nature of frontotemporal dementia (FTD), sensitive biomarkers are greatly needed for the accurate diagnosis of this neurodegenerative disorder. Circulating miRNAs have been reported as promising biomarkers for neurodegenerative disorders and processes affecting the central nervous system, especially in aging. The objective of the study was to evaluate if some circulating miRNAs linked with apoptosis (miR-29b-3p, miR-34a-5p, miR-16-5p, miR-17-5p, miR-107, miR-19b-3p, let-7b-5p, miR-26b-5p, and 127-3p) were able to distinguish between FTD patients and healthy controls. For this study, we enrolled 127 subjects, including 54 patients with FTD, 20 patients with Alzheimer's disease (AD), and 53 healthy controls. The qRT-PCR analysis showed a downregulation of miR-127-3p in FTD compared to controls, while the levels of other miRNAs remained unchanged. Then, miR-127-3p expression was also analyzed in AD patients, finding a different expression between two patient groups. A receiver operating characteristic curve was then created for miR-127-3p to discriminate FTD versus AD (AUC: 0.8986), and versus healthy controls (AUC: 0.8057). In conclusion, miR-127-3p could help to diagnose FTD and to distinguish it from AD.
Collapse
Affiliation(s)
- Paola Piscopo
- Department of Neuroscience, Istituto Superiore di Sanità, Rome, Italy
| | | | - Maria Puopolo
- Department of Neuroscience, Istituto Superiore di Sanità, Rome, Italy
| | - Emanuela D'Acunto
- Department of Neuroscience, Istituto Superiore di Sanità, Rome, Italy.,Department of Biology and Biotechnologies 'Charles Darwin', University of Rome "Sapienza", Rome, Italy
| | - Giuseppina Talarico
- Department of Human Neuroscience, University of Rome "Sapienza", Rome, Italy
| | - Alessio Crestini
- Department of Neuroscience, Istituto Superiore di Sanità, Rome, Italy
| | - Marina Gasparini
- Department of Human Neuroscience, University of Rome "Sapienza", Rome, Italy
| | - Rosa Campopiano
- Department of Neurology, IRCCS Neuromed Institute, Pozzilli, IS, Italy
| | | | | | - Giuseppe Bruno
- Department of Human Neuroscience, University of Rome "Sapienza", Rome, Italy
| | - Michela A Denti
- Centre for Integrative Biology, University of Trento, Trento, Italy
| | | |
Collapse
|
20
|
He K, Guo C, Guo M, Tong S, Zhang Q, Sun H, He L, Shi Y. Identification of serum microRNAs as diagnostic biomarkers for schizophrenia. Hereditas 2019; 156:23. [PMID: 31297041 PMCID: PMC6598381 DOI: 10.1186/s41065-019-0099-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 06/20/2019] [Indexed: 12/18/2022] Open
Abstract
Background At present, the schizophrenia diagnoses are based on the clinical symptoms and behaviors neglecting the laboratory test indicators. Results To better investigate the diagnostic potential of miRNAs for schizophrenia, we selected 14 candidate miRNAs and examined their expressions in the serums of 40 schizophrenia patients and 40 healthy controls by qRT-PCR. Ultimately three abnormally expressed microRNAs were identified, i.e., miR-34a-5p, miR-432-5p and miR-449a. Then, binary regression analysis was employed to combine 3 dysregulated miRNAs. ROC analysis revealed that the AUC of the combination of miR-432-5p + miR-449a in serums was 0.841 (95% CI: 0.791~0.887) with 90% sensitivity and 80% specificity. The AUC of the combination of miR-34a-5p + miR-432-5p + miR-449a in serums was 0.843 (95% CI: 0.791~0.887) with 90% sensitivity and 77.5% specificity. The results indicated that the combined model of miR-432-5p + miR-449a and miR-34a-5p + miR-432-5p + miR-449a have better prediction performances. Conclusions The study concludes that the two miRNAs combinations have the potential to be used as biomarkers for schizophrenia diagnoses. The finding may be conducive to overcoming the dilemmas faced by current schizophrenia diagnosis.
Collapse
Affiliation(s)
- Kuanjun He
- 1College of Life Science, Inner Mongolia University for Nationalities, Tongliao, 028043 People's Republic of China
| | - Chuang Guo
- 1College of Life Science, Inner Mongolia University for Nationalities, Tongliao, 028043 People's Republic of China
| | - Meng Guo
- 2Network Center, Inner Mongolia University for Nationalities, Tongliao, 028043 People's Republic of China
| | - Shuping Tong
- 3Affiliated Hospital of Inner Mongolia University for Nationalities, Tongliao, 028043 People's Republic of China
| | - Qiuli Zhang
- 3Affiliated Hospital of Inner Mongolia University for Nationalities, Tongliao, 028043 People's Republic of China
| | - Hongjun Sun
- Tongliao Institute of Mental Health, Tongliao, 028043 People's Republic of China
| | - Lin He
- 5Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, 200030 People's Republic of China.,6Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030 People's Republic of China.,7Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, Shanghai, 200042 People's Republic of China
| | - Yongyong Shi
- 5Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, 200030 People's Republic of China.,6Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030 People's Republic of China.,7Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, Shanghai, 200042 People's Republic of China
| |
Collapse
|
21
|
Shen JJ, Wang YF, Yang W. Sex-Interacting mRNA- and miRNA-eQTLs and Their Implications in Gene Expression Regulation and Disease. Front Genet 2019; 10:313. [PMID: 31024623 PMCID: PMC6465513 DOI: 10.3389/fgene.2019.00313] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 03/21/2019] [Indexed: 12/26/2022] Open
Abstract
Despite sex being an important epidemiological and physiological factor, not much is known about how sex works to interact with genotypes to result in different phenotypes. Both messenger RNA (mRNA) and microRNA (miRNA) may be differentially expressed between the sexes in different physiological conditions, and both may be differentially regulated between males and females. Using whole transcriptome data on lymphoblastoid cell lines from 338 samples of European origin, we tried to uncover genes differentially expressed between the two sexes and sex-interacting expression quantitative trait loci (ss-eQTLs). Two miRNAs were found to be differentially expressed between the two sexes, both of which were found to be functionally implicated in breast cancer. Using two stage linear regression analysis, 21 mRNA ss-eQTL and 3 miRNA ss-eQTLs were discovered. We replicated two of the mRNA ss-eQTLs (p < 0.1) using a separate dataset of gene expression data derived from monocytes. Three mRNA ss-eQTLs are in high linkage disequilibrium with variants also found to be associated with sexually dimorphic traits. Taken together, we believe the ss-eQTLs presented will assist researchers in uncovering the basis of sex-biased gene expression regulation, and ultimately help us understand the genetic basis of differences in phenotypes between sexes.
