1
|
Penning A, Snoeck S, Garritsen O, Tosoni G, Hof A, de Boer F, van Hasenbroek J, Zhang L, Thrupp N, Craessaerts K, Fiers M, Salta E. NACC2, a molecular effector of miR-132 regulation at the interface between adult neurogenesis and Alzheimer's disease. Sci Rep 2024; 14:21163. [PMID: 39256511 PMCID: PMC11387632 DOI: 10.1038/s41598-024-72096-6] [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: 05/03/2024] [Accepted: 09/03/2024] [Indexed: 09/12/2024] Open
Abstract
The generation of new neurons at the hippocampal neurogenic niche, known as adult hippocampal neurogenesis (AHN), and its impairment, have been implicated in Alzheimer's disease (AD). MicroRNA-132 (miR-132), the most consistently downregulated microRNA (miRNA) in AD, was recently identified as a potent regulator of AHN, exerting multilayered proneurogenic effects in adult neural stem cells (NSCs) and their progeny. Supplementing miR-132 in AD mouse brain restores AHN and relevant memory deficits, yet the exact mechanisms involved are still unknown. Here, we identify NACC2 as a novel miR-132 target implicated in both AHN and AD. miR-132 deficiency in mouse hippocampus induces Nacc2 expression and inflammatory signaling in adult NSCs. We show that miR-132-dependent regulation of NACC2 is involved in the initial stages of human NSC differentiation towards astrocytes and neurons. Later, NACC2 function in astrocytic maturation becomes uncoupled from miR-132. We demonstrate that NACC2 is present in reactive astrocytes surrounding amyloid plaques in mouse and human AD hippocampus, and that there is an anticorrelation between miR-132 and NACC2 levels in AD and upon induction of inflammation. Unraveling the molecular mechanisms by which miR-132 regulates neurogenesis and cellular reactivity in AD, will provide valuable insights towards its possible application as a therapeutic target.
Collapse
Affiliation(s)
- Amber Penning
- Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Sarah Snoeck
- Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Oxana Garritsen
- UMC Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Giorgia Tosoni
- Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Amber Hof
- Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Fleur de Boer
- Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | | | - Lin Zhang
- Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Nicky Thrupp
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
| | | | - Mark Fiers
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
| | - Evgenia Salta
- Netherlands Institute for Neuroscience, Amsterdam, The Netherlands.
| |
Collapse
|
2
|
Nateghi B, Keraudren R, Boulay G, Bazin M, Goupil C, Canet G, Loiselle A, St-Amour I, Planel E, Soulet D, Hébert SS. Beneficial effects of miR-132/212 deficiency in the zQ175 mouse model of Huntington's disease. Front Neurosci 2024; 18:1421680. [PMID: 39170678 PMCID: PMC11337869 DOI: 10.3389/fnins.2024.1421680] [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: 04/22/2024] [Accepted: 07/10/2024] [Indexed: 08/23/2024] Open
Abstract
Huntington's disease (HD) is a rare genetic neurodegenerative disorder caused by an expansion of CAG repeats in the Huntingtin (HTT) gene. One hypothesis suggests that the mutant HTT gene contributes to HD neuropathology through transcriptional dysregulation involving microRNAs (miRNAs). In particular, the miR-132/212 cluster is strongly diminished in the HD brain. This study explores the effects of miR-132/212 deficiency specifically in adult HD zQ175 mice. The absence of miR-132/212 did not impact body weight, body temperature, or survival rates. Surprisingly, miR-132/212 loss seemed to alleviate, in part, the effects on endogenous Htt expression, HTT inclusions, and neuronal integrity in HD zQ175 mice. Additionally, miR-132/212 depletion led to age-dependent improvements in certain motor functions. Transcriptomic analysis revealed alterations in HD-related networks in WT- and HD zQ175-miR-132/212-deficient mice, including significant overlap in BDNF and Creb1 signaling pathways. Interestingly, however, a higher number of miR-132/212 gene targets was observed in HD zQ175 mice lacking the miR-132/212 cluster, especially in the striatum. These findings suggest a nuanced interplay between miR-132/212 expression and HD pathogenesis, providing potential insights into therapeutic interventions. Further investigation is needed to fully understand the underlying mechanisms and therapeutic potential of modulating miR-132/212 expression during HD progression.
Collapse
Affiliation(s)
- Behnaz Nateghi
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, CHUL, Québec, QC, Canada
- Département de Psychiatrie et de Neurosciences, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Remi Keraudren
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, CHUL, Québec, QC, Canada
- Département de Psychiatrie et de Neurosciences, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Gabriel Boulay
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, CHUL, Québec, QC, Canada
- Département de Psychiatrie et de Neurosciences, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Marc Bazin
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, CHUL, Québec, QC, Canada
| | - Claudia Goupil
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, CHUL, Québec, QC, Canada
| | - Geoffrey Canet
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, CHUL, Québec, QC, Canada
| | - Andréanne Loiselle
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, CHUL, Québec, QC, Canada
| | - Isabelle St-Amour
- CERVO Brain Research Centre, Centre Intégré Universitaire de Santé et des Services Sociaux de la Capitale-Nationale, Québec, QC, Canada
| | - Emmanuel Planel
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, CHUL, Québec, QC, Canada
- Département de Psychiatrie et de Neurosciences, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Denis Soulet
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, CHUL, Québec, QC, Canada
- Faculté de Pharmacie, Université Laval, Québec, QC, Canada
| | - Sébastien S. Hébert
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, CHUL, Québec, QC, Canada
- Département de Psychiatrie et de Neurosciences, Faculté de Médecine, Université Laval, Québec, QC, Canada
| |
Collapse
|
3
|
Walgrave H, Penning A, Tosoni G, Snoeck S, Davie K, Davis E, Wolfs L, Sierksma A, Mars M, Bu T, Thrupp N, Zhou L, Moechars D, Mancuso R, Fiers M, Howden AJ, De Strooper B, Salta E. microRNA-132 regulates gene expression programs involved in microglial homeostasis. iScience 2023; 26:106829. [PMID: 37250784 PMCID: PMC10213004 DOI: 10.1016/j.isci.2023.106829] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 03/13/2023] [Accepted: 05/03/2023] [Indexed: 05/31/2023] Open
Abstract
microRNA-132 (miR-132), a known neuronal regulator, is one of the most robustly downregulated microRNAs (miRNAs) in the brain of Alzheimer's disease (AD) patients. Increasing miR-132 in AD mouse brain ameliorates amyloid and Tau pathologies, and also restores adult hippocampal neurogenesis and memory deficits. However, the functional pleiotropy of miRNAs requires in-depth analysis of the effects of miR-132 supplementation before it can be moved forward for AD therapy. We employ here miR-132 loss- and gain-of-function approaches using single-cell transcriptomics, proteomics, and in silico AGO-CLIP datasets to identify molecular pathways targeted by miR-132 in mouse hippocampus. We find that miR-132 modulation significantly affects the transition of microglia from a disease-associated to a homeostatic cell state. We confirm the regulatory role of miR-132 in shifting microglial cell states using human microglial cultures derived from induced pluripotent stem cells.
