1
|
Chowdhury MAR, Haq MM, Lee JH, Jeong S. Multi-faceted regulation of CREB family transcription factors. Front Mol Neurosci 2024; 17:1408949. [PMID: 39165717 PMCID: PMC11333461 DOI: 10.3389/fnmol.2024.1408949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 07/12/2024] [Indexed: 08/22/2024] Open
Abstract
cAMP response element-binding protein (CREB) is a ubiquitously expressed nuclear transcription factor, which can be constitutively activated regardless of external stimuli or be inducibly activated by external factors such as stressors, hormones, neurotransmitters, and growth factors. However, CREB controls diverse biological processes including cell growth, differentiation, proliferation, survival, apoptosis in a cell-type-specific manner. The diverse functions of CREB appear to be due to CREB-mediated differential gene expression that depends on cAMP response elements and multi-faceted regulation of CREB activity. Indeed, the transcriptional activity of CREB is controlled at several levels including alternative splicing, post-translational modification, dimerization, specific transcriptional co-activators, non-coding small RNAs, and epigenetic regulation. In this review, we present versatile regulatory modes of CREB family transcription factors and discuss their functional consequences.
Collapse
Affiliation(s)
- Md Arifur Rahman Chowdhury
- Department of Bioactive Material Sciences, Jeonbuk National University, Jeonju, Republic of Korea
- Department of Molecular Biology, and Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, Republic of Korea
| | - Md Mazedul Haq
- Department of Bioactive Material Sciences, Jeonbuk National University, Jeonju, Republic of Korea
- Department of Molecular Biology, and Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, Republic of Korea
| | - Jeong Hwan Lee
- Division of Life Sciences, Jeonbuk National University, Jeonju, Republic of Korea
| | - Sangyun Jeong
- Department of Bioactive Material Sciences, Jeonbuk National University, Jeonju, Republic of Korea
- Department of Molecular Biology, and Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, Republic of Korea
| |
Collapse
|
2
|
Zhu J, Hou Y, Li W, Wang X, Li F, Li N, Hu Y, Wang X, Ge SN. miR-181a expressed in the dorsal hippocampus regulates the reinstatement of cocaine CPP by targeting PRKAA1. Behav Brain Res 2024; 471:115097. [PMID: 38878971 DOI: 10.1016/j.bbr.2024.115097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 06/04/2024] [Accepted: 06/04/2024] [Indexed: 06/21/2024]
Abstract
Neuroadaptive changes in the hippocampus underlie addictive-like behaviors in humans or animals chronically exposed to cocaine. miR-181a, which is widely expressed in the hippocampus, acts as a regulator for synaptic plasticity, while its role in drug reinstatement is unclear. In this study, we found that miR-181a regulates the reinstatement of cocaine conditioned place preference(CPP), and altered miR-181a expression changes the complexity of hippocampal neurons and the density and morphology of dendritic spines. By using a luciferase gene reporter, we found that miR-181a targets PRKAA1, an upstream molecule in the mTOR pathway. High miR-181a expression reduced the expression of the PRKAA1 mRNA and promoted mTOR activity and the reinstatement of cocaine CPP. These results indicate that miR-181a is involved in neuronal structural plasticity induced by reinstatement of cocaine CPP, possibly through the activation of the mTOR signaling pathway. This study provides new microRNA targets and a theoretical foundation for the prevention of cocaine-induced reinstatement.
Collapse
Affiliation(s)
- Jun Zhu
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710038, China; Shaanxi University of Chinese Medicine, Xian Yang, Shaanxi 712046, China
| | - Yueru Hou
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710038, China; Shaanxi University of Chinese Medicine, Xian Yang, Shaanxi 712046, China
| | - Wan Li
- Xi'an Technological University, Xi'an 710021, China
| | - Xin Wang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710038, China
| | - Fei Li
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710038, China
| | - Nan Li
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710038, China
| | - Yan Hu
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710038, China
| | - Xuelian Wang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710038, China.
| | - Shun-Nan Ge
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710038, China.
| |
Collapse
|
3
|
Adam M, Ozcan S, Dalkilic S, Tektemur NK, Tekin S, Bilgin B, Hekim MG, Bulut F, Kelestemur MM, Canpolat S, Ozcan M. Modulation of Neuronal Damage in DRG by Asprosin in a High-Glucose Environment and Its Impact on miRNA181-a Expression in Diabetic DRG. Neurotox Res 2023; 42:5. [PMID: 38133838 DOI: 10.1007/s12640-023-00678-9] [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: 05/18/2023] [Revised: 10/09/2023] [Accepted: 12/02/2023] [Indexed: 12/23/2023]
Abstract
Asprosin, a hormone secreted from adipose tissue, has been implicated in the modulation of cell viability. Current studies suggest that neurological impairments are increased in individuals with obesity-linked diabetes, likely due to the presence of excess adipose tissue, but the precise molecular mechanism behind this association remains poorly understood. In this study, our hypothesis that asprosin has the potential to mitigate neuronal damage in a high glucose (HG) environment while also regulating the expression of microRNA (miRNA)-181a, which is involved in critical biological processes such as cellular survival, apoptosis, and autophagy. To investigate this, dorsal root ganglion (DRG) neurons were exposed to asprosin in a HG (45 mmol/L) environment for 24 hours, with a focus on the role of the protein kinase A (PKA) pathway. Expression of miRNA-181a was measured by using real-time polymerase chain reaction (RT-PCR) in diabetic DRG. Our findings revealed a decline in cell viability and an upregulation of apoptosis under HG conditions. However, pretreatment with asprosin in sensory neurons effectively improved cell viability and reduced apoptosis by activating the PKA pathway. Furthermore, we observed that asprosin modulated the expression of miRNA-181a in diabetic DRG. Our study demonstrates that asprosin has the potential to protect DRG neurons from HG-induced damage while influencing miRNA-181a expression in diabetic DRG. These findings provide valuable insights for the development of clinical interventions targeting neurotoxicity in diabetes, with asprosin emerging as a promising therapeutic target for managing neurological complications in affected individuals.
Collapse
Affiliation(s)
- Muhammed Adam
- Faculty of Medicine (TIP FAKULTESI), Department of Biophysics, University of Firat, Elazig, TR23119, Turkey
| | - Sibel Ozcan
- Department of Anaesthesiology and Reanimation, University of Firat, Elazig, Turkey
| | - Semih Dalkilic
- Department of Biology, University of Firat, Elazig, Turkey
| | | | - Suat Tekin
- Department of Physiology, University of Inonu, Malatya, Turkey
| | - Batuhan Bilgin
- Faculty of Medicine (TIP FAKULTESI), Department of Biophysics, University of Firat, Elazig, TR23119, Turkey
| | | | - Ferah Bulut
- Faculty of Medicine (TIP FAKULTESI), Department of Biophysics, University of Firat, Elazig, TR23119, Turkey
| | | | - Sinan Canpolat
- Department of Physiology, University of Firat, Elazig, Turkey
| | - Mete Ozcan
- Faculty of Medicine (TIP FAKULTESI), Department of Biophysics, University of Firat, Elazig, TR23119, Turkey.