Collapse
Affiliation(s)
- Jiangshan J Shen
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong.,Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Jining, China.,Lupus Research Institute, Affiliated Hospital of Jining Medical University, Jining, China
| | - Yong-Fei Wang
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Wanling Yang
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| |
Collapse
|
22
|
Migdalska-Richards A, Mill J. Epigenetic studies of schizophrenia: current status and future directions. Curr Opin Behav Sci 2019. [DOI: 10.1016/j.cobeha.2018.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
23
|
Zhu L, Li C, Liu Q, Xu W, Zhou X. Molecular biomarkers in cardiac hypertrophy. J Cell Mol Med 2019; 23:1671-1677. [PMID: 30648807 PMCID: PMC6378174 DOI: 10.1111/jcmm.14129] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 11/30/2018] [Accepted: 12/10/2018] [Indexed: 12/21/2022] Open
Abstract
Cardiac hypertrophy is characterized by an increase in myocyte size in the absence of cell division. This condition is thought to be an adaptive response to cardiac wall stress resulting from the enhanced cardiac afterload. The pathogenesis of heart dysfunction, which is one of the primary causes of morbidity and mortality in elderly people, is often associated with myocardial remodelling caused by cardiac hypertrophy. In order to well understand the potential mechanisms, we described the molecules involved in the development and progression of myocardial hypertrophy. Increasing evidence has indicated that micro‐RNAs are involved in the pathogenesis of cardiac hypertrophy. In addition, molecular biomarkers including vascular endothelial growth factor B, NAD‐dependent deacetylase sirtuin‐3, growth/differentiation factor 15 and glycoprotein 130, also play important roles in the development of myocardial hypertrophy. Knowing the regulatory mechanisms of these biomarkers in the heart may help identify new molecular targets for the treatment of cardiac hypertrophy.
Collapse
Affiliation(s)
- Liu Zhu
- Department of Cardiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Chao Li
- Department of Cardiology, Fuwai Hospital, Chinese Academy of Medical Sciences, Shenzhen, China
| | - Qiang Liu
- Department of Cardiology, Fuwai Hospital, Chinese Academy of Medical Sciences, Shenzhen, China
| | - Weiting Xu
- Department of Cardiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiang Zhou
- Department of Cardiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
24
|
Role of miRNA in the Regulatory Mechanisms of Estrogens in Cardiovascular Ageing. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:6082387. [PMID: 30671171 PMCID: PMC6317101 DOI: 10.1155/2018/6082387] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 11/13/2018] [Indexed: 12/24/2022]
Abstract
Cardiovascular diseases are a worldwide health problem and are the leading cause of mortality in developed countries. Together with experimental data, the lower incidence of cardiovascular diseases in women than in men of reproductive age points to the influence of sex hormones at the cardiovascular level and suggests that estrogens play a protective role against cardiovascular disease and that this role is also modified by ageing. Estrogens affect cardiovascular function via their specific estrogen receptors to trigger gene expression changes at the transcriptional level. In addition, emerging studies have proposed a role for microRNAs in the vascular effects mediated by estrogens. miRNAs regulate gene expression by repressing translational processes and have been estimated to be involved in the regulation of approximately 30% of all protein-coding genes in mammals. In this review, we highlight the current knowledge of the role of estrogen-sensitive miRNAs, and their influence in regulating vascular ageing.
Collapse
|
25
|
Hepatitis B virus promotes proliferation and metastasis in male Chinese hepatocellular carcinoma patients through the LEF-1/miR-371a-5p/SRCIN1/pleiotrophin/Slug pathway. Exp Cell Res 2018; 370:174-188. [DOI: 10.1016/j.yexcr.2018.06.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 06/14/2018] [Accepted: 06/16/2018] [Indexed: 12/13/2022]
|
26
|
Giatti S, Garcia-Segura LM, Barreto GE, Melcangi RC. Neuroactive steroids, neurosteroidogenesis and sex. Prog Neurobiol 2018; 176:1-17. [PMID: 29981391 DOI: 10.1016/j.pneurobio.2018.06.007] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 05/25/2018] [Accepted: 06/30/2018] [Indexed: 12/12/2022]
Abstract
The nervous system is a target and a source of steroids. Neuroactive steroids are steroids that target neurons and glial cells. They include hormonal steroids originated in the peripheral glands, steroids locally synthesized by the neurons and glial cells (neurosteroids) and synthetic steroids, some of them used in clinical practice. Here we review the mechanisms of synthesis, metabolism and action of neuroactive steroids, including the role of epigenetic modifications and the mitochondria in their sex specific actions. We examine sex differences in neuroactive steroid levels under physiological conditions and their role in the establishment of sex dimorphic structures in the nervous system and sex differences in its function. In addition, particular attention is paid to neuroactive steroids under pathological conditions, analyzing how pathology alters their levels and their role as neuroprotective factors, considering the influence of sex in both cases.
Collapse
Affiliation(s)
- Silvia Giatti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Luis M Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| | - Roberto C Melcangi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy.
| |
Collapse
|
27
|
Denley MCS, Gatford NJF, Sellers KJ, Srivastava DP. Estradiol and the Development of the Cerebral Cortex: An Unexpected Role? Front Neurosci 2018; 12:245. [PMID: 29887794 PMCID: PMC5981095 DOI: 10.3389/fnins.2018.00245] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Accepted: 03/28/2018] [Indexed: 12/16/2022] Open
Abstract
The cerebral cortex undergoes rapid folding in an "inside-outside" manner during embryonic development resulting in the establishment of six discrete cortical layers. This unique cytoarchitecture occurs via the coordinated processes of neurogenesis and cell migration. In addition, these processes are fine-tuned by a number of extracellular cues, which exert their effects by regulating intracellular signaling pathways. Interestingly, multiple brain regions have been shown to develop in a sexually dimorphic manner. In many cases, estrogens have been demonstrated to play an integral role in mediating these sexual dimorphisms in both males and females. Indeed, 17β-estradiol, the main biologically active estrogen, plays a critical organizational role during early brain development and has been shown to be pivotal in the sexually dimorphic development and regulation of the neural circuitry underlying sex-typical and socio-aggressive behaviors in males and females. However, whether and how estrogens, and 17β-estradiol in particular, regulate the development of the cerebral cortex is less well understood. In this review, we outline the evidence that estrogens are not only present but are engaged and regulate molecular machinery required for the fine-tuning of processes central to the cortex. We discuss how estrogens are thought to regulate the function of key molecular players and signaling pathways involved in corticogenesis, and where possible, highlight if these processes are sexually dimorphic. Collectively, we hope this review highlights the need to consider how estrogens may influence the development of brain regions directly involved in the sex-typical and socio-aggressive behaviors as well as development of sexually dimorphic regions such as the cerebral cortex.