Collapse
Affiliation(s)
- Hannah Walgrave
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute (LBI), 3000 Leuven, Belgium
| | - Amber Penning
- Netherlands Institute for Neuroscience, 1105 BA Amsterdam, the Netherlands
| | - Giorgia Tosoni
- Netherlands Institute for Neuroscience, 1105 BA Amsterdam, the Netherlands
| | - Sarah Snoeck
- Netherlands Institute for Neuroscience, 1105 BA Amsterdam, the Netherlands
| | - Kristofer Davie
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium
- VIB-KU Leuven Center for Brain & Disease Research, Bioinformatics Core Facility, 3000 Leuven, Belgium
| | - Emma Davis
- UK Dementia Research Institute at UCL, London WC1E 6BT, UK
| | - Leen Wolfs
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute (LBI), 3000 Leuven, Belgium
| | - Annerieke Sierksma
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute (LBI), 3000 Leuven, Belgium
| | - Mayte Mars
- Netherlands Institute for Neuroscience, 1105 BA Amsterdam, the Netherlands
| | - Taofeng Bu
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute (LBI), 3000 Leuven, Belgium
| | - Nicola Thrupp
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute (LBI), 3000 Leuven, Belgium
| | - Lujia Zhou
- Discovery Neuroscience, Janssen Research and Development, Division of Janssen Pharmaceutica NV, 2340 Beerse, Belgium
| | - Diederik Moechars
- Discovery Neuroscience, Janssen Research and Development, Division of Janssen Pharmaceutica NV, 2340 Beerse, Belgium
| | - Renzo Mancuso
- Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB Center for Molecular Neurology, VIB, 2610 Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, 2610 Antwerp, Belgium
| | - Mark Fiers
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute (LBI), 3000 Leuven, Belgium
| | - Andrew J.M. Howden
- UK Dementia Research Institute, University of Dundee, Dundee DD1 4HN, UK
| | - Bart De Strooper
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute (LBI), 3000 Leuven, Belgium
- UK Dementia Research Institute at UCL, London WC1E 6BT, UK
| | - Evgenia Salta
- Netherlands Institute for Neuroscience, 1105 BA Amsterdam, the Netherlands
| |
Collapse
|
4
|
Tan Z, Li W, Cheng X, Zhu Q, Zhang X. Non-Coding RNAs in the Regulation of Hippocampal Neurogenesis and Potential Treatment Targets for Related Disorders. Biomolecules 2022; 13:biom13010018. [PMID: 36671403 PMCID: PMC9855933 DOI: 10.3390/biom13010018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/17/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Non-coding RNAs (ncRNAs), including miRNAs, lncRNAs, circRNAs, and piRNAs, do not encode proteins. Nonetheless, they have critical roles in a variety of cellular activities-such as development, neurogenesis, degeneration, and the response to injury to the nervous system-via protein translation, RNA splicing, gene activation, silencing, modifications, and editing; thus, they may serve as potential targets for disease treatment. The activity of adult neural stem cells (NSCs) in the subgranular zone of the hippocampal dentate gyrus critically influences hippocampal function, including learning, memory, and emotion. ncRNAs have been shown to be involved in the regulation of hippocampal neurogenesis, including proliferation, differentiation, and migration of NSCs and synapse formation. The interaction among ncRNAs is complex and diverse and has become a major topic within the life science. This review outlines advances in research on the roles of ncRNAs in modulating NSC bioactivity in the hippocampus and discusses their potential applications in the treatment of illnesses affecting the hippocampus.