| |
Collapse
|
4
|
Vasilieva AA, Timechko EE, Lysova KD, Paramonova AI, Yakimov AM, Kantimirova EA, Dmitrenko DV. MicroRNAs as Potential Biomarkers of Post-Traumatic Epileptogenesis: A Systematic Review. Int J Mol Sci 2023; 24:15366. [PMID: 37895044 PMCID: PMC10607802 DOI: 10.3390/ijms242015366] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Structural or post-traumatic epilepsy often develops after brain tissue damage caused by traumatic brain injury, stroke, infectious diseases of the brain, etc. Most often, between the initiating event and epilepsy, there is a period without seizures-a latent period. At this time, the process of restructuring of neural networks begins, leading to the formation of epileptiform activity, called epileptogenesis. The prediction of the development of the epileptogenic process is currently an urgent and difficult task. MicroRNAs are inexpensive and minimally invasive biomarkers of biological and pathological processes. The aim of this study is to evaluate the predictive ability of microRNAs to detect the risk of epileptogenesis. In this study, we conducted a systematic search on the MDPI, PubMed, ScienceDirect, and Web of Science platforms. We analyzed publications that studied the aberrant expression of circulating microRNAs in epilepsy, traumatic brain injury, and ischemic stroke in order to search for microRNAs-potential biomarkers for predicting epileptogenesis. Thus, 31 manuscripts examining biomarkers of epilepsy, 19 manuscripts examining biomarkers of traumatic brain injury, and 48 manuscripts examining biomarkers of ischemic stroke based on circulating miRNAs were analyzed. Three miRNAs were studied: miR-21, miR-181a, and miR-155. The findings showed that miR-21 and miR-155 are associated with cell proliferation and apoptosis, and miR-181a is associated with protein modifications. These miRNAs are not strictly specific, but they are involved in processes that may be indirectly associated with epileptogenesis. Also, these microRNAs may be of interest when they are studied in a cohort with each other and with other microRNAs. To further study the microRNA-based biomarkers of epileptogenesis, many factors must be taken into account: the time of sampling, the type of biological fluid, and other nuances. Currently, there is a need for more in-depth and prolonged studies of epileptogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Diana V. Dmitrenko
- Department of Medical Genetics and Clinical Neurophysiology of Postgraduate Education, V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk 660022, Russia; (A.A.V.); (E.E.T.); (K.D.L.); (A.I.P.)
| |
Collapse
|
5
|
Jiang MJ, Li J, Luo CH, Zhu C, Chen ZJ, Bai W, Hu TY, Feng CH, Li C, Mo ZX. Rhynchophylline inhibits methamphetamine dependence via modulating the miR-181a-5p/GABRA1 axis. JOURNAL OF ETHNOPHARMACOLOGY 2023; 314:116635. [PMID: 37182675 DOI: 10.1016/j.jep.2023.116635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 04/25/2023] [Accepted: 05/12/2023] [Indexed: 05/16/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Uncaria rhynchophylla (Miq.) Miq. ex Havil. is a plant species that is routinely devoted in traditional Chinese medicine to treat central nervous system disorders. Rhynchophylline (Rhy), a predominant alkaloid isolated from Uncaria rhynchophylla (Miq.) Miq. ex Havil., has been demonstrated to reverse methamphetamine-induced (METH-induced) conditioned place preference (CPP) effects in mice, rats and zebrafish. The precise mechanism is still poorly understood, thus further research is necessary. AIM OF STUDY This study aimed to investigate the role of miRNAs in the inhibitory effect of Rhy on METH dependence. MATERIALS AND METHODS A rat CPP paradigm and a PC12 cell addiction model were established. Microarray assays were used to screen and identify the candidate miRNA. Behavioral assessment, real-time PCR, dual-luciferase reporter assay, western blotting, stereotaxic injection of antagomir/agomir and cell transfection experiments were performed to elucidate the effect of the candidate miRNA and intervention mechanism of Rhy on METH dependence. RESULTS Rhy successfully reversed METH-induced CPP effect and the upregulated miR-181a-5p expression in METH-dependent rat hippocampus and PC12 cells. Moreover, suppression of miR-181a-5p by antagomir 181a reversed METH-induced CPP effect. Meanwhile, overexpression of miR-181a-5p by agomir 181a in combination with low-dose METH (0.5 mg/kg) elicited a significant CPP effect, which was blocked by Rhy through inhibiting miR-181a-5p. Finally, the result demonstrated that miR-181a-5p exerted its regulatory role by targeting γ-aminobutyric acid A receptor α1 (GABRA1) both in vivo and in vitro. CONCLUSION This finding reveals that Rhy inhibits METH dependence via modulating the miR-181a-5p/GABRA1 axis, which may be a promising target for treatment of METH dependence.
Collapse
Affiliation(s)
- Ming-Jin Jiang
- Jiangxi Provincial Institute of Translational Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China.
| | - Jing Li
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China.
| | - Chao-Hua Luo
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China.
| | - Chen Zhu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China.
| | - Zhi-Jie Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China.
| | - Wei Bai
- Jiangxi Provincial Institute of Translational Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| | - Tian-Yu Hu
- Jiangxi Provincial Institute of Translational Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| | - Chuan-Hua Feng
- Jiangxi Provincial Institute of Translational Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| | - Chan Li
- School of Life Sciences, Guangzhou University, Guangzhou, 510006, China.
| | - Zhi-Xian Mo
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
6
|
Schäfer A, Evers L, Meier L, Schlomann U, Bopp MHA, Dreizner GL, Lassmann O, Ben Bacha A, Benescu AC, Pojskic M, Preußer C, von Strandmann EP, Carl B, Nimsky C, Bartsch JW. The Metalloprotease-Disintegrin ADAM8 Alters the Tumor Suppressor miR-181a-5p Expression Profile in Glioblastoma Thereby Contributing to Its Aggressiveness. Front Oncol 2022; 12:826273. [PMID: 35371977 PMCID: PMC8964949 DOI: 10.3389/fonc.2022.826273] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 02/16/2022] [Indexed: 01/08/2023] Open
Abstract
Glioblastoma (GBM) as the most common and aggressive brain tumor is characterized by genetic heterogeneity, invasiveness, radio-/chemoresistance, and occurrence of GBM stem-like cells. The metalloprotease-disintegrin ADAM8 is highly expressed in GBM tumor and immune cells and correlates with poor survival. In GBM, ADAM8 affects intracellular kinase signaling and increases expression levels of osteopontin/SPP1 and matrix metalloproteinase 9 (MMP9) by an unknown mechanism. Here we explored whether microRNA (miRNA) expression levels could be regulators of MMP9 expression in GBM cells expressing ADAM8. Initially, we identified several miRNAs as dysregulated in ADAM8-deficient U87 GBM cells. Among these, the tumor suppressor miR-181a-5p was significantly upregulated in ADAM8 knockout clones. By inhibiting kinase signaling, we found that ADAM8 downregulates expression of miR-181a-5p via activation of signal transducer and activator of transcription 3 (STAT3) and mitogen-activated protein kinase (MAPK) signaling suggesting an ADAM8-dependent silencing of miR-181a-5p. In turn, mimic miR-181a-5p transfection caused decreased cell proliferation and lower MMP9 expression in GBM cells. Furthermore, miR-181a-5p was detected in GBM cell-derived extracellular vesicles (EVs) as well as patient serum-derived EVs. We identified miR-181a-5p downregulating MMP9 expression via targeting the MAPK pathway. Analysis of patient tissue samples (n=22) revealed that in GBM, miR-181a-5p is strongly downregulated compared to ADAM8 and MMP9 mRNA expression, even in localized tumor areas. Taken together, we provide evidence for a functional axis involving ADAM8/miR-181a-5p/MAPK/MMP9 in GBM tumor cells.