Collapse
Affiliation(s)
- Matthew C. S. Denley
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom
| | - Nicholas J. F. Gatford
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom
| | - Katherine J. Sellers
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom
| | - Deepak P. Srivastava
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, United Kingdom
| |
Collapse
|
28
|
The Involvement of MicroRNAs in Modulation of Innate and Adaptive Immunity in Systemic Lupus Erythematosus and Lupus Nephritis. J Immunol Res 2018; 2018:4126106. [PMID: 29854836 PMCID: PMC5964414 DOI: 10.1155/2018/4126106] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 04/03/2018] [Indexed: 12/20/2022] Open
Abstract
Noncoding RNAs (ncRNAs), including microRNAs (miRNAs), represent a family of RNA molecules that do not translate into protein. Nevertheless, they have the ability to regulate gene expression and play an essential role in immune cell differentiation and function. MicroRNAs were found to be differentially expressed in various tissues, and changes in their expression have been associated with several pathological processes. Yet, their roles in systemic lupus erythematosus (SLE) and lupus nephritis (LN) remain to be elucidated. Both SLE and LN are characterized by a complex dysfunction of the innate and adaptive immunity. Recently, significant findings have been made in understanding SLE through the use of genetic variant identification and expression pattern analysis and mouse models, as well as epigenetic analyses. Abnormalities in immune cell responses, cytokine and chemokine production, cell activation, and apoptosis have been linked to a unique expression pattern of a number of miRNAs that have been implicated in the immune pathogenesis of this autoimmune disease. The recent evidence that significantly increased the understanding of the pathogenesis of SLE drives a renewed interest in efficient therapy targets. This review aims at providing an overview of the current state of research on the expression and role of miRNAs in the immune pathogenesis of SLE and LN.
Collapse
|
29
|
Gibbons A, Udawela M, Dean B. Non-Coding RNA as Novel Players in the Pathophysiology of Schizophrenia. Noncoding RNA 2018; 4:E11. [PMID: 29657307 PMCID: PMC6027250 DOI: 10.3390/ncrna4020011] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 03/29/2018] [Accepted: 04/06/2018] [Indexed: 12/22/2022] Open
Abstract
Schizophrenia is associated with diverse changes in the brain's transcriptome and proteome. Underlying these changes is the complex dysregulation of gene expression and protein production that varies both spatially across brain regions and temporally with the progression of the illness. The growing body of literature showing changes in non-coding RNA in individuals with schizophrenia offers new insights into the mechanisms causing this dysregulation. A large number of studies have reported that the expression of microRNA (miRNA) is altered in the brains of individuals with schizophrenia. This evidence is complemented by findings that single nucleotide polymorphisms (SNPs) in miRNA host gene sequences can confer an increased risk of developing the disorder. Additionally, recent evidence suggests the expression of other non-coding RNAs, such as small nucleolar RNA and long non-coding RNA, may also be affected in schizophrenia. Understanding how these changes in non-coding RNAs contribute to the development and progression of schizophrenia offers potential avenues for the better treatment and diagnosis of the disorder. This review will focus on the evidence supporting the involvement of non-coding RNA in schizophrenia and its therapeutic potential.
Collapse
Affiliation(s)
- Andrew Gibbons
- The Florey Institute for Neuroscience and Mental Health, 30 Royal Parade, Parkville, VIC 3052, Australia.
- The Department of Psychiatry, the University of Melbourne, Parkville, Victoria, Australia.
| | - Madhara Udawela
- The Florey Institute for Neuroscience and Mental Health, 30 Royal Parade, Parkville, VIC 3052, Australia.
| | - Brian Dean
- The Florey Institute for Neuroscience and Mental Health, 30 Royal Parade, Parkville, VIC 3052, Australia.
- The Centre for Mental Health, Swinburne University of Technology, Hawthorn, Victoria, Australia.
| |
Collapse
|
30
|
Diagnostic value of blood-derived microRNAs for schizophrenia: results of a meta-analysis and validation. Sci Rep 2017; 7:15328. [PMID: 29127368 PMCID: PMC5681644 DOI: 10.1038/s41598-017-15751-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 11/01/2017] [Indexed: 12/31/2022] Open
Abstract
There is an increasing interest in searching biomarkers for schizophrenia (SZ) diagnosis, which overcomes the drawbacks inherent with the subjective diagnostic methods. MicroRNA (miRNA) fingerprints have been explored for disease diagnosis. We performed a meta-analysis to examine miRNA diagnostic value for SZ and further validated the meta-analysis results. Using following terms: schizophrenia/SZ, microRNA/miRNA, diagnosis, sensitivity and specificity, we searched databases restricted to English language and reviewed all articles published from January 1990 to October 2016. All extracted data were statistically analyzed and the results were further validated with peripheral blood mononuclear cells (PBMNCs) isolated from patients and healthy controls using RT-qPCR and receiver operating characteristic (ROC) analysis. A total of 6 studies involving 330 patients and 202 healthy controls were included for meta-analysis. The pooled sensitivity, specificity and diagnostic odds ratio were 0.81 (95% CI: 0.75-0.86), 0.81 (95% CI: 0.72-0.88) and 18 (95% CI: 9-34), respectively; the positive and negative likelihood ratio was 4.3 and 0.24 respectively; the area under the curve in summary ROC was 0.87 (95% CI: 0.84-0.90). Validation revealed that miR-181b-5p, miR-21-5p, miR-195-5p, miR-137, miR-346 and miR-34a-5p in PBMNCs had high diagnostic sensitivity and specificity in the context of schizophrenia. In conclusion, blood-derived miRNAs might be promising biomarkers for SZ diagnosis.
Collapse
|
31
|
Tsuji M, Kawasaki T, Matsuda T, Arai T, Gojo S, Takeuchi JK. Sexual dimorphisms of mRNA and miRNA in human/murine heart disease. PLoS One 2017; 12:e0177988. [PMID: 28704447 PMCID: PMC5509429 DOI: 10.1371/journal.pone.0177988] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 05/05/2017] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Sexual dimorphisms are well recognized in various cardiac diseases such as ischemic cardiomyopathy (ICM), hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM). Thorough understanding of the underlying genetic programs is crucial to optimize treatment strategies specified for each gender. By performing meta-analysis and microarray analysis, we sought to comprehensively characterize the sexual dimorphisms in the healthy and diseased heart at the level of both mRNA and miRNA transcriptome. RESULTS Existing mRNA microarray data of both mouse and human heart were integrated, identifying dozens/ hundreds of sexually dimorphic genes in healthy heart, ICM, HCM, and DCM. These sexually dimorphic genes overrepresented gene ontologies (GOs) important for cardiac homeostasis. Further, microarray of miRNA, isolated from mouse sham left ventricle (LV) (n = 6 & n = 5 for male & female) and chronic MI LV (n = 19 & n = 19) and from human normal LV (n = 6 & n = 6) and ICM LV (n = 4 & n = 5), was conducted. This revealed that 13 mouse miRNAs are sexually dimorphic in MI and 6 in normal heart. In human, 3 miRNAs were sexually dimorphic in ICM and 15 in normal heart. These data revealed miRNA-mRNA networks that operate in a sexually-biased fashion. CONCLUSIONS mRNA and miRNA transcriptome of normal and disease heart show significant sex differences, which might impact the cardiac homeostasis. Together this study provides the first comprehensive picture of the genome-wide program underlying the heart sexual dimorphisms, laying the foundation for gender specific treatment strategies.