Collapse
Affiliation(s)
- Zhengye Tan
- Department of Anatomy, Institute of Neurobiology, Medical School, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Wen Li
- Department of Anatomy, Institute of Neurobiology, Medical School, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Xiang Cheng
- Department of Anatomy, Institute of Neurobiology, Medical School, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Qing Zhu
- School of Pharmacy, Nantong University, Nantong 226001, China
- Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, Nantong 226001, China
| | - Xinhua Zhang
- Department of Anatomy, Institute of Neurobiology, Medical School, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
- Central Lab, Yancheng Third People’s Hospital, The Sixth Affiliated Hospital of Nantong University, Yancheng 224001, China
- Correspondence:
| |
Collapse
|
5
|
Zhang W, Ye F, Xiong J, He F, Yang L, Yin F, Peng J, Wang X. Silencing of miR-132-3p protects against neuronal injury following status epilepticus by inhibiting IL-1β-induced reactive astrocyte (A1) polarization. FASEB J 2022; 36:e22554. [PMID: 36111973 DOI: 10.1096/fj.202200110rr] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 08/28/2022] [Accepted: 09/06/2022] [Indexed: 02/05/2023]
Abstract
Mesial temporal lobe epilepsy (MTLE) is one of the most common refractory epilepsies and is usually accompanied by a range of brain pathological changes, such as neuronal injury and astrocytosis. Naïve astrocytes are readily converted to cytotoxic reactive astrocytes (A1) in response to inflammatory stimulation, suppressing the polarization of A1 protects against neuronal death in early central nervous system injury. Our previous study found that pro-inflammatory cytokines and miR-132-3p (hereinafter referred to as "miR-132") expression were upregulated, but how miR-132 affected reactive astrocyte polarization and neuronal damage during epilepsy is not fully understood. Here, we aimed to explore the effect and mechanism of miR-132 on A1 polarization. Our results confirmed that A1 markers were significantly elevated in the hippocampus of MTLE rats and IL-1β-treated primary astrocytes. In vivo, knockdown of miR-132 by lateral ventricular injection reduced A1 astrocytes, neuronal loss, mossy fiber sprouting, and remitted the severity of status epilepticus and the recurrence of spontaneous recurrent seizures. In vitro, the neuronal cell viability and axon length were reduced by additional treatment with A1 astrocyte conditioned media (ACM), and downregulation of astrocyte miR-132 rescued the inhibition of cell activity by A1 ACM, while the length of axons was further inhibited. The regulation of miR-132 on A1 astrocytes may be related to its target gene expression. Our results show that interfering with astrocyte polarization may be a breakthrough in the treatment of refractory epilepsy, which may extend to the research of other astrocyte polarization-mediated brain injuries.
Collapse
Affiliation(s)
- Wen Zhang
- Department of Pediatrics, Xiangya Hospital of Central South University, Changsha, China
| | - Fanghua Ye
- Department of Pediatrics, Xiangya Hospital of Central South University, Changsha, China
| | - Juan Xiong
- Department of Pediatrics, Xiangya Hospital of Central South University, Changsha, China
| | - Fang He
- Department of Pediatrics, Xiangya Hospital of Central South University, Changsha, China
| | - Li Yang
- Department of Pediatrics, Xiangya Hospital of Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Pediatrics, Changsha, China
| | - Fei Yin
- Department of Pediatrics, Xiangya Hospital of Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Pediatrics, Changsha, China
| | - Jing Peng
- Department of Pediatrics, Xiangya Hospital of Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Pediatrics, Changsha, China
| | - Xiaole Wang
- Department of Pediatrics, Xiangya Hospital of Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Pediatrics, Changsha, China
| |
Collapse
|
6
|
C-terminal binding protein 2 promotes high-glucose-triggered cell proliferation, angiogenesis and cellular adhesion of human retinal endothelial cell line. Int Ophthalmol 2022; 42:2975-2985. [PMID: 35353294 DOI: 10.1007/s10792-022-02283-9] [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: 10/21/2021] [Accepted: 03/12/2022] [Indexed: 10/18/2022]
Abstract
PURPOSE The proliferation and angiogenesis of human retinal endothelial cells (HRECs) are critical for the pathophysiology of diabetic retinopathy (DR). C-terminal binding protein 2 (CtBP2) has multiple biologic functions, but its effect on HRECs under high-glucose (HG) conditions is unclear. METHODS The cell viability, angiogenesis, cellular adhesion and CtBP2 expression levels of HRECs were measured following treatment with different concentrations of glucose. Small interfering CtBP2-targeting RNA, wide-type and function mutant plasmid of CtBP2 were constructed and then were transfected into HRECs to evaluate the effects of CtBP2 on cell functions of HRECs. RESULTS The expression of CtBP2 in HRECs was increased after HG treatment. HG treatment significantly increased cell proliferation, angiogenesis, and decreased relative gene expressions in gap junctions, tight junctions and adherens junctions. After CtBP2 was inhibited via siRNA, the changes induced by HG were partially restored. Conversely, only wild-type CtBP2 could increase cell proliferation and angiogenesis under HG condition. Mechanistically, we also found that CtBP2 exerted its functions to effect HG-induced changes via Akt signaling pathway. CONCLUSION This study implicates that CtBP2 promotes HG-induced cell proliferation, angiogenesis and cellular adhesion, and CtBP2 might be a potential target in the prevention of DR.
Collapse
|
7
|
miR-132-5p regulates apoptosis and autophagy in MPTP model of Parkinson's disease by targeting ULK1. Neuroreport 2021; 31:959-965. [PMID: 32658123 DOI: 10.1097/wnr.0000000000001494] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder that is characterized by a loss of dopaminergic neurons in the substantia nigra of the brain. Numerous investigations have focused on the underlying mechanism involved in the progression of PD in recent decades. miR-132 is abnormal expression in many diseases including PD. However, the functional role and molecular mechanism of miR-132-5p in PD pathogenesis are still not elucidated. In our study, we found miR-132-5p was upregulated in 1-methyl-4-pheny-1,2,3,6-tetrahydropyridine (MPTP) model of PD. MTT assay and flow cytometric analysis revealed that inhibition of miR-132-5p increased cell survival ability and reduced MPTP-induced apoptosis of SH-SY5Y cells. Furthermore, inhibition of miR-132-5p could significantly suppressed mRNA and protein expression levels of LC3 and Beclin 1, indicating inhibition of miR-132-5p might restrain autophagy in PD. Subsequently, ULK1 was identified as a target of miR-132-5p and positively regulated by miR-132-5p at both mRNA and protein levels. Additionally, ectopic expression of ULK1 was able to reverse the effects of miR-132-5p inhibition. Taken together, our results demonstrated that miR-132-5p inhibition might exert a protective role in MPTP-treated PD models by targeting ULK1, indicating that miR-132-5p may be a prospective therapeutic target for PD.