Collapse
Affiliation(s)
- Agnes Schäfer
- Department of Neurosurgery, Philipps University Marburg, Marburg, Germany
| | - Lara Evers
- Department of Neurosurgery, Philipps University Marburg, Marburg, Germany
| | - Lara Meier
- Department of Neurosurgery, Philipps University Marburg, Marburg, Germany
| | - Uwe Schlomann
- Department of Neurosurgery, Philipps University Marburg, Marburg, Germany
| | - Miriam H A Bopp
- Department of Neurosurgery, Philipps University Marburg, Marburg, Germany.,Marburg Center for Mind, Brain and Behavior (MCMBB), Marburg, Germany
| | - Gian-Luca Dreizner
- Department of Neurosurgery, Philipps University Marburg, Marburg, Germany
| | - Olivia Lassmann
- Department of Neurosurgery, Philipps University Marburg, Marburg, Germany
| | - Aaron Ben Bacha
- Department of Neurosurgery, Philipps University Marburg, Marburg, Germany
| | | | - Mirza Pojskic
- Department of Neurosurgery, Philipps University Marburg, Marburg, Germany
| | - Christian Preußer
- Core Facility Extracellular Vesicles, Philipps University of Marburg - Medical Faculty, Marburg, Germany
| | - Elke Pogge von Strandmann
- Core Facility Extracellular Vesicles, Philipps University of Marburg - Medical Faculty, Marburg, Germany
| | - Barbara Carl
- Department of Neurosurgery, Philipps University Marburg, Marburg, Germany
| | - Christopher Nimsky
- Department of Neurosurgery, Philipps University Marburg, Marburg, Germany.,Marburg Center for Mind, Brain and Behavior (MCMBB), Marburg, Germany
| | - Jörg W Bartsch
- Department of Neurosurgery, Philipps University Marburg, Marburg, Germany.,Marburg Center for Mind, Brain and Behavior (MCMBB), Marburg, Germany
| |
Collapse
|
7
|
Fu Y, Xin Z, Ling Z, Xie H, Xiao T, Shen X, Lin J, Xu L, Jiang H. A CREB1-miR-181a-5p loop regulates the pathophysiologic features of bone marrow stromal cells in fibrous dysplasia of bone. Mol Med 2021; 27:81. [PMID: 34294046 PMCID: PMC8296714 DOI: 10.1186/s10020-021-00341-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 07/06/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Fibrous dysplasia (FD) is a bone marrow stromal cell (BMSC) disease caused by activating mutations of guanine nucleotide-binding protein alpha-stimulating activity polypeptide (GNAS) and is characterized by increased proliferative activity and disrupted osteogenesis of BMSCs. However, the molecular mechanisms regulating the pathophysiologic features of BMSCs in FD remain unknown. This study aimed to identify and verify the roles of the CREB1-miR-181a-5p regulatory loop in FD pathophysiology. METHODS MicroRNA (miRNA) sequencing analysis was used to identify the possible miRNAs implicated in FD. The proliferation, apoptosis, and osteogenic differentiation of BMSCs, as well as the osteoclast-induced phenotype, were measured and compared after exogenous miR-181a-5p transfection into FD BMSCs or miR-181a-5p inhibitor transfection into normal BMSCs. Chromatin immunoprecipitation and luciferase reporter assays were performed to verify the interactions between CREB1 and miR-181a-5p and their effects on the FD pathological phenotype. RESULTS Compared to normal BMSCs, FD BMSCs showed decreased miR-181a-5p levels and exhibited increased proliferative activity, decreased apoptotic capacity, and impaired osteogenesis. FD BMSCs also showed a stronger osteoclast activation effect. miR-181a-5p overexpression reversed the pathophysiologic features of FD BMSCs, whereas miR-181a-5p suppression induced an FD-like phenotype in normal BMSCs. Mechanistically, miR-181a-5p was the downstream target of CREB1, and CREB1 was posttranscriptionally regulated by miR-181a-5p. CONCLUSIONS Our study identifies that the interaction loop between CREB1 and miR-181a-5p plays a crucial role in regulating the pathophysiologic features of FD BMSCs. MiR-181a-5p may be a potential therapeutic target for the treatment of FD.
Collapse
Affiliation(s)
- Yu Fu
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, No.136, Hanzhong Road, Nanjing, 210029, Jiangsu Province, China. .,Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, 210029, China. .,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, 210029, China.
| | - Zhili Xin
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, No.136, Hanzhong Road, Nanjing, 210029, Jiangsu Province, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Ziji Ling
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, No.136, Hanzhong Road, Nanjing, 210029, Jiangsu Province, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Hanyu Xie
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, No.136, Hanzhong Road, Nanjing, 210029, Jiangsu Province, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Tao Xiao
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, No.136, Hanzhong Road, Nanjing, 210029, Jiangsu Province, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, 210029, China.,Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, 210008, China
| | - Xin Shen
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, No.136, Hanzhong Road, Nanjing, 210029, Jiangsu Province, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Jialin Lin
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, No.136, Hanzhong Road, Nanjing, 210029, Jiangsu Province, China
| | - Ling Xu
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, No.136, Hanzhong Road, Nanjing, 210029, Jiangsu Province, China
| | - Hongbing Jiang
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, No.136, Hanzhong Road, Nanjing, 210029, Jiangsu Province, China. .,Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, 210029, China. .,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
8
|
Steven A, Friedrich M, Jank P, Heimer N, Budczies J, Denkert C, Seliger B. What turns CREB on? And off? And why does it matter? Cell Mol Life Sci 2020; 77:4049-4067. [PMID: 32347317 PMCID: PMC7532970 DOI: 10.1007/s00018-020-03525-8] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 03/21/2020] [Accepted: 04/06/2020] [Indexed: 12/16/2022]
Abstract
Altered expression and function of the transcription factor cyclic AMP response-binding protein (CREB) has been identified to play an important role in cancer and is associated with the overall survival and therapy response of tumor patients. This review focuses on the expression and activation of CREB under physiologic conditions and in tumors of distinct origin as well as the underlying mechanisms of CREB regulation by diverse stimuli and inhibitors. In addition, the clinical relevance of CREB is summarized, including its use as a prognostic and/or predictive marker as well as a therapeutic target.
Collapse
Affiliation(s)
- André Steven
- Institute for Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany
| | - Michael Friedrich
- Institute for Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany
| | - Paul Jank
- Institute of Pathology, Philipps University Marburg, 35043, Marburg, Germany
| | - Nadine Heimer
- Institute for Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany
| | - Jan Budczies
- Institute of Pathology, University Clinic Heidelberg, 69120, Heidelberg, Germany
| | - Carsten Denkert
- Institute of Pathology, Philipps University Marburg, 35043, Marburg, Germany
| | - Barbara Seliger
- Institute for Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany.
| |
Collapse
|
9
|
Astrocyte-Derived Small Extracellular Vesicles Regulate Dendritic Complexity through miR-26a-5p Activity. Cells 2020; 9:cells9040930. [PMID: 32290095 PMCID: PMC7226994 DOI: 10.3390/cells9040930] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 04/02/2020] [Accepted: 04/05/2020] [Indexed: 02/07/2023] Open
Abstract
In the last few decades, it has been established that astrocytes play key roles in the regulation of neuronal morphology. However, the contribution of astrocyte-derived small extracellular vesicles (sEVs) to morphological differentiation of neurons has only recently been addressed. Here, we showed that cultured astrocytes expressing a GFP-tagged version of the stress-regulated astrocytic enzyme Aldolase C (Aldo C-GFP) release small extracellular vesicles (sEVs) that are transferred into cultured hippocampal neurons. Surprisingly, Aldo C-GFP-containing sEVs (Aldo C-GFP sEVs) displayed an exacerbated capacity to reduce the dendritic complexity in developing hippocampal neurons compared to sEVs derived from control (i.e., GFP-expressing) astrocytes. Using bioinformatics and biochemical tools, we found that the total content of overexpressed Aldo C-GFP correlates with an increased content of endogenous miRNA-26a-5p in both total astrocyte homogenates and sEVs. Notably, neurons magnetofected with a nucleotide sequence that mimics endogenous miRNA-26a-5p (mimic 26a-5p) not only decreased the levels of neuronal proteins associated to morphogenesis regulation, but also reproduced morphological changes induced by Aldo-C-GFP sEVs. Furthermore, neurons magnetofected with a sequence targeting miRNA-26a-5p (antago 26a-5p) were largely resistant to Aldo C-GFP sEVs. Our results support a novel and complex level of astrocyte-to-neuron communication mediated by astrocyte-derived sEVs and the activity of their miRNA content.