Collapse
Affiliation(s)
- Masato Tsuji
- Division of Bio-informational Pharmacology, Medical Research Institute, Tokyo Medical Dental University, Tokyo, Japan
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
- * E-mail: (MT); (JKT)
| | - Takanori Kawasaki
- Department of Cardiovascular Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takeru Matsuda
- Department of Mathematical Informatics, Graduate School of Information Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Tomio Arai
- Department of Pathology, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Tokyo, Japan
| | - Satoshi Gojo
- Department of Cardiovascular Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Jun K. Takeuchi
- Division of Bio-informational Pharmacology, Medical Research Institute, Tokyo Medical Dental University, Tokyo, Japan
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
- * E-mail: (MT); (JKT)
| |
Collapse
|
32
|
Ragan C, Patel K, Edson J, Zhang ZH, Gratten J, Mowry B. Small non-coding RNA expression from anterior cingulate cortex in schizophrenia shows sex specific regulation. Schizophr Res 2017; 183:82-87. [PMID: 27916288 DOI: 10.1016/j.schres.2016.11.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 11/15/2016] [Accepted: 11/16/2016] [Indexed: 01/09/2023]
Abstract
MicroRNAs (miRNAs) are known to regulate the expression of genes that are important for brain development and function, but the roles of other classes of small non-coding RNAs (sncRNAs) are less well understood. Additionally, although miRNA expression studies have been conducted in post-mortem brain samples from schizophrenia (SCZ) patients, other classes of sncRNAs are yet to be investigated in SCZ. We profiled the expression of miRNAs, piwi-interacting RNAs (piRNAs), small nucleolar RNAs (snoRNAs) and small nuclear RNAs (snRNAs) in SCZ by applying small RNA sequencing (RNA-Seq) to sncRNA isolated from post-mortem anterior cingulate cortex (ACC) of SCZ-affected individuals (n=22) and matched controls (n=22). We identified about one-third of annotated miRNAs, one-quarter of snoRNAs and a small proportion of piRNAs and snRNAs. No sncRNAs were significantly differentially expressed between SCZ and controls, but there was evidence for an interaction between disease status and sex on the expression level of a number of miRNAs and snoRNAs. Many of these transcripts exhibited differential expression between male and female cases, and/or between female cases and controls, suggesting sex based dysregulation in ACC of SCZ. These findings require replication in an independent sample, but our study provides further insights into the potential involvement of sncRNAs in brain function and SCZ.
Collapse
Affiliation(s)
- Chikako Ragan
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Kalpana Patel
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia; Queensland Centre for Mental Health Research, The University of Queensland, Brisbane, Queensland, Australia
| | - Janette Edson
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia; The Diamantina Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Zong-Hong Zhang
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Jacob Gratten
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Bryan Mowry
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia; Queensland Centre for Mental Health Research, The University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
33
|
Swathy B, Banerjee M. Understanding epigenetics of schizophrenia in the backdrop of its antipsychotic drug therapy. Epigenomics 2017; 9:721-736. [DOI: 10.2217/epi-2016-0106] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
The diatheses of gene and environment interaction in schizophrenia (SCZ) are becoming increasingly evident. Genetic and epigenetic backgrounds are being considered in stratifying and addressing phenotypic variation and drug response in SCZ. But how much of these epigenetic alterations are the primary contributing factor, toward disease pathogenesis and drug response, needs further clarity. Evidence indicates that antipsychotic drugs can also alter the epigenetic homeostasis thereby inducing pharmacoepigenomic effects. We re-examine the context of epigenetics in disease pathogenesis and antipsychotic drug therapy in SCZ to understand how much of these observations act as real indicators of the disease or therapeutic response. We propose that epigenetic viewpoint in SCZ needs to be critically examined under the genetic, epigenetic and pharmacoepigenetic background.
Collapse
Affiliation(s)
- Babu Swathy
- Human Molecular Genetics Laboratory, Rajiv Gandhi Center for Biotechnology, Thiruvananthapuram, India
| | - Moinak Banerjee
- Human Molecular Genetics Laboratory, Rajiv Gandhi Center for Biotechnology, Thiruvananthapuram, India
| |
Collapse
|
34
|
Alural B, Genc S, Haggarty SJ. Diagnostic and therapeutic potential of microRNAs in neuropsychiatric disorders: Past, present, and future. Prog Neuropsychopharmacol Biol Psychiatry 2017; 73:87-103. [PMID: 27072377 PMCID: PMC5292013 DOI: 10.1016/j.pnpbp.2016.03.010] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 03/28/2016] [Accepted: 03/30/2016] [Indexed: 12/12/2022]
Abstract
Neuropsychiatric disorders are common health problems affecting approximately 1% of the population. Twin, adoption, and family studies have displayed a strong genetic component for many of these disorders; however, the underlying pathophysiological mechanisms and neural substrates remain largely unknown. Given the critical need for new diagnostic markers and disease-modifying treatments, expanding the focus of genomic studies of neuropsychiatric disorders to include the role of non-coding RNAs (ncRNAs) is of growing interest. Of known types of ncRNAs, microRNAs (miRNAs) are 20-25-nucleotide, single-stranded, molecules that regulate gene expression through post-transcriptional mechanisms and have the potential to coordinately regulate complex regulatory networks. In this review, we summarize the current knowledge on miRNA alteration/dysregulation in neuropsychiatric disorders, with a special emphasis on schizophrenia (SCZ), bipolar disorder (BD), and major depressive disorder (MDD). With an eye toward the future, we also discuss the diagnostic and prognostic potential of miRNAs for neuropsychiatric disorders in the context of personalized treatments and network medicine.
Collapse
Affiliation(s)
- Begum Alural
- Department of Neuroscience, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey; Izmir Biomedicine and Genome Center, Dokuz Eylul University, Izmir, Turkey
| | - Sermin Genc
- Department of Neuroscience, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey; Izmir Biomedicine and Genome Center, Dokuz Eylul University, Izmir, Turkey
| | - Stephen J Haggarty
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA; Chemical Neurobiology Laboratory, Departments of Neurology and Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
35
|
Guo L, Zhang Q, Ma X, Wang J, Liang T. miRNA and mRNA expression analysis reveals potential sex-biased miRNA expression. Sci Rep 2017; 7:39812. [PMID: 28045090 PMCID: PMC5206641 DOI: 10.1038/srep39812] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 11/25/2016] [Indexed: 12/22/2022] Open
Abstract
Recent studies suggest that mRNAs may be differentially expressed between males and females. This study aimed to perform expression analysis of mRNA and its main regulatory molecule, microRNA (miRNA), to discuss the potential sex-specific expression patterns using abnormal expression profiles from The Cancer Genome Atlas database. Generally, deregulated miRNAs and mRNAs had consistent expression between males and females, but some miRNAs may be oppositely expressed in specific diseases: up-regulated in one group and down-regulated in another. Studies of miRNA gene families and clusters further confirmed that these sequence or location related miRNAs might have opposing expression between sexes. The specific miRNA might have greater expression divergence across different groups, suggesting flexible expression across different individuals, especially in tumor samples. The typical analysis regardless of the sex will ignore or balance these sex-specific deregulated miRNAs. Compared with flexible miRNAs, their targets of mRNAs showed relative stable expression between males and females. These relevant results provide new insights into miRNA-mRNA interaction and sex difference.