Collapse
|
8
|
Walgrave H, Balusu S, Snoeck S, Vanden Eynden E, Craessaerts K, Thrupp N, Wolfs L, Horré K, Fourne Y, Ronisz A, Silajdžić E, Penning A, Tosoni G, Callaerts-Vegh Z, D'Hooge R, Thal DR, Zetterberg H, Thuret S, Fiers M, Frigerio CS, De Strooper B, Salta E. Restoring miR-132 expression rescues adult hippocampal neurogenesis and memory deficits in Alzheimer's disease. Cell Stem Cell 2021; 28:1805-1821.e8. [PMID: 34033742 DOI: 10.1016/j.stem.2021.05.001] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/24/2021] [Accepted: 04/30/2021] [Indexed: 12/14/2022]
Abstract
Neural stem cells residing in the hippocampal neurogenic niche sustain lifelong neurogenesis in the adult brain. Adult hippocampal neurogenesis (AHN) is functionally linked to mnemonic and cognitive plasticity in humans and rodents. In Alzheimer's disease (AD), the process of generating new neurons at the hippocampal neurogenic niche is impeded, yet the mechanisms involved are unknown. Here we identify miR-132, one of the most consistently downregulated microRNAs in AD, as a potent regulator of AHN, exerting cell-autonomous proneurogenic effects in adult neural stem cells and their progeny. Using distinct AD mouse models, cultured human primary and established neural stem cells, and human patient material, we demonstrate that AHN is directly affected by AD pathology. miR-132 replacement in adult mouse AD hippocampus restores AHN and relevant memory deficits. Our findings corroborate the significance of AHN in mouse models of AD and reveal the possible therapeutic potential of targeting miR-132 in neurodegeneration.
Collapse
Affiliation(s)
- Hannah Walgrave
- VIB Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Sriram Balusu
- VIB Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Sarah Snoeck
- Laboratory of Neurogenesis and Neurodegeneration, Netherlands Institute for Neuroscience, 1105BA Amsterdam, the Netherlands
| | - Elke Vanden Eynden
- VIB Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Katleen Craessaerts
- VIB Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Nicky Thrupp
- VIB Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Leen Wolfs
- VIB Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Katrien Horré
- VIB Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Yannick Fourne
- VIB Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Alicja Ronisz
- KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium; Laboratory for Neuropathology, KU Leuven, and Department of Pathology, UZ Leuven, 3000 Leuven, Belgium
| | - Edina Silajdžić
- Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE5 9RX, UK
| | - Amber Penning
- Laboratory of Neurogenesis and Neurodegeneration, Netherlands Institute for Neuroscience, 1105BA Amsterdam, the Netherlands
| | - Giorgia Tosoni
- Laboratory of Neurogenesis and Neurodegeneration, Netherlands Institute for Neuroscience, 1105BA Amsterdam, the Netherlands
| | - Zsuzsanna Callaerts-Vegh
- KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium; Laboratory for Biological Psychology, KU Leuven, 3000 Leuven, Belgium
| | - Rudi D'Hooge
- KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium; Laboratory for Biological Psychology, KU Leuven, 3000 Leuven, Belgium
| | - Dietmar Rudolf Thal
- KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium; Laboratory for Neuropathology, KU Leuven, and Department of Pathology, UZ Leuven, 3000 Leuven, Belgium
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 431 80 Mölndal, Sweden; Department of Neurodegenerative Disease, UCL Institute of Neurology, London, WC1N 3BG, UK; Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, 431 80 Mölndal, Sweden; UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
| | - Sandrine Thuret
- Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE5 9RX, UK
| | - Mark Fiers
- VIB Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium
| | | | - Bart De Strooper
- VIB Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium; UK Dementia Research Institute at UCL, London, WC1E 6BT, UK.
| | - Evgenia Salta
- Laboratory of Neurogenesis and Neurodegeneration, Netherlands Institute for Neuroscience, 1105BA Amsterdam, the Netherlands.
| |
Collapse
|
9
|
Stojanovic T, Benes H, Awad A, Bormann D, Monje FJ. Nicotine abolishes memory-related synaptic strengthening and promotes synaptic depression in the neurogenic dentate gyrus of miR-132/212 knockout mice. Addict Biol 2021; 26:e12905. [PMID: 32293776 PMCID: PMC7988623 DOI: 10.1111/adb.12905] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/21/2020] [Accepted: 03/30/2020] [Indexed: 12/25/2022]
Abstract
Micro-RNAs (miRNAs) are highly evolutionarily conserved short-length/noncoding RNA molecules that modulate a wide range of cellular functions in many cell types by regulating the expression of a variety of targeted genes. miRNAs have also recently emerged as key regulators of neuronal genes mediating the effects of psychostimulant drugs and memory-related neuroplasticity processes. Smoking is a predominant addictive behaviour associated with millions of deaths worldwide, and nicotine is a potent natural psychoactive agonist of cholinergic receptors, highly abundant in cigarettes. The influence of miRNAs modulation on cholinergic signalling in the nervous system remains however poorly explored. Using miRNA knockout mice and biochemical, electrophysiological and pharmacological approaches, we examined the effects of miR-132/212 gene disruption on the levels of hippocampal nicotinic acetylcholine receptors, total ERK and phosphorylated ERK (pERK) and MeCP2 protein levels, and studied the impact of nicotine stimulation on hippocampal synaptic transmission and synaptic depression and strengthening. miR-132/212 deletion significantly altered α7-nAChR and pERK protein levels, but not total ERK or MeCP2, and resulted in both exacerbated synaptic depression and virtually abolished memory-related synaptic strengthening upon nicotine stimulation. These observations reveal a functional miRNAs/nicotinergic signalling interplay critical for nicotinic-receptor expression and neuroplasticity in brain structures relevant for drug addiction and learning and memory functions.