Collapse
|
10
|
Rodriguez‐Ortiz CJ, Prieto GA, Martini AC, Forner S, Trujillo‐Estrada L, LaFerla FM, Baglietto‐Vargas D, Cotman CW, Kitazawa M. miR-181a negatively modulates synaptic plasticity in hippocampal cultures and its inhibition rescues memory deficits in a mouse model of Alzheimer's disease. Aging Cell 2020; 19:e13118. [PMID: 32087004 PMCID: PMC7059142 DOI: 10.1111/acel.13118] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 11/21/2019] [Accepted: 12/23/2019] [Indexed: 12/11/2022] Open
Abstract
MicroRNAs play a pivotal role in rapid, dynamic, and spatiotemporal modulation of synaptic functions. Among them, recent emerging evidence highlights that microRNA-181a (miR-181a) is particularly abundant in hippocampal neurons and controls the expression of key plasticity-related proteins at synapses. We have previously demonstrated that miR-181a was upregulated in the hippocampus of a mouse model of Alzheimer's disease (AD) and correlated with reduced levels of plasticity-related proteins. Here, we further investigated the underlying mechanisms by which miR-181a negatively modulated synaptic plasticity and memory. In primary hippocampal cultures, we found that an activity-dependent upregulation of the microRNA-regulating protein, translin, correlated with reduction of miR-181a upon chemical long-term potentiation (cLTP), which induced upregulation of GluA2, a predicted target for miR-181a, and other plasticity-related proteins. Additionally, Aβ treatment inhibited cLTP-dependent induction of translin and subsequent reduction of miR-181a, and cotreatment with miR-181a antagomir effectively reversed the effects elicited by Aβ but did not rescue translin levels, suggesting that the activity-dependent upregulation of translin was upstream of miR-181a. In mice, a learning episode markedly decreased miR-181a in the hippocampus and raised the protein levels of GluA2. Lastly, we observed that inhibition of miR-181a alleviated memory deficits and increased GluA2 and GluA1 levels, without restoring translin, in the 3xTg-AD model. Taken together, our results indicate that miR-181a is a major negative regulator of the cellular events that underlie synaptic plasticity and memory through AMPA receptors, and importantly, Aβ disrupts this process by suppressing translin and leads to synaptic dysfunction and memory impairments in AD.
Collapse
Affiliation(s)
| | - Gilberto Aleph Prieto
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCalifornia
- Departamento de Neurobiología Celular y MolecularInstituto de NeurobiologíaUniversidad Nacional Autonoma de MéxicoQuerétaroMexico
| | - Alessandra C. Martini
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCalifornia
| | - Stefania Forner
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCalifornia
| | - Laura Trujillo‐Estrada
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCalifornia
| | - Frank M. LaFerla
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCalifornia
| | - David Baglietto‐Vargas
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCalifornia
| | - Carl W. Cotman
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCalifornia
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCAUSA
| | | |
Collapse
|
11
|
Inhibition of miR-181a promotes midbrain neuronal growth through a Smad1/5-dependent mechanism: implications for Parkinson's disease. Neuronal Signal 2018; 2:NS20170181. [PMID: 32714583 PMCID: PMC7371012 DOI: 10.1042/ns20170181] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 12/22/2017] [Accepted: 01/10/2018] [Indexed: 12/14/2022] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease, and is characterized by the progressive degeneration of nigrostriatal dopaminergic (DA) neurons. Current PD treatments are symptomatic, wear off over time and do not protect against DA neuronal loss. Finding a way to re-grow midbrain DA (mDA) neurons is a promising disease-modifying therapeutic strategy for PD. However, reliable biomarkers are required to allow such growth-promoting approaches to be applied early in the disease progression. miR-181a has been shown to be dysregulated in PD patients, and has been identified as a potential biomarker for PD. Despite studies demonstrating the enrichment of miR-181a in the brain, specifically in neurites of postmitotic neurons, the role of miR-181a in mDA neurons remains unknown. Herein, we used cell culture models of human mDA neurons to investigate a potential role for miR-181a in mDA neurons. We used a bioninformatics analysis to identify that miR-181a targets components of the bone morphogenetic protein (BMP) signalling pathway, including the transcription factors Smad1 and Smad5, which we find are expressed by rat mDA neurons and are required for BMP-induced neurite growth. We also found that inhibition of neuronal miR-181a, resulted in increased Smad signalling, and induced neurite growth in SH-SY5Y cells. Finally, using embryonic rat cultures, we demonstrated that miR-181a inhibition induces ventral midbrain (VM) and cortical neuronal growth. These data describe a new role for miR-181a in mDA neurons, and provide proof of principle that miR-181a dysresgulation in PD may alter the activation state of signalling pathways important for neuronal growth in neurons affected in PD.
Collapse
|
12
|
Steven A, Seliger B. Control of CREB expression in tumors: from molecular mechanisms and signal transduction pathways to therapeutic target. Oncotarget 2018; 7:35454-65. [PMID: 26934558 PMCID: PMC5085243 DOI: 10.18632/oncotarget.7721] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 01/26/2016] [Indexed: 12/11/2022] Open
Abstract
The cyclic AMP response element binding (CREB) protein has pleiotropic activities in physiologic processes. Due to its central position downstream of many growth signaling pathways CREB has the ability to influence cell survival, growth and differentiation of normal, but also of tumor cells suggesting an oncogenic potential of CREB. Indeed, increased CREB expression and activation is associated with tumor progression, chemotherapy resistance and reduced patients' survival. We summarize here the different cellular functions of CREB in tumors of distinct histology as well as its use as potential prognostic marker. In addition, the underlying molecular mechanisms to achieve constitutive activation of CREB including structural alterations, such as gene amplification and chromosomal translocation, and deregulation, which could occur at the transcriptional, post-transcriptional and post-translational level, will be described. Since downregulation of CREB by different strategies resulted in inhibition of cell proliferation, invasion and induction of apoptosis, the role of CREB as a promising target for cancer therapy will be also discussed.