Collapse
Affiliation(s)
- Li Guo
- Department of Bioinformatics, School of Geographic and Biologic Information, Nanjing University of Posts and Telecommunications, Nanjing, 210023, China
| | - Qiang Zhang
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Xiao Ma
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Jun Wang
- Department of Bioinformatics, School of Geographic and Biologic Information, Nanjing University of Posts and Telecommunications, Nanjing, 210023, China
| | - Tingming Liang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210023, China
| |
Collapse
|
36
|
The early growth response protein 1-miR-30a-5p-neurogenic differentiation factor 1 axis as a novel biomarker for schizophrenia diagnosis and treatment monitoring. Transl Psychiatry 2017; 7:e998. [PMID: 28072411 PMCID: PMC5545732 DOI: 10.1038/tp.2016.268] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 10/22/2016] [Accepted: 11/13/2016] [Indexed: 01/09/2023] Open
Abstract
To date, diagnosis of schizophrenia is still based on clinical interviews and careful observations, which is subjective and variable, and can lead to misdiagnosis and/or delay in diagnosis. As early intervention in schizophrenia is important in improving outcomes, objective tests that can be used for schizophrenia diagnosis or treatment monitoring are thus in great need. MicroRNAs (miRNAs) negatively regulate target gene expression and their biogenesis is tightly controlled by various factors including transcription factors (TFs). Dysregulation of miRNAs in brain tissue and peripheral blood mononuclear cells (PBMNCs) from patients with schizophrenia has been well documented, but analysis of the sensitivity and specificity for potential diagnostic utility of these alternations is limited. In this study, we explored the TF-miRNA-30-target gene axis as a novel biomarker for schizophrenia diagnosis and treatment monitoring. Using bioinformatics analysis, we retrieved all TFs that control the biogenesis of miRNA 30 members as well as all target genes that are regulated by miRNA-30 members. Further, reverse transcription-quantitative PCR analysis revealed that the early growth response protein 1 (EGR1) and miR-30a-5p were remarkably downregulated, whereas neurogenic differentiation factor 1 (NEUROD1) was significantly upregulated in PBMNCs from patients in acute psychotic state. Antipsychotics treatment resulted in the elevation of EGR1 and miR-30a-5p but the reduction of NEUROD1. Receiver operating characteristic analysis showed that the EGR1-miR-30a-5p-NEUROD1 axis possessed significantly greater diagnostic value than miR-30a-5p alone. Our data suggest the EGR1-miR-30a-5p-NEUROD1 axis might serve as a promising biomarker for diagnosis and treatment monitoring for those patients in acute psychotic state.
Collapse
|
37
|
Lim CH, Zainal NZ, Kanagasundram S, Zain SM, Mohamed Z. Preliminary examination of microRNA expression profiling in bipolar disorder I patients during antipsychotic treatment. Am J Med Genet B Neuropsychiatr Genet 2016; 171:867-74. [PMID: 27177356 DOI: 10.1002/ajmg.b.32457] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 05/02/2016] [Indexed: 02/01/2023]
Abstract
Although major progress has been achieved in research and development of antipsychotic medications for bipolar disorder (BPD), knowledge of the molecular mechanisms underlying this disorder and the action of atypical antipsychotics remains incomplete. The levels of microRNAs (miRNAs)-small non-coding RNA molecules that regulate gene expression, including genes involved in neuronal function and plasticity-are frequently altered in psychiatric disorders. This study aimed to examine changes in miRNA expression in bipolar mania patients after treatment with asenapine and risperidone. Using a miRNA microarray, we analyzed miRNA expression in the blood of 10 bipolar mania patients following 12 weeks of treatment with asenapine or risperidone. Selected miRNAs were validated by using real-time PCR. A total of 16 miRNAs were differentially expressed after treatment in the asenapine group, 14 of which were significantly upregulated and the other two significantly downregulated. However, all three differentially expressed miRNAs in the risperidone group were downregulated. MiRNA target gene prediction and gene ontology analysis revealed significant enrichment for pathways associated with immune system response and regulation of programmed cell death and transcription. Our results suggest that candidate miRNAs may be involved in the mechanism of action of both antipsychotics in bipolar mania. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Chor Hong Lim
- The Pharmacogenomics Laboratory, Faculty of Medicine, Department of Pharmacology, University of Malaya, Kuala Lumpur, Malaysia
| | - Nor Zuraida Zainal
- Faculty of Medicine, Department of Psychological Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Sharmilla Kanagasundram
- Faculty of Medicine, Department of Psychological Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Shamsul Mohd Zain
- The Pharmacogenomics Laboratory, Faculty of Medicine, Department of Pharmacology, University of Malaya, Kuala Lumpur, Malaysia
| | - Zahurin Mohamed
- The Pharmacogenomics Laboratory, Faculty of Medicine, Department of Pharmacology, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
38
|
Liao Q, Wang Y, Cheng J, Dai D, Zhou X, Zhang Y, Li J, Yin H, Gao S, Duan S. DNA methylation patterns of protein-coding genes and long non-coding RNAs in males with schizophrenia. Mol Med Rep 2016; 12:6568-76. [PMID: 26503909 PMCID: PMC4626154 DOI: 10.3892/mmr.2015.4249] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2014] [Accepted: 04/20/2015] [Indexed: 01/06/2023] Open
Abstract
Schizophrenia (SCZ) is one of the most complex mental illnesses affecting ~1% of the population worldwide. SCZ pathogenesis is considered to be a result of genetic as well as epigenetic alterations. Previous studies have aimed to identify the causative genes of SCZ. However, DNA methylation of long non-coding RNAs (lncRNAs) involved in SCZ has not been fully elucidated. In the present study, a comprehensive genome-wide analysis of DNA methylation was conducted using samples from two male patients with paranoid and undifferentiated SCZ, respectively. Methyl-CpG binding domain protein-enriched genome sequencing was used. In the two patients with paranoid and undifferentiated SCZ, 1,397 and 1,437 peaks were identified, respectively. Bioinformatic analysis demonstrated that peaks were enriched in protein-coding genes, which exhibited nervous system and brain functions. A number of these peaks in gene promoter regions may affect gene expression and, therefore, influence SCZ-associated pathways. Furthermore, 7 and 20 lncRNAs, respectively, in the Refseq database were hypermethylated. According to the lncRNA dataset in the NONCODE database, ~30% of intergenic peaks overlapped with novel lncRNA loci. The results of the present study demonstrated that aberrant hypermethylation of lncRNA genes may be an important epigenetic factor associated with SCZ. However, further studies using larger sample sizes are required.