Collapse
Affiliation(s)
- Tamara Stojanovic
- Center for Physiology and Pharmacology, Department of Neurophysiology and NeuropharmacologyMedical University of ViennaViennaAustria
| | - Hannah Benes
- Center for Physiology and Pharmacology, Department of Neurophysiology and NeuropharmacologyMedical University of ViennaViennaAustria
| | - Amena Awad
- Center for Physiology and Pharmacology, Department of Neurophysiology and NeuropharmacologyMedical University of ViennaViennaAustria
| | - Daniel Bormann
- Center for Physiology and Pharmacology, Department of Neurophysiology and NeuropharmacologyMedical University of ViennaViennaAustria
| | - Francisco J. Monje
- Center for Physiology and Pharmacology, Department of Neurophysiology and NeuropharmacologyMedical University of ViennaViennaAustria
| |
Collapse
|
10
|
Liu W, Shi X, Wang B. microRNA-133a exerts tumor suppressive role in oral squamous cell carcinoma through the Notch signaling pathway via downregulation of CTBP2. Cancer Gene Ther 2021; 29:62-72. [PMID: 33531645 DOI: 10.1038/s41417-020-00289-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 12/07/2020] [Accepted: 12/11/2020] [Indexed: 12/14/2022]
Abstract
microRNAs (miRNAs) have been revealed to participate in some oral cancers and are proved to be effective. In the present study, we tried to explore the biological function of miR-133a in oral squamous cell carcinoma (OSCC) cells. The relationship that C-terminal-binding proteins 2 (CTBP2) was the putative target gene of miR-133a revealed from bioinformatics analysis was further was further validated by dual-luciferase reporter gene assay. In total, 40 patients with OSCC were enrolled for characterization of miR-133a, CTBP2, and Notch signaling pathway-related gene expression in clinical OSCC tissues. Low expression of miR-133a and high expression of CTBP2, Hes1, Notch-1, and Notch-3 were determined in OSCC tissues. OSCC cell lines were transfected with miR-133a inhibitor, miR-133a mimic, or shRNA targeting CTBP2, in response to which cell proliferation, migration, invasion, cell cycle, and apoptosis were evaluated. Transfection of miR-133a mimic induced apoptosis and inhibited OSCC cell proliferation, migration, and invasion and this was demonstrated to be attributable to decreased CTBP2 expression and suppression of the Notch signaling pathway. Taken together, we concluded that miR-133a acted as a tumor suppressor in OSCC through inhibition of the Notch signaling pathway via binding to CTBP2.
Collapse
Affiliation(s)
- Wei Liu
- Department of Stomatology, Linyi People's Hospital, 276000, Linyi, P. R. China
| | - Xiaoming Shi
- Department of Stomatology, Linyi People's Hospital, 276000, Linyi, P. R. China
| | - Baoliang Wang
- Department of Stomatology, Linyi People's Hospital, 276000, Linyi, P. R. China.
| |
Collapse
|
11
|
Chen D, Liu J, Wu Z, Li SH. Role of miR-132/methyl-CpG-binding protein 2 in the regulation of neural stem cell differentiation. Neural Regen Res 2021; 16:345-349. [PMID: 32859795 PMCID: PMC7896221 DOI: 10.4103/1673-5374.290908] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Methyl-CpG-binding protein 2 (MeCP2) is a well-known transcription repressor, and mutations in MECP2 cause serious neurological disorders. Many studies have suggested that MeCP2 is involved in neural maturation only, and have not reported its role in neural stem cell differentiation. In the present study, we investigated this possible role of MeCP2 in neural stem cells. We used two different differentiation methods to explore how MeCP2 influences neural stem cell differentiation. When we transfected MeCP2-overexpressing lentivirus into neural stem cells, astrocytic differentiation was impaired. This impaired astrocytic differentiation occurred even in conditions of 20% fetal bovine serum, which favored astrocytic differentiation. In addition, miR-132 had the largest expression change after differentiation among several central nervous system related miRNAs. A luciferase assay confirmed that miR-132 directly targeted MeCP2, and that miR-132 was able to reduce MeCP2 expression at both the RNA and protein levels. The upregulation of miR-132 by miRNA mimics promoted astrocytic differentiation, which was fully recovered by MeCP2 overexpression. These results indicate that miR-132 regulates cell lineage differentiation by reducing MeCP2. The study was approved by the Ethics Committee of Shanghai Tenth People's Hospital of TongJi University, China (approval No. SHDSYY-2018-4748) on March 10, 2018.
Collapse
Affiliation(s)
- Dong Chen
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Jie Liu
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Zhong Wu
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Shao-Hua Li
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| |
Collapse
|
12
|
Intracranial Self-Stimulation Modulates Levels of SIRT1 Protein and Neural Plasticity-Related microRNAs. Mol Neurobiol 2020; 57:2551-2562. [PMID: 32219698 DOI: 10.1007/s12035-020-01901-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 03/09/2020] [Indexed: 02/07/2023]
Abstract
Deep brain stimulation (DBS) of reward system brain areas, such as the medial forebrain bundle (MFB), by means of intracranial self-stimulation (ICSS), facilitates learning and memory in rodents. MFB-ICSS has been found capable of modifying different plasticity-related proteins, but its underlying molecular mechanisms require further elucidation. MicroRNAs (miRNAs) and the longevity-associated SIRT1 protein have emerged as important regulatory molecules implicated in neural plasticity. Thus, we aimed to analyze the effects of MFB-ICSS on miRNAs expression and SIRT1 protein levels in hippocampal subfields and serum. We used OpenArray to select miRNA candidates differentially expressed in the dentate gyrus (DG) of ICSS-treated (3 sessions, 45' session/day) and sham rats. We further analyzed the expression of these miRNAs, together with candidates selected after bibliographic screening (miR-132-3p, miR-134-5p, miR-146a-5p, miR-181c-5p) in DG, CA1, and CA3, as well as in serum, by qRT-PCR. We also assessed tissue and serum SIRT1 protein levels by Western Blot and ELISA, respectively. Expression of miR-132-3p, miR-181c-5p, miR-495-3p, and SIRT1 protein was upregulated in DG of ICSS rats (P < 0.05). None of the analyzed molecules was regulated in CA3, while miR-132-3p was also increased in CA1 (P = 0.011) and serum (P = 0.048). This work shows for the first time that a DBS procedure, specifically MFB-ICSS, modulates the levels of plasticity-related miRNAs and SIRT1 in specific hippocampal subfields. The mechanistic role of these molecules could be key to the improvement of memory by MFB-ICSS. Moreover, regarding the proposed clinical applicability of DBS, serum miR-132 is suggested as a potential treatment biomarker.