Collapse
Affiliation(s)
- André Steven
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Barbara Seliger
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| |
Collapse
|
13
|
Du X, Yang Y, Xu C, Peng Z, Zhang M, Lei L, Gao W, Dong Y, Shi Z, Sun X, Wang Z, Li X, Li X, Liu G. Upregulation of miR-181a impairs hepatic glucose and lipid homeostasis. Oncotarget 2017; 8:91362-91378. [PMID: 29207650 PMCID: PMC5710930 DOI: 10.18632/oncotarget.20523] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 08/06/2017] [Indexed: 01/20/2023] Open
Abstract
The contributions of altered post-transcriptional gene silencing to the development of metabolic disorders remain poorly understood thus far. The objective of this study was to evaluate the roles of miR-181a in the regulation of hepatic glucose and lipid metabolism. MiR-181a is abundantly expressed in the liver, and we found that blood and hepatic miR-181a levels were significantly increased in patients and dairy cows with non-alcoholic fatty liver disease, as well as in high-fat diet and ob/ob mice. We determined that sirtuin1 is a target of miR-181a. Moreover, we found that hepatic sirtuin1 and peroxisome proliferator-activated receptor-γ coactivator-1α expression levels are downregulated, and acetylated peroxisome proliferator-activated receptor-γ coactivator-1α expression levels are upregulated in patients and dairy cows with non-alcoholic fatty liver disease, as well as in high-fat diet and ob/ob mice. MiR-181a overexpression inhibits the sirtuin1-peroxisome proliferator-activated receptor-γ coactivator-1α pathway, reduces insulin sensitivity, and increases gluconeogenesis and lipid synthesis in dairy cow hepatocytes and HepG2 cells. Conversely, silencing of miR-181a over-activates the sirtuin1-peroxisome proliferator-activated receptor-γ coactivator-1α pathway, increases insulin sensitivity and glycogen content, and decreases gluconeogenesis and lipid synthesis in hepatocytes, even under non-esterified fatty acids treatment conditions. Furthermore, miR-181a overexpression or sirtuin1 knockdown in mice increases lipid accumulation and decreases insulin sensitivity and glycogen content in the liver. Taken together, these findings indicate that increased hepatic miR-181a impairs glucose and lipid homeostasis by silencing sirtuin1 in non-alcoholic fatty liver disease.
Collapse
Affiliation(s)
- Xiliang Du
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yuchen Yang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Chuang Xu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Zhicheng Peng
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Min Zhang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Lin Lei
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Wenwen Gao
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yuhao Dong
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Zhen Shi
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Xudong Sun
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Zhe Wang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Xiaobing Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Xinwei Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Guowen Liu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| |
Collapse
|
14
|
Zhang SF, Chen JC, Zhang J, Xu JG. miR-181a involves in the hippocampus-dependent memory formation via targeting PRKAA1. Sci Rep 2017; 7:8480. [PMID: 28814760 PMCID: PMC5559581 DOI: 10.1038/s41598-017-09095-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 07/20/2017] [Indexed: 02/05/2023] Open
Abstract
Post-transcriptional gene regulation by microRNAs (miRNAs) is involved in memory formation. However, the roles of individual miRNAs in these processes remain largely unknown. In this study, we want to clarify the role of miR-181a in hippocampus-dependent memory formation. A transient increase in miR-181a expression was observed after conditioned fear conditioning (CFC) and object location task (OLT) training. Selective overexpression or inhibition of miR-181a in the dorsal hippocampus (DH) via the injection of a miR-181a agomir or antagomir enhanced or impaired the CFC- and OLT-dependent memory formation, respectively. Using bioinformatics and luciferase assays, we identified PRKAA1 as a potential target gene of miR-181a. After CFC or OLT training, the expression and activity of PRKAA1 decreased as miR-181a expression increased and was effectively blocked by the miR-181a antagomir. Moreover, microinjection of the PRKAA1 agonist AICAR or inhibitor compound C in the DH reversed the roles of the miR-181a agomir or antagomir in CFC- and OLT-dependent memory formation. In conclusion, this work provides novel evidence describing the role and mechanism of miR-181a in hippocampus-dependent memory formation, which sheds light on the potential regulation of cognition and future treatments for cognitive disorders.
Collapse
Affiliation(s)
- Sun-Fu Zhang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, P. R. China.,Department of Neurosurgery, The First People's Hospital of Yibin, Yibin, Sichuan, P. R. China
| | - Jun-Chen Chen
- Department of Neurosurgery, Sichuan 81 Rehabilitation Center, Chengdu, Sichuan, P. R. China
| | - Jing Zhang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, P. R. China
| | - Jian-Guo Xu
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, P. R. China.
| |
Collapse
|
15
|
Inhibition of miR-181a protects female mice from transient focal cerebral ischemia by targeting astrocyte estrogen receptor-α. Mol Cell Neurosci 2017; 82:118-125. [PMID: 28522364 DOI: 10.1016/j.mcn.2017.05.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 05/03/2017] [Accepted: 05/08/2017] [Indexed: 02/03/2023] Open
Abstract
Whether the effect of miR-181a is sexually dimorphic in stroke is unknown. Prior work showed protection of male mice with miR-181a inhibition. Estrogen receptor-α (ERα) is an identified target of miR181 in endometrium. Therefore we investigated the separate and joint effects of miR-181a inhibition and 17β-estradiol (E2) replacement after ovariectomy. Adult female mice were ovariectomized and implanted with an E2- or vehicle-containing capsule for 14d prior to 1h middle cerebral artery occlusion (MCAO). Each group received either miR-181a antagomir or mismatch control by intracerebroventricular injection 24h before MCAO. After MCAO neurologic deficit and infarct volume were assessed. Primary male and female astrocyte cultures were subjected to glucose deprivation with miR-181a inhibitor or transfection control, and E2 or vehicle control, with/without ESRα knockdown with small interfering RNA. Cell death was assessed by propidium iodide staining, and lactate dehydrogenase assay. A miR-181a/ERα target site blocker (TSB), with/without miR-181a mimic, was used to confirm targeting of ERα by miR-181a in astrocytes. Individually, miR-181a inhibition or E2 decreased infarct volume and improved neurologic score in female mice, and protected male and female astrocyte cultures. Combined miR-181a inhibition plus E2 afforded greater protection of female mice and female astrocyte cultures, but not in male astrocyte cultures. MiR-181a inhibition only increased ERα levels in vivo and in female cultures, while ERα knockdown with siRNA increased cell death in both sexes. Treatment with ERα TSB was strongly protective in both sexes. In conclusion, the results of the present study suggest miR-181a inhibition enhances E2-mediated stroke protection in females in part by augmenting ERα production, a mechanism detected in female mice and female astrocytes. Sex differences were observed with combined miR-181a inhibition/E2 treatment, and miR-181a targeting of ERα.
Collapse
|
16
|
Wingo AP, Almli LM, Stevens JS, Jovanovic T, Wingo TS, Tharp G, Li Y, Lori A, Briscione M, Jin P, Binder EB, Bradley B, Gibson G, Ressler KJ. Genome-wide association study of positive emotion identifies a genetic variant and a role for microRNAs. Mol Psychiatry 2017; 22:774-783. [PMID: 27595594 PMCID: PMC5339071 DOI: 10.1038/mp.2016.143] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 05/30/2016] [Accepted: 07/01/2016] [Indexed: 01/14/2023]
Abstract
Positive affect denotes a state of pleasurable engagement with the environment eliciting positive emotion such as contentment, enthusiasm or happiness. Positive affect is associated with favorable psychological, physical and economic outcomes in many longitudinal studies. With a heritability of ⩽64%, positive affect is substantially influenced by genetic factors; however, our understanding of genetic pathways underlying individual differences in positive affect is still limited. Here, through a genome-wide association study of positive affect in African-American participants, we identify a single-nucleotide polymorphism, rs322931, significantly associated with positive affect at P<5 × 10-8, and replicate this association in another cohort. Furthermore, we show that the minor allele of rs322931 predicts expression of microRNAs miR-181a and miR-181b in human brain and blood, greater nucleus accumbens reactivity to positive emotional stimuli and enhanced fear inhibition. Prior studies have suggested that miR-181a is part of the reward neurocircuitry. Taken together, we identify a novel genetic variant for further elucidation of genetic underpinning of positive affect that mediates positive emotionality potentially via the nucleus accumbens and miR-181.