Collapse
|
39
|
Li X, Gao Y, Meng Z, Zhang C, Qi Q. Regulatory role of microRNA-30b and plasminogen activator inhibitor-1 in the pathogenesis of cognitive impairment. Exp Ther Med 2016; 11:1993-1998. [PMID: 27168840 DOI: 10.3892/etm.2016.3162] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 01/15/2016] [Indexed: 02/06/2023] Open
Abstract
The present study aimed to investigate the role of plasminogen activator inhibitor-1 (PAI-1) in drug-induced early cognitive impairment and the underlying mechanism concerning microRNA (miR)-30b. A mouse model of cognitive impairment was established by intraperitoneal injection of scopolamine (2 mg/kg body weight) for 13 days. Behavioral performance was assessed using the Morris water maze (MWM) test. The mRNA expression levels of PAI-1 and miR-30b were detected using quantitative polymerase chain reaction (qPCR). The protein expression levels of PAI-1 in the hippocampus and blood were determined using western blot analysis and enzyme-linked immunosorbent assays. The MWM test demonstrated that, on days 3 and 4, the escape latency was significantly elevated in the model mice in comparison with control group (P<0.05). In addition, the length of swimming path was significantly increased (P<0.05), while the number of times of crossing the platform location was significantly reduced in the model mouse group (P<0.05) in comparison with the control group. qPCR demonstrated that the mRNA expression levels of PAI-1 in the model mice was significantly elevated in the hippocampus and blood in comparison with the control group (P<0.01). Furthermore, western blot analysis and enzyme-linked immunosorbent assay demonstrated that the protein expression levels of PAI-1 were significantly elevated in the hippocampus and blood in the model group, in comparison with the control group (P<0.05). Notably, the levels of miR-30b in the hippocampus and blood were significantly decreased in the model mice in comparison with the control group (P<0.01). To conclude, the expression levels of PAI-1 were significantly elevated in mice with scopolamine-induced cognitive impairment, which may be associated with the downregulation of miR-30b. The findings from the present study suggest that miR-30b may be involved in the regulation of PAI-1, which would contribute to the pathogenesis of cognitive impairment.
Collapse
Affiliation(s)
- Xiuqin Li
- Second Department of Health, Laiwu Hospital Affiliated to Taishan Medical University, Laiwu, Shandong 271100, P.R. China
| | - Yong Gao
- Department of Neurosurgery, Laiwu Hospital Affiliated to Taishan Medical University, Laiwu, Shandong 271100, P.R. China
| | - Zhaoyun Meng
- Second Department of Health, Laiwu Hospital Affiliated to Taishan Medical University, Laiwu, Shandong 271100, P.R. China
| | - Cui Zhang
- Department of Neurology, Laiwu Hospital Affiliated to Taishan Medical University, Laiwu, Shandong 271100, P.R. China
| | - Qinde Qi
- Department of Neurology, Laiwu Hospital Affiliated to Taishan Medical University, Laiwu, Shandong 271100, P.R. China
| |
Collapse
|
40
|
Geaghan M, Cairns MJ. MicroRNA and Posttranscriptional Dysregulation in Psychiatry. Biol Psychiatry 2015; 78:231-9. [PMID: 25636176 DOI: 10.1016/j.biopsych.2014.12.009] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 11/11/2014] [Accepted: 12/03/2014] [Indexed: 11/27/2022]
Abstract
Psychiatric syndromes, including schizophrenia, mood disorders, and autism spectrum disorders, are characterized by a complex range of symptoms, including psychosis, depression, mania, and cognitive deficits. Although the mechanisms driving pathophysiology are complex and remain largely unknown, advances in the understanding of gene association and gene networks are providing significant clues to their etiology. In recent years, small noncoding RNA molecules known as microRNA (miRNA) have emerged as potential players in the pathophysiology of mental illness. These small RNAs regulate hundreds of target transcripts by modifying their stability and translation on a broad scale, influencing entire gene networks in the process. There is evidence to suggest that numerous miRNAs are dysregulated in postmortem neuropathology of neuropsychiatric disorders, and there is strong genetic support for association of miRNA genes and their targets with these conditions. This review presents the accumulated evidence linking miRNA dysregulation and dysfunction with schizophrenia, bipolar disorder, major depressive disorder, and autism spectrum disorders and the potential of miRNAs as biomarkers or therapeutics for these disorders. We further assess the functional roles of some outstanding miRNAs associated with these conditions and how they may be influencing the development of psychiatric symptoms.
Collapse
Affiliation(s)
- Michael Geaghan
- School of Biomedical Sciences, Faculty of Health and Medicine, University of Newcastle, Callaghan, Australia.; Centre for Translational Neuroscience and Mental Health, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Murray J Cairns
- School of Biomedical Sciences, Faculty of Health and Medicine, University of Newcastle, Callaghan, Australia.; Schizophrenia Research Institute, Sydney, Australia.; Centre for Translational Neuroscience and Mental Health, Hunter Medical Research Institute, Newcastle, New South Wales, Australia..
| |
Collapse
|
41
|
Cathcart P, Lucchesi W, Ottaviani S, De Giorgio A, Krell J, Stebbing J, Castellano L. Noncoding RNAs and the control of signalling via nuclear receptor regulation in health and disease. Best Pract Res Clin Endocrinol Metab 2015; 29:529-43. [PMID: 26303081 DOI: 10.1016/j.beem.2015.07.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Nuclear receptors belong to a superfamily of proteins that play central roles in human biology, orchestrating a large variety of biological functions in both health and disease. Understanding the interactions and regulatory pathways of NRs will allow development of potential therapeutic interventions for a multitude of disease processes. Non-coding RNAs have recently been discovered to have significant interactions with NR signalling pathways via a variety of biological connections. This review summarises the known interactions between ncRNAs and the NR superfamily in health, embryogenesis and a plethora of human diseases.