Collapse
|
13
|
Karaca E, Li X, Lewicki J, Neofytou C, Guérout N, Barnabé-Heider F, Hermanson O. The corepressor CtBP2 is required for proper development of the mouse cerebral cortex. Mol Cell Neurosci 2020; 104:103481. [PMID: 32169478 DOI: 10.1016/j.mcn.2020.103481] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 03/05/2020] [Accepted: 03/09/2020] [Indexed: 12/21/2022] Open
Abstract
The development of the cerebral cortex depends on numerous parameters, including extracellular cues and microenvironmental factors that also affect gene expression. C-Terminal Binding Proteins (CtBPs) 1 and 2 are transcriptional co-repressors which have been shown to be critically involved in embryonic development. CtBPs are oxygen sensing molecules, and we have previously demonstrated an important role for CtBP1 in integrating oxygen levels and BMP-signaling to influence neural progenitor fate choice. In turn, CtBP2 has been associated with neurodevelopment and neurological disease, and we have shown that CtBP2 acetylation and dimerization, required for proper transcriptional activity, are regulated by microenvironmental oxygen levels. Yet, the putative function of CtBP2 in mammalian cortical development and neurogenesis in vivo is still largely unknown. Here we show that CtBP2 was widely expressed by neural stem and progenitor cells (NSPCs) as well as neurons during cortical development in mice. By using in utero electroporation of siRNA to reduce the levels of CtBP2 mRNA and protein in the developing mouse brain, we found that the NSPC proliferation and migration were largely perturbed, while glial differentiation under these conditions remained unchanged. Our study provides evidence that CtBP2 is required for the maintenance and migration of the NSPCs during mouse cortical development.
Collapse
Affiliation(s)
- Esra Karaca
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden; Department of Cardiothoracic Surgery, Stanford University, California, USA.
| | - Xiaofei Li
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden; Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Jakub Lewicki
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | - Nicolas Guérout
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden; Normandie Université, UNIROUEN, EA3830 GRHV, Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | | | - Ola Hermanson
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
14
|
Nagarajan B, Harder A, Japp A, Häberlein F, Mingardo E, Kleinert H, Yilmaz Ö, Zoons A, Rau B, Christ A, Kubitscheck U, Eiberger B, Sandhoff R, Eckhardt M, Hartmann D, Odermatt B. CNS myelin protein 36K regulates oligodendrocyte differentiation through Notch. Glia 2019; 68:509-527. [PMID: 31702067 DOI: 10.1002/glia.23732] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 09/23/2019] [Accepted: 09/24/2019] [Indexed: 12/12/2022]
Abstract
In contrast to humans and other mammals, zebrafish can successfully regenerate and remyelinate central nervous system (CNS) axons following injury. In addition to common myelin proteins found in mammalian myelin, 36K protein is a major component of teleost fish CNS myelin. Although 36K is one of the most abundant proteins in zebrafish brain, its function remains unknown. Here we investigate the function of 36K using translation-blocking Morpholinos. Morphant larvae showed fewer dorsally migrated oligodendrocyte precursor cells as well as upregulation of Notch ligand. A gamma secretase inhibitor, which prevents activation of Notch, could rescue oligodendrocyte precursor cell numbers in 36K morphants, suggesting that 36K regulates initial myelination through inhibition of Notch signaling. Since 36K like other short chain dehydrogenases might act on lipids, we performed thin layer chromatography and mass spectrometry of lipids and found changes in lipid composition in 36K morphant larvae. Altogether, we suggest that during early development 36K regulates membrane lipid composition, thereby altering the amount of transmembrane Notch ligands and the efficiency of intramembrane gamma secretase processing of Notch and thereby influencing oligodendrocyte precursor cell differentiation and further myelination. Further studies on the role of 36K short chain dehydrogenase in oligodendrocyte precursor cell differentiation during remyelination might open up new strategies for remyelination therapies in human patients.