Collapse
Affiliation(s)
- Aliza P. Wingo
- Atlanta VAMC, Atlanta, GA, USA
- Emory University, Department of Psychiatry, School of Medicine, Atlanta, GA, USA
| | - Lynn M. Almli
- Emory University, Department of Psychiatry, School of Medicine, Atlanta, GA, USA
| | - Jennifer S. Stevens
- Emory University, Department of Psychiatry, School of Medicine, Atlanta, GA, USA
| | - Tanja Jovanovic
- Emory University, Department of Psychiatry, School of Medicine, Atlanta, GA, USA
| | - Thomas S. Wingo
- Atlanta VAMC, Atlanta, GA, USA
- Emory University, Department of Neurology, School of Medicine, Atlanta, GA, USA
- Department of Human Genetics, School of Medicine, Emory University, Atlanta, GA, USA
| | - Gregory Tharp
- Yerkes Research Center, Emory University, Atlanta, GA, USA
| | - Yujing Li
- Department of Human Genetics, School of Medicine, Emory University, Atlanta, GA, USA
| | - Adriana Lori
- Emory University, Department of Psychiatry, School of Medicine, Atlanta, GA, USA
| | - Maria Briscione
- Atlanta VAMC, Atlanta, GA, USA
- Emory University, Department of Psychiatry, School of Medicine, Atlanta, GA, USA
| | - Peng Jin
- Department of Human Genetics, School of Medicine, Emory University, Atlanta, GA, USA
| | - Elisabeth B. Binder
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| | - Bekh Bradley
- Atlanta VAMC, Atlanta, GA, USA
- Emory University, Department of Psychiatry, School of Medicine, Atlanta, GA, USA
| | - Greg Gibson
- Center for Integrative Genomics, School of Biology, Georgia Institute of Technology, Atlanta, GA, USA
| | - Kerry J. Ressler
- Emory University, Department of Psychiatry, School of Medicine, Atlanta, GA, USA
- McLean Hospital, Harvard Medical School, Belmont, Massachusetts
| |
Collapse
|
17
|
Deng M, Tufan T, Raza MU, Jones TC, Zhu MY. MicroRNAs 29b and 181a down-regulate the expression of the norepinephrine transporter and glucocorticoid receptors in PC12 cells. J Neurochem 2016; 139:197-207. [PMID: 27501468 DOI: 10.1111/jnc.13761] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 07/25/2016] [Accepted: 08/01/2016] [Indexed: 12/13/2022]
Abstract
MicroRNAs are short non-coding RNAs that provide global regulation of gene expression at the post-transcriptional level. Such regulation has been found to play a role in stress-induced epigenetic responses in the brain. The norepinephrine transporter (NET) and glucocorticoid receptors are closely related to the homeostatic integration and regulation after stress. Our previous studies demonstrated that NET mRNA and protein levels in rats are regulated by chronic stress and by administration of corticosterone, which is mediated through glucocorticoid receptors. Whether miRNAs are intermediaries in the regulation of these proteins remains to be elucidated. This study was undertaken to determine possible regulatory effects of miRNAs on the expression of NET and glucocorticoid receptors in the noradrenergic neuronal cell line. Using computational target prediction, we identified several candidate miRNAs potentially targeting NET and glucocorticoid receptors. Western blot results showed that over-expression of miR-181a and miR-29b significantly repressed protein levels of NET, which is accompanied by a reduced [3 H] norepinephrine uptake, and glucocorticoid receptors in PC12 cells. Luciferase reporter assays verified that both miR-181a and miR-29b bind the 3'UTR of mRNA of NET and glucocorticoid receptors. Furthermore, exposure of PC12 cells to corticosterone markedly reduced the endogenous levels of miR-29b, which was not reversed by the application of glucocorticoid receptor antagonist mifepristone. These observations indicate that miR-181a and miR-29b can function as the negative regulators of NET and glucocorticoid receptor translation in vitro. This regulatory effect may be related to stress-induced up-regulation of the noradrenergic phenotype, a phenomenon observed in stress models and depressive patients. This study demonstrated that miR-29b and miR-181a, two short non-coding RNAs that provide global regulation of gene expression, markedly repressed protein levels of norepinephrine (NE) transporter and glucocorticoid receptor (GR), as well as NE uptake by binding the 3'UTR of their mRNAs in PC12 cells. Also, exposure of cells to corticosterone significantly reduced miR-29b levels through a GR-independent way.
Collapse
Affiliation(s)
- Maoxian Deng
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA.,Department of Veterinary Medicine and Animal Husbandry, Jiangsu Polytechnic College of A&F, Jurong, Jiangsu, China
| | - Turan Tufan
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| | - Muhammad U Raza
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| | - Thomas C Jones
- Department of Biological Sciences, College of Arts and Sciences, East Tennessee State University, Johnson City, Tennessee, USA
| | - Meng-Yang Zhu
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA.
| |
Collapse
|
18
|
Martin E, Qureshi A, Dasa V, Freitas M, Gimble J, Davis T. MicroRNA regulation of stem cell differentiation and diseases of the bone and adipose tissue: Perspectives on miRNA biogenesis and cellular transcriptome. Biochimie 2016; 124:98-111. [DOI: 10.1016/j.biochi.2015.02.012] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 02/17/2015] [Indexed: 12/19/2022]
|
19
|
Lian S, Guo JR, Nan XM, Ma L, Loor JJ, Bu DP. MicroRNA Bta-miR-181a regulates the biosynthesis of bovine milk fat by targeting ACSL1. J Dairy Sci 2016; 99:3916-3924. [PMID: 26971144 DOI: 10.3168/jds.2015-10484] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 01/21/2016] [Indexed: 01/19/2023]
Abstract
MicroRNA (miRNA) are a class of small noncoding RNA that function as important posttranscriptional regulators of gene expression. The acyl-CoA synthetase long-chain family member 1 (ACSL1) is an important enzyme in the process of milk lipid synthesis. In a previous study dealing with incubations of stearic acid in bovine mammary epithelial cells, an opposite expression pattern was observed between ACSL1 and miR-181a. Bioinformatics analysis with TargetScan and PicTar revealed ACSL1 as a potential target gene of miR-181a. The objective of this work was to determine the potential function of miR-181a on milk fat synthesis by defining the regulatory relationship between miR-181a and ACSL1. Primary bovine mammary epithelial cells were harvested from mid-lactation cows and cultured in Dulbecco's modified Eagle's medium/F-12 medium with 10% fetal bovine serum, 0.5μg/mL of insulin, 10 ng/mL of epidermal growth factor, 5μg/mL of transferrin, 1μg/mL of hydrocortisone, 1μg/mL of progesterone, 5μg/mL of estradiol, and 5μg/mL of prolactin. Cells were transfected with an miR-181a mimic to increase its expression and an miR-181a inhibitor to decrease its expression before culturing for 48 h. The results revealed that the overexpression of miR-181a inhibited the expression of ACSL1, whereas the downregulation of miR-181a increased ACSL1 expression. Western blot analysis of ACSL1 revealed similar effects. Oil-red-O staining indicated that cellular lipid droplet synthesis was decreased with the overexpression of bta-miR-181a, and treatment with the bta-miR-181a inhibitor increased concentration of lipid droplets. Furthermore, overexpression of bta-miR-181a resulted in a decrease in concentration of triacylglycerol in the cells, whereas inhibition of bta-miR-181a increased concentration of triacylglycerol. Therefore, the results indicated that bta-miR-181a may contribute to negative regulation of lipid synthesis in mammary cells via targeting ACSL1.