Collapse
Affiliation(s)
- Paul Cathcart
- Department of Surgery and Cancer, Imperial College London, Imperial Centre for Translational and Experimental Medicine, London W12 0NN, UK
| | - Walter Lucchesi
- School of Pharmacy, University of Reading, Whiteknights Reading Berks RG6 6AP, UK
| | - Silvia Ottaviani
- Department of Surgery and Cancer, Imperial College London, Imperial Centre for Translational and Experimental Medicine, London W12 0NN, UK
| | - Alex De Giorgio
- Department of Surgery and Cancer, Imperial College London, Imperial Centre for Translational and Experimental Medicine, London W12 0NN, UK
| | - Jonathan Krell
- Department of Surgery and Cancer, Imperial College London, Imperial Centre for Translational and Experimental Medicine, London W12 0NN, UK
| | - Justin Stebbing
- Department of Surgery and Cancer, Imperial College London, Imperial Centre for Translational and Experimental Medicine, London W12 0NN, UK
| | - Leandro Castellano
- Department of Surgery and Cancer, Imperial College London, Imperial Centre for Translational and Experimental Medicine, London W12 0NN, UK.
| |
Collapse
|
42
|
Khan D, Dai R, Ansar Ahmed S. Sex differences and estrogen regulation of miRNAs in lupus, a prototypical autoimmune disease. Cell Immunol 2015; 294:70-9. [DOI: 10.1016/j.cellimm.2015.01.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 01/05/2015] [Accepted: 01/06/2015] [Indexed: 12/12/2022]
|
43
|
Han F, Huo Y, Huang CJ, Chen CL, Ye J. MicroRNA-30b promotes axon outgrowth of retinal ganglion cells by inhibiting Semaphorin3A expression. Brain Res 2015; 1611:65-73. [PMID: 25791621 DOI: 10.1016/j.brainres.2015.03.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 02/08/2015] [Accepted: 03/06/2015] [Indexed: 10/23/2022]
Abstract
Semaphorin3A (Sema3A) is a major inhibitory factor of optic nerve (ON) regeneration post-injury. Many microRNAs (miRNAs) are expressed specifically in the mammalian brain and retina and are dynamically regulated during development, suggesting that this group of miRNAs may be associated with neural development. We found that microRNA-30b (miR-30b) bound to the three prime untranslated region (3' UTR) of Sema3A and inhibited the expression of Sema3A mRNA. The mRNA expression level of miR-30b and the protein expression levels of Sema3A, Neuropilin1 (NRP1), PlexinA1 (PlexA1), phosphorylated p38MAPK (p-p38MAPK), and active caspase-3 were all upregulated in retinas from rats with a damaged ON relative to those with an intact ON. Transfection of cultured retinal ganglion cells (RGCs) with an miR-30b mimic led to decreased levels of Sema3A, NRP1, PlexA1, p-p38MAPK, and active caspase-3 protein expression, as well as axon elongation and reduced levels of apoptosis. These findings provide evidence that miR-30b inhibits Sema3A expression. Decreased Sema3A expression promotes axon outgrowth in RGCs due to reduced levels of Sema3A binding to NRP1 and PlexA1 and simultaneously reduces apoptosis by inhibiting the p38MAPK and caspase-3 pathways. Our findings provide the first evidence that miR-30b-mediated Sema3A downregulation may serve as a new strategy for the clinical treatment of ON injury.
Collapse
Affiliation(s)
- F Han
- Department of Ophthalmology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Y Huo
- Department of Ophthalmology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - C-J Huang
- Department of Ophthalmology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - C-L Chen
- Department of Ophthalmology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - J Ye
- Department of Ophthalmology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China.
| |
Collapse
|
44
|
DNA methylation patterns of protein coding genes and long noncoding RNAs in female schizophrenic patients. Eur J Med Genet 2015; 58:95-104. [DOI: 10.1016/j.ejmg.2014.12.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 12/04/2014] [Indexed: 12/11/2022]
|
45
|
Chen Y, Fang J, Chen X, Pan C, Liu X, Liu J. Effects of the Treg/Th17 cell balance and their associated cytokines in patients with hepatitis B infection. Exp Ther Med 2014; 8:1671-1676. [PMID: 25371713 PMCID: PMC4217773 DOI: 10.3892/etm.2014.2014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 09/11/2014] [Indexed: 01/13/2023] Open
Abstract
The extent to which T-cell-mediated immunity is impaired in patients with hepatitis B virus (HBV) infection remains controversial. In addition, the role of T regulatory (Treg) and T helper 17 (Th17) cells and their associated cytokines in immunity is not clear. In the present study, peripheral blood samples were collected from 44 patients with chronic hepatitis B virus, 14 asymptomatic hepatitis B carriers, 19 patients with liver cirrhosis and 20 healthy individuals. Flow cytometry was used to detect the percentages of T cell subsets in the samples, including CD3+, CD4+ and CD8+ T cells, Treg cells and Th17 cells. A cytometric bead array was conducted to detect the levels of interleukin (IL)-17, -6, -10 and -21, and transforming growth factor (TGF)-β. The data revealed that Treg cell levels decreased, while Th17 cell levels increased in the peripheral blood of HBV patients. As the extent of inflammation and fibrosis in the hepatic tissue increased, the frequency of Treg and Th17 cells in the peripheral blood did not significantly differ. In addition, the levels of Th17 cells were found to positively correlate with TGF-β and IL-21 levels. Therefore, analyzing the balance between Treg/Th17 cells and their associated cytokines may be a useful indicator in the diagnosis of HBV.