Collapse
Affiliation(s)
- Bhuvaneswari Nagarajan
- Institute of Anatomy, Division of Anatomy and Cell Biology, University Clinics, University of Bonn, Bonn, Germany
| | - Alexander Harder
- Institute of Physical and Theoretical Chemistry, University of Bonn, Bonn, Germany
| | - Anna Japp
- Institute of Neuropathology, University Clinics, University of Bonn, Bonn, Germany
| | - Felix Häberlein
- Institute of Anatomy, Division of Anatomy and Cell Biology, University Clinics, University of Bonn, Bonn, Germany.,Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Enrico Mingardo
- Institute of Anatomy, Division of Anatomy and Cell Biology, University Clinics, University of Bonn, Bonn, Germany
| | - Henning Kleinert
- Institute of Anatomy, Division of Anatomy and Cell Biology, University Clinics, University of Bonn, Bonn, Germany
| | - Öznur Yilmaz
- Institute of Anatomy, Division of Anatomy and Cell Biology, University Clinics, University of Bonn, Bonn, Germany
| | - Angelika Zoons
- Institute of Anatomy, Division of Neuroanatomy, University Clinics, University of Bonn, Bonn, Germany
| | - Birgit Rau
- Institute of Anatomy, Division of Neuroanatomy, University Clinics, University of Bonn, Bonn, Germany
| | - Andrea Christ
- Institute of Anatomy, Division of Anatomy and Cell Biology, University Clinics, University of Bonn, Bonn, Germany
| | - Ulrich Kubitscheck
- Institute of Physical and Theoretical Chemistry, University of Bonn, Bonn, Germany
| | - Britta Eiberger
- Institute of Anatomy, Division of Anatomy and Cell Biology, University Clinics, University of Bonn, Bonn, Germany
| | - Roger Sandhoff
- Lipid Pathobiochemistry Group, German Cancer Research Centre, Heidelberg, Germany
| | - Matthias Eckhardt
- Institute of Biochemistry and Molecular Biology, University Clinics, University of Bonn, Bonn, Germany
| | - Dieter Hartmann
- Institute of Anatomy, Division of Neuroanatomy, University Clinics, University of Bonn, Bonn, Germany
| | - Benjamin Odermatt
- Institute of Anatomy, Division of Anatomy and Cell Biology, University Clinics, University of Bonn, Bonn, Germany.,Institute of Anatomy, Division of Neuroanatomy, University Clinics, University of Bonn, Bonn, Germany
| |
Collapse
|
15
|
Kizil C, Bhattarai P. Is Alzheimer's Also a Stem Cell Disease? - The Zebrafish Perspective. Front Cell Dev Biol 2018; 6:159. [PMID: 30533414 PMCID: PMC6265475 DOI: 10.3389/fcell.2018.00159] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 11/06/2018] [Indexed: 12/22/2022] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease and is the leading form of dementia. AD entails chronic inflammation, impaired synaptic integrity and reduced neurogenesis. The clinical and molecular onsets of the disease do not temporally overlap and the initiation phase of the cellular changes might start with a complex causativeness between chronic inflammation, reduced neural stem cell plasticity and neurogenesis. Although the immune and neuronal aspects in AD are well studied, the neural stem cell-related features are far less investigated. An intriguing question is, therefore, whether a stem cell can ever be made proliferative and neurogenic during the prevalent AD in the brain. Recent findings affirm this hypothesis and thus a plausible way to circumvent the AD phenotypes could be to mobilize the endogenous stem cells by enhancing their proliferative and neurogenic capacity as well as to provide the newborn neurons the potential to survive and integrate into the existing circuitry. To address these questions, zebrafish offers unprecedented information and tools, which can be effectively translated into mammalian experimental systems.
Collapse
Affiliation(s)
- Caghan Kizil
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Helmholtz Association, Dresden, Germany
- Center for Regenerative Therapies Dresden, Cluster of Excellence, Technische Universität Dresden, Dresden, Germany
| | - Prabesh Bhattarai
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Helmholtz Association, Dresden, Germany
- Center for Regenerative Therapies Dresden, Cluster of Excellence, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
16
|
Chao G, Wang Y, Ye F, Zhang S. Regulation of Colonic Mucosal MicroRNA Expression via Multiple Targets in Visceral Hypersensitivity Rats by Tongxieyaofang. Yonsei Med J 2018; 59:945-950. [PMID: 30187701 PMCID: PMC6127421 DOI: 10.3349/ymj.2018.59.8.945] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 07/30/2018] [Accepted: 08/02/2018] [Indexed: 02/07/2023] Open
Abstract
PURPOSE This study aimed to screen for differentially expressed microRNAs (miRNAs) in the colons of rats with visceral hypersensitivity to build the expression profiles of miRNAs therein and to determine the mechanism of Tongxieyaofang use in the treatment of irritable bowel syndrome (IBS). MATERIALS AND METHODS Forty Sprague-Dawley rats were divided randomly into four groups: control group, model control group (induced by rectum stimulus and evaluated by abdominal withdraw reaction), treatment control group (normal saline), and Tongxieyaofang group (treated with Tongxieyaofang). We screened for differential expression of colonic mucosal miRNAs using liquid chip technology and verified the expression thereof using reverse transcription-PCR. RESULTS The visceral hypersensitivity rat model was successfully established. We found the expression of let-7f, let-7i, miR-130b, miR-29a, miR-132, miR-21, and miR-375 to be up-regulated (p<0.05), while the expression of miR-24, miR-31a, miR-192, miR-221, and miR-223 was down-regulated (p<0.05) in the visceral hypersensitivity rats. After treatment with Tongxieyaofang, the expression of let-7f, let-7i, miR-130b, miR-29a, miR-132, miR-21, and miR-375 was reduced (p<0.05), whereas the expression of miR-24, miR-31a, miR-192, miR-221, miR-223 was increased, compared to the treatment control group (p<0.05). CONCLUSION MiRNAs play a pivotal role in visceral hypersensitivity and might be targets in the treatment of IBS by Tongxieyaofang.
Collapse
Affiliation(s)
- Guanqun Chao
- Department of Family Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Yingying Wang
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fangxu Ye
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shuo Zhang
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
17
|
Du XF, Xu B, Zhang Y, Chen MJ, Du JL. A transgenic zebrafish model for in vivo long-term imaging of retinotectal synaptogenesis. Sci Rep 2018; 8:14077. [PMID: 30232367 PMCID: PMC6145912 DOI: 10.1038/s41598-018-32409-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 09/04/2018] [Indexed: 01/07/2023] Open
Abstract
The retinotectal synapse in larval zebrafish, combined with live time-lapse imaging, provides an advantageous model for study of the development and remodelling of central synapses in vivo. In previous studies, these synapses were labelled by transient expression of fluorescence-tagged synaptic proteins, which resulted in the dramatic variation of labelling patterns in each larva. Here, using GAL4-Upstream Activating Sequence (GAL4-UAS) methodology, we generated stable transgenic lines, which express EGFP-tagged synaptophysin (a presynaptic protein) in retinal ganglion cells (RGCs), to reliably label the pre-synaptic site of retinotectal synapses. This tool avoids the variable labelling of RGCs that occurs in transient transgenic larvae. We obtained several stable transgenic lines that differ consistently in the number of labelled RGCs. Using stable lines that consistently had a single labelled RGC, we could trace synaptogenic dynamics on an individual RGC axonal arbor across different developmental stages. In the stable lines that consistently had multiple labelled RGCs, we could simultaneously monitor both pre- and post-synaptic compartments by combining transient labelling of post-synaptic sites on individual tectal neurons. These tools allowed us to investigate molecular events underlying synaptogenesis and found that the microRNA-132 (miR-132) is required for developmental synaptogenesis. Thus, these transgenic zebrafish stable lines provide appropriate tools for studying central synaptogenesis and underlying molecular mechanisms in intact vertebrate brain.
Collapse
Affiliation(s)
- Xu-Fei Du
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, 200031, China.
| | - Bing Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, 200031, China
| | - Yu Zhang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, 200031, China.,School of Future Technology, University of Chinese Academy of Sciences, 19A Yu-Quan Road, Beijing, 100049, China
| | - Min-Jia Chen
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, 200031, China.,School of Life Science and Technology, ShanghaiTech University, 319 Yue-Yang Road, Shanghai, 200031, China
| | - Jiu-Lin Du
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, 200031, China. .,School of Future Technology, University of Chinese Academy of Sciences, 19A Yu-Quan Road, Beijing, 100049, China. .,School of Life Science and Technology, ShanghaiTech University, 319 Yue-Yang Road, Shanghai, 200031, China.
| |
Collapse
|
18
|
The pericyte secretome: Potential impact on regeneration. Biochimie 2018; 155:16-25. [PMID: 29698670 DOI: 10.1016/j.biochi.2018.04.015] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Accepted: 04/20/2018] [Indexed: 12/11/2022]
Abstract
Personalized and regenerative medicine is an emerging therapeutic strategy that is based on cell biology and biomedical engineering used to develop biological substitutes to maintain normal function or restore damaged tissues and organs. The secretory capacities of different cell types are now explored as such possible therapeutic regenerative agents in a variety of diseases. A secretome can comprise chemokines, cytokines, growth factors, but also extracellular matrix components, microvesicles and exosomes as well as genetic material and may differ depending on the tissue and the stimulus applied to the cell. With regard to clinical applications, the secretome of mesenchymal stem cells (MSC) is currently the most widely explored. However, other cell types such as pericytes may have similar properties as MSC and the potential therapeutic possibilities of these cells are only just beginning to emerge. In this review, we will summarize the currently available data describing the secretome of pericytes and its potential implications for tissue regeneration, whereby we especially focus on brain pericytes as potential new target cell for neuroregeneration and brain repair.
Collapse
|
19
|
Qian Y, Song J, Ouyang Y, Han Q, Chen W, Zhao X, Xie Y, Chen Y, Yuan W, Fan C. Advances in Roles of miR-132 in the Nervous System. Front Pharmacol 2017; 8:770. [PMID: 29118714 PMCID: PMC5660991 DOI: 10.3389/fphar.2017.00770] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 10/12/2017] [Indexed: 12/24/2022] Open
Abstract
miR-132 is an endogenous small RNA and controls post-transcriptional regulation of gene expression via controlled degradation of mRNA or transcription inhibition. In the nervous system, miR-132 is significant for regulating neuronal differentiation, maturation and functioning, and widely participates in axon growth, neural migration, and plasticity. The miR-132 is affected by factors like mRNA expression, functional redundancy, and signaling cascades. It targets multiple downstream molecules to influence physiological and pathological neuronal activities. MiR-132 can influence the pathogenesis of many diseases, especially in the nervous system. The dysregulation of miR-132 results in the occurrence and exacerbation of neural developmental, degenerative diseases, like Alzheimer’s disease, Parkinson’s disease and epilepsy, neural infection and psychiatric disorders including disturbance of consciousness, cognition and memory, depression and schizophrenia. Regulation of miR-132 expression relieves symptoms, alleviates severity and finally effects a cure. This review aims to discuss the clinical potentials of miR-132 in the nervous system.
Collapse
Affiliation(s)
- Yun Qian
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Shanghai Sixth People's Hospital East Campus, Shanghai University of Medicine and Health, Shanghai, China
| | - Jialin Song
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Shanghai Sixth People's Hospital East Campus, Shanghai University of Medicine and Health, Shanghai, China
| | - Yuanming Ouyang
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Shanghai Sixth People's Hospital East Campus, Shanghai University of Medicine and Health, Shanghai, China
| | - Qixin Han
- Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Chen
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Shanghai Sixth People's Hospital East Campus, Shanghai University of Medicine and Health, Shanghai, China
| | - Xiaotian Zhao
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Yangmei Xie
- Department of Neurology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Yinghui Chen
- Department of Neurology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Weien Yuan
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Cunyi Fan
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
20
|
Salta E, De Strooper B. microRNA-132: a key noncoding RNA operating in the cellular phase of Alzheimer's disease. FASEB J 2017; 31:424-433. [PMID: 28148775 DOI: 10.1096/fj.201601308] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 12/12/2016] [Indexed: 12/20/2022]
Abstract
With the consideration of the broad involvement of microRNAs (miRNAs) in the regulation of molecular networks in the brain, it is not surprising that miRNA dysregulation causes neurodegeneration in animal models. miRNA profiling in the human brain has revealed miR-132 as one of the most severely down-regulated miRNAs at the intermediate and late Braak stages of Alzheimer's disease (AD), as well as in other neurodegenerative disorders. Suppression of miR-132 aggravates multiple layers of pathology at the molecular and functional level. We describe the potential therapeutic implications of these findings and suggest miRNA targeting or replacement as a realistic multi-hit, therapeutic strategy for AD. Salta, E., De Strooper, B. microRNA-132: a key noncoding RNA operating in the cellular phase of Alzheimer's disease.
Collapse
Affiliation(s)
- Evgenia Salta
- Vlaams Instituut voor Biotechnologie (VIB) Center for Brain and Disease, VIB-Leuven, Leuven, Belgium; .,Center for Human Genetics, Universitaire Ziekenhuizen and Leuven Research Institute for Neuroscience and Disease, KU-Leuven, Leuven, Belgium; and
| | - Bart De Strooper
- Vlaams Instituut voor Biotechnologie (VIB) Center for Brain and Disease, VIB-Leuven, Leuven, Belgium