Collapse
Affiliation(s)
- S Lian
- Institute of Animal Science, State Key Laboratory of Animal Nutrition, Chinese Academy of Agricultural Sciences, Beijing, 100193, P. R. China; College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - J R Guo
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - X M Nan
- Institute of Animal Science, State Key Laboratory of Animal Nutrition, Chinese Academy of Agricultural Sciences, Beijing, 100193, P. R. China
| | - L Ma
- Institute of Animal Science, State Key Laboratory of Animal Nutrition, Chinese Academy of Agricultural Sciences, Beijing, 100193, P. R. China
| | - J J Loor
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - D P Bu
- Institute of Animal Science, State Key Laboratory of Animal Nutrition, Chinese Academy of Agricultural Sciences, Beijing, 100193, P. R. China; CAAS-ICRAF Joint Laboratory on Agroforestry and Sustainable Animal Husbandry, World Agroforestry Centre, East and Central Asia, Beijing 100193, China; Synergetic Innovation Center of Food Safety and Nutrition, Harbin, 150030, China.
| |
Collapse
|
20
|
Guibinga GH. MicroRNAs: tools of mechanistic insights and biological therapeutics discovery for the rare neurogenetic syndrome Lesch-Nyhan disease (LND). ADVANCES IN GENETICS 2015; 90:103-131. [PMID: 26296934 DOI: 10.1016/bs.adgen.2015.06.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
MicroRNAs (miRNAs) are small regulatory RNAs that modulate the translation of mRNA. They have emerged over the past few years as indispensable entities in the transcriptional regulation of genes. Their discovery has added additional layers of complexity to regulatory networks that control cellular homeostasis. Also, their dysregulated pattern of expression is now well demonstrated in myriad diseases and pathogenic processes. In the current review, we highlight the role of miRNAs in Lesch-Nyhan disease (LND), a rare neurogenetic syndrome caused by mutations in the purine metabolic gene encoding the hypoxanthine-guanine phosphoribosyltransferase (HPRT) enzyme. We describe how experimental and biocomputational approaches have helped to unravel genetic and signaling pathways that provide mechanistic understanding of some of the molecular and cellular basis of this ill-defined neurogenetic disorder. Through miRNA-based target predictions, we have identified signaling pathways that may be of significance in guiding biological therapeutic discovery for this incurable neurological disorder. We also propose a model to explain how a gene such as HPRT, mostly known for its housekeeping metabolic functions, can have pleiotropic effects on disparate genes and signal transduction pathways. Our hypothetical model suggests that HPRT mRNA transcripts may be acting as competitive endogenous RNAs (ceRNAs) intertwined in multiregulatory cross talk between key neural transcripts and miRNAs. Overall, this approach of using miRNA-based genomic approaches to elucidate the molecular and cellular basis of LND and guide biological target identification might be applicable to other ill-defined rare inborn-error metabolic diseases.
Collapse
Affiliation(s)
- Ghiabe-Henri Guibinga
- Division of Genetics, Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
21
|
Störchel PH, Thümmler J, Siegel G, Aksoy-Aksel A, Zampa F, Sumer S, Schratt G. A large-scale functional screen identifies Nova1 and Ncoa3 as regulators of neuronal miRNA function. EMBO J 2015; 34:2237-54. [PMID: 26105073 DOI: 10.15252/embj.201490643] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 05/20/2015] [Indexed: 01/12/2023] Open
Abstract
MicroRNAs (miRNAs) are important regulators of neuronal development, network connectivity, and synaptic plasticity. While many neuronal miRNAs were previously shown to modulate neuronal morphogenesis, little is known regarding the regulation of miRNA function. In a large-scale functional screen, we identified two novel regulators of neuronal miRNA function, Nova1 and Ncoa3. Both proteins are expressed in the nucleus and the cytoplasm of developing hippocampal neurons. We found that Nova1 and Ncoa3 stimulate miRNA function by different mechanisms that converge on Argonaute (Ago) proteins, core components of the miRNA-induced silencing complex (miRISC). While Nova1 physically interacts with Ago proteins, Ncoa3 selectively promotes the expression of Ago2 at the transcriptional level. We further show that Ncoa3 regulates dendritic complexity and dendritic spine maturation of hippocampal neurons in a miRNA-dependent fashion. Importantly, both the loss of miRNA activity and increased dendrite complexity upon Ncoa3 knockdown were rescued by Ago2 overexpression. Together, we uncovered two novel factors that control neuronal miRISC function at the level of Ago proteins, with possible implications for the regulation of synapse development and plasticity.
Collapse
Affiliation(s)
- Peter H Störchel
- Institute for Physiological Chemistry Biochemical-Pharmacological Center Marburg Philipps-University Marburg, Marburg, Germany
| | - Juliane Thümmler
- Institute for Physiological Chemistry Biochemical-Pharmacological Center Marburg Philipps-University Marburg, Marburg, Germany
| | - Gabriele Siegel
- Institute for Physiological Chemistry Biochemical-Pharmacological Center Marburg Philipps-University Marburg, Marburg, Germany
| | - Ayla Aksoy-Aksel
- Institute for Physiological Chemistry Biochemical-Pharmacological Center Marburg Philipps-University Marburg, Marburg, Germany
| | - Federico Zampa
- Institute for Physiological Chemistry Biochemical-Pharmacological Center Marburg Philipps-University Marburg, Marburg, Germany
| | - Simon Sumer
- Institute for Physiological Chemistry Biochemical-Pharmacological Center Marburg Philipps-University Marburg, Marburg, Germany
| | - Gerhard Schratt
- Institute for Physiological Chemistry Biochemical-Pharmacological Center Marburg Philipps-University Marburg, Marburg, Germany
| |
Collapse
|
22
|
Modulation of hippocampal neural plasticity by glucose-related signaling. Neural Plast 2015; 2015:657928. [PMID: 25977822 PMCID: PMC4419237 DOI: 10.1155/2015/657928] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 04/02/2015] [Accepted: 04/05/2015] [Indexed: 12/20/2022] Open
Abstract
Hormones and peptides involved in glucose homeostasis are emerging as important modulators of neural plasticity. In this regard, increasing evidence shows that molecules such as insulin, insulin-like growth factor-I, glucagon-like peptide-1, and ghrelin impact on the function of the hippocampus, which is a key area for learning and memory. Indeed, all these factors affect fundamental hippocampal properties including synaptic plasticity (i.e., synapse potentiation and depression), structural plasticity (i.e., dynamics of dendritic spines), and adult neurogenesis, thus leading to modifications in cognitive performance. Here, we review the main mechanisms underlying the effects of glucose metabolism on hippocampal physiology. In particular, we discuss the role of these signals in the modulation of cognitive functions and their potential implications in dysmetabolism-related cognitive decline.
Collapse
|
23
|
Kempf SJ, Moertl S, Sepe S, von Toerne C, Hauck SM, Atkinson MJ, Mastroberardino PG, Tapio S. Low-dose ionizing radiation rapidly affects mitochondrial and synaptic signaling pathways in murine hippocampus and cortex. J Proteome Res 2015; 14:2055-64. [PMID: 25807253 DOI: 10.1021/acs.jproteome.5b00114] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The increased use of radiation-based medical imaging methods such as computer tomography is a matter of concern due to potential radiation-induced adverse effects. Efficient protection against such detrimental effects has not been possible due to inadequate understanding of radiation-induced alterations in signaling pathways. The aim of this study was to elucidate the molecular mechanisms behind learning and memory deficits after acute low and moderate doses of ionizing radiation. Female C57BL/6J mice were irradiated on postnatal day 10 (PND10) with gamma doses of 0.1 or 0.5 Gy. This was followed by evaluation of the cellular proteome, pathway-focused transcriptome, and neurological development/disease-focused miRNAome of hippocampus and cortex 24 h postirradiation. Our analysis showed that signaling pathways related to mitochondrial and synaptic functions were changed by acute irradiation. This may lead to reduced mitochondrial function paralleled by enhanced number of dendritic spines and neurite outgrowth due to elevated long-term potentiation, triggered by increased phosphorylated CREB. This was predominately observed in the cortex at 0.1 and 0.5 Gy and in the hippocampus only at 0.5 Gy. Moreover, a radiation-induced increase in the expression of several neural miRNAs associated with synaptic plasticity was found. The early changes in signaling pathways related to memory formation may be associated with the acute neurocognitive side effects in patients after brain radiotherapy but might also contribute to late radiation-induced cognitive injury.