Collapse
Affiliation(s)
- Yi Chen
- The Liver Center of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fuijan 350025, P.R. China
| | - Jiankai Fang
- The Liver Center of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fuijan 350025, P.R. China
| | - Xuzheng Chen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fuijan 350108, P.R. China
| | - Chen Pan
- The Liver Center of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fuijan 350025, P.R. China
| | - Xiaolong Liu
- The Liver Center of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fuijan 350025, P.R. China
| | - Jingfeng Liu
- The Liver Center of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fuijan 350025, P.R. China
| |
Collapse
|
46
|
Hommers LG, Domschke K, Deckert J. Heterogeneity and individuality: microRNAs in mental disorders. J Neural Transm (Vienna) 2014; 122:79-97. [PMID: 25395183 DOI: 10.1007/s00702-014-1338-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 11/07/2014] [Indexed: 12/21/2022]
Abstract
MicroRNAs are about 22 nucleotide long single-stranded RNA molecules, negatively regulating gene expression of a single gene or a gene network. In neural tissues, they have been implicated in developmental and neuroplasticity-related processes, such as neurogenesis, differentiation, apoptosis and long-term potentiation. Their molecular mode of action is reminiscent of findings of genome-wide association studies in mental disorders, unable to attribute the risk of disease to a specific gene, but rather to multiple genes, gene-networks and gene-environment interaction. As such, microRNAs are an attractive target for research. Here, we review clinical studies conducted in humans on microRNAs in mental disorders with a particular focus on schizophrenia, bipolar disorder, major depressive disorder and anxiety disorders. The majority of clinical studies have focused on schizophrenia. The most robust finding has been reported for rs1625579 located in MIR137HG, which was associated with schizophrenia on a genome-wide level. Concerning bipolar disorder, major depression and anxiety disorders, promising results have been published, but only a considerably smaller number of clinical studies is available and genome-wide association studies did not suggest a direct link to microRNAs so far. Expression of microRNAs as biomarkers of mental disorders and treatment response is currently emerging with preliminary results. Larger-scaled genetic and functional studies along with translational research are needed to enhance our understanding of microRNAs in mental disorders. These studies will aid in disentangling the complex genetic nature of these disorders and possibly contribute to the development of novel, individualized diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Leif G Hommers
- Center of Mental Health, Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital Würzburg, Füchsleinstrasse 15, 97080, Würzburg, Germany,
| | | | | |
Collapse
|
47
|
MicroRNAs in Schizophrenia: Implications for Synaptic Plasticity and Dopamine-Glutamate Interaction at the Postsynaptic Density. New Avenues for Antipsychotic Treatment Under a Theranostic Perspective. Mol Neurobiol 2014; 52:1771-1790. [PMID: 25394379 DOI: 10.1007/s12035-014-8962-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 10/23/2014] [Indexed: 12/17/2022]
Abstract
Despite dopamine-glutamate aberrant interaction that has long been considered a relevant landmark of psychosis pathophysiology, several aspects of these two neurotransmitters reciprocal interaction remain to be defined. The emerging role of postsynaptic density (PSD) proteins at glutamate synapse as a molecular "lego" making a functional hub where different signals converge may add a new piece of information to understand how dopamine-glutamate interaction may work with regard to schizophrenia pathophysiology and treatment. More recently, compelling evidence suggests a relevant role for microRNA (miRNA) as a new class of dopamine and glutamate modulators with regulatory functions in the reciprocal interaction of these two neurotransmitters. Here, we aimed at addressing the following issues: (i) Do miRNAs have a role in schizophrenia pathophysiology in the context of dopamine-glutamate aberrant interaction? (ii) If miRNAs are relevant for dopamine-glutamate interaction, at what level this modulation takes place? (iii) Finally, will this knowledge open the door to innovative diagnostic and therapeutic tools? The biogenesis of miRNAs and their role in synaptic plasticity with relevance to schizophrenia will be considered in the context of dopamine-glutamate interaction, with special focus on miRNA interaction with PSD elements. From this framework, implications both for biomarkers identification and potential innovative interventions will be considered.
Collapse
|
48
|
Sun E, Shi Y. MicroRNAs: Small molecules with big roles in neurodevelopment and diseases. Exp Neurol 2014; 268:46-53. [PMID: 25128264 DOI: 10.1016/j.expneurol.2014.08.005] [Citation(s) in RCA: 134] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 07/29/2014] [Accepted: 08/05/2014] [Indexed: 01/13/2023]
Abstract
MicroRNAs (miRNAs) are single-stranded, non-coding RNA molecules that play important roles in the development and functions of the brain. Extensive studies have revealed critical roles for miRNAs in brain development and function. Dysregulation or altered expression of miRNAs is associated with abnormal brain development and pathogenesis of neurodevelopmental diseases. This review serves to highlight the versatile roles of these small RNA molecules in normal brain development and their association with neurodevelopmental disorders, in particular, two closely related neuropsychiatric disorders of neurodevelopmental origin, schizophrenia and bipolar disorder.
Collapse
Affiliation(s)
- Emily Sun
- Department of Neurosciences, Cancer Center, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Yanhong Shi
- Department of Neurosciences, Cancer Center, Beckman Research Institute of City of Hope, Duarte, CA, USA.
| |
Collapse
|
49
|
Kolshus E, Dalton VS, Ryan KM, McLoughlin DM. When less is more--microRNAs and psychiatric disorders. Acta Psychiatr Scand 2014; 129:241-56. [PMID: 23952691 DOI: 10.1111/acps.12191] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/11/2013] [Indexed: 12/16/2022]
Abstract
OBJECTIVE MicroRNAs are small non-coding RNA molecules that regulate gene expression, including genes involved in neuronal function and plasticity that have relevance for brain function and mental health. We therefore performed a systematic review of miRNAs in general adult psychiatric disorders. METHOD Systematic searches in PubMed/MEDLINE and Web of Science were conducted to identify published clinical articles on microRNAs in general adult psychiatric disorders. We also reviewed references from included articles. RESULTS There is mounting evidence of microRNAs' regulatory roles in a number of central nervous system processes, including neurogenesis and synaptic plasticity. The majority of clinical studies of microRNAs in psychiatric disorders are in schizophrenia, where a number of specific microRNAs have been identified in separate studies. There is some evidence of marked downregulation of some microRNAs in affective disorders. Treatment with antidepressants appears to upregulate microRNA levels. There is currently little evidence from human studies in anxiety, addiction or other psychiatric disorders. CONCLUSION MicroRNA research in psychiatry is currently in a nascent period, but represents an emerging and exciting area, with the potential to clarify molecular mechanisms of disease and identify novel biomarkers and therapeutic agents.
Collapse
Affiliation(s)
- E Kolshus
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland; Department of Psychiatry, Trinity College Dublin, St. Patrick's University Hospital, Dublin 8, Ireland
| | | | | | | |
Collapse
|
50
|
Kohen R, Dobra A, Tracy JH, Haugen E. Transcriptome profiling of human hippocampus dentate gyrus granule cells in mental illness. Transl Psychiatry 2014; 4:e366. [PMID: 24594777 PMCID: PMC3966046 DOI: 10.1038/tp.2014.9] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 01/06/2014] [Indexed: 12/20/2022] Open
Abstract
This study is, to the best of our knowledge, the first application of whole transcriptome sequencing (RNA-seq) to cells isolated from postmortem human brain by laser capture microdissection. We investigated the transcriptome of dentate gyrus (DG) granule cells in postmortem human hippocampus in 79 subjects with mental illness (schizophrenia, bipolar disorder, major depression) and nonpsychiatric controls. We show that the choice of normalization approach for analysis of RNA-seq data had a strong effect on results; under our experimental conditions a nonstandard normalization method gave superior results. We found evidence of disrupted signaling by miR-182 in mental illness. This was confirmed using a novel method of leveraging microRNA genetic variant information to indicate active targeting. In healthy subjects and those with bipolar disorder, carriers of a high- vs those with a low-expressing genotype of miR-182 had different levels of miR-182 target gene expression, indicating an active role of miR-182 in shaping the DG transcriptome for those subject groups. By contrast, comparing the transcriptome between carriers of different genotypes among subjects with major depression and schizophrenia suggested a loss of DG miR-182 signaling in these conditions.
Collapse
Affiliation(s)
- R Kohen
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, USA,Department of Psychiatry and Behavioral Sciences, University of Washington, 1959 Pacific Avenue NE, Seattle, WA 98195-6560, USA. E-mail:
| | - A Dobra
- Department of Statistics, University of Washington, Seattle, WA, USA,Department of Biobehavioral Nursing and Health Systems, University of Washington, Seattle, WA, USA,Center for Statistics and The Social Sciences, University of Washington, Seattle, WA, USA
| | - J H Tracy
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, USA
| | - E Haugen
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| |
Collapse
|