Collapse
Affiliation(s)
- Stefan J Kempf
- †Institute of Radiation Biology, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Ingolstaedter Landstrasse 1, 85764 Neuherberg, Germany
| | - Simone Moertl
- †Institute of Radiation Biology, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Ingolstaedter Landstrasse 1, 85764 Neuherberg, Germany
| | - Sara Sepe
- ‡Department of Genetics, Erasmus Medical Center, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
| | - Christine von Toerne
- §Research Unit Protein Science, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Ingolstaedter Landstrasse 1, 85764 Neuherberg, Germany
| | - Stefanie M Hauck
- §Research Unit Protein Science, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Ingolstaedter Landstrasse 1, 85764 Neuherberg, Germany
| | - Michael J Atkinson
- †Institute of Radiation Biology, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Ingolstaedter Landstrasse 1, 85764 Neuherberg, Germany.,∥Chair of Radiation Biology, Technical University Munich, Arcisstrasse 21, 80333 Munich, Germany
| | - Pier G Mastroberardino
- ‡Department of Genetics, Erasmus Medical Center, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
| | - Soile Tapio
- †Institute of Radiation Biology, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Ingolstaedter Landstrasse 1, 85764 Neuherberg, Germany
| |
Collapse
|
24
|
Giusti SA, Vogl AM, Brockmann MM, Vercelli CA, Rein ML, Trümbach D, Wurst W, Cazalla D, Stein V, Deussing JM, Refojo D. MicroRNA-9 controls dendritic development by targeting REST. eLife 2014; 3. [PMID: 25406064 PMCID: PMC4235007 DOI: 10.7554/elife.02755] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 10/15/2014] [Indexed: 12/14/2022] Open
Abstract
MicroRNAs (miRNAs) are conserved noncoding RNAs that function as posttranscriptional regulators of gene expression. miR-9 is one of the most abundant miRNAs in the brain. Although the function of miR-9 has been well characterized in neural progenitors, its role in dendritic and synaptic development remains largely unknown. In order to target miR-9 in vivo, we developed a transgenic miRNA sponge mouse line allowing conditional inactivation of the miR-9 family in a spatio-temporal-controlled manner. Using this novel approach, we found that miR-9 controls dendritic growth and synaptic transmission in vivo. Furthermore, we demonstrate that miR-9-mediated downregulation of the transcriptional repressor REST is essential for proper dendritic growth. DOI:http://dx.doi.org/10.7554/eLife.02755.001 Messages are sent back and forth in our brains by cells called neurons that connect to each other in complex networks. Neurons develop from stem cells in a complicated process that involves a number of different stages. In one of the final stages, tree-like structures called dendrites emerge from the neurons and connect with neighboring neurons via special junctions called synapses. A group of small RNA molecules called microRNAs have roles in controlling the development of neurons. One microRNA, called miR-9, is abundant in the brain and is known to be involved in the early stages of neuron development. However, its role in the formation of dendrites and synapses remains unclear. Giusti et al. studied this microRNA in mice. A length of DNA, coding for an RNA molecule that binds to miR-9 molecules and stops them performing their normal function, was inserted into the mice. These experiments showed that miR-9 is involved in controlling dendrite growth and synaptic function. To enable a neuron to produce dendrites, miR-9 binds to and interferes with the RNA molecules that are needed to make a protein called REST. This protein is a transcription factor that switches off the expression of other genes so, in effect, miR-9 allows a set of genes that are needed for dendrite growth to be switched on. The methodology developed by Giusti et al. could be used to study the functions of other microRNAs. DOI:http://dx.doi.org/10.7554/eLife.02755.002
Collapse
Affiliation(s)
- Sebastian A Giusti
- Department of Molecular Neurobiology, Max Planck Institute of Psychiatry, Munich, Germany
| | - Annette M Vogl
- Department of Molecular Neurobiology, Max Planck Institute of Psychiatry, Munich, Germany
| | - Marisa M Brockmann
- Department of Molecular Neurobiology, Max Planck Institute of Psychiatry, Munich, Germany
| | - Claudia A Vercelli
- Department of Molecular Neurobiology, Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Martin L Rein
- Department of Neurobiology of Stress and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Dietrich Trümbach
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Demian Cazalla
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, United States
| | - Valentin Stein
- Institute of Physiology, University of Bonn, Bonn, Germany
| | - Jan M Deussing
- Department of Neurobiology of Stress and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Damian Refojo
- Department of Molecular Neurobiology, Max Planck Institute of Psychiatry, Munich, Germany
| |
Collapse
|
25
|
Goto G, Hori Y, Ishikawa M, Tanaka S, Sakamoto A. Changes in the gene expression levels of microRNAs in the rat hippocampus by sevoflurane and propofol anesthesia. Mol Med Rep 2014; 9:1715-22. [PMID: 24626427 DOI: 10.3892/mmr.2014.2038] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 02/13/2014] [Indexed: 11/05/2022] Open
Abstract
General anesthesia is commonly used in the surgical arena, but little is known regarding its influence at the genomic and molecular levels. MicroRNAs (miRNAs) belong to a new class of non-coding RNA molecules which influence cell biology. In the present study, it was hypothesized that miRNAs alter gene expression levels under general anesthesia. The aim was to compare the miRNA expression profiles in the rat hippocampus in response to anesthesia with representative volatile (sevoflurane) and intravenous (propofol) anesthetics. Wistar Rats were randomly assigned to either a 2.4% sevoflurane, 600 µg/kg/min propofol or control (without anesthetics) group. Total RNA from hippocampal samples which contained miRNA was subjected to quantitative reverse transcription-polymerase chain reaction and Taqman Low-Density Arrays (TLDA). A total of 373 miRNAs are associated with rats and the TLDA analysis revealed that 279 expressed miRNAs (74.8%) were expressed in all three groups. Significant differences in the levels of 33 of the 279 expressed miRNAs (11.8%) were observed among the three groups in response to the anesthetic agents, and when compared with the control group, significant differences were found in 26 of the 279 expressed miRNAs (9.3%). Following sevoflurane anesthesia, the levels of four miRNAs were significantly increased and those of 12 were significantly reduced. By contrast, following propofol anesthesia, the levels of 11 miRNAs were significantly reduced but no miRNAs exhibited significantly elevated levels. One miRNA was common between the two anesthesia groups, whereas 14 miRNAs were significantly differentially expressed. In conclusion, sevoflurane and propofol exerted different effects on miRNA expression in the rat hippocampus.
Collapse
Affiliation(s)
- Gentaro Goto
- Department of Anesthesiology and Pain Medicine, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan
| | - Yoko Hori
- Department of Anesthesiology and Pain Medicine, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan
| | - Masashi Ishikawa
- Department of Anesthesiology and Pain Medicine, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan
| | - Shunsuke Tanaka
- Department of Anesthesiology and Pain Medicine, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan
| | - Atsuhiro Sakamoto
- Department of Anesthesiology and Pain Medicine, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan
| |
Collapse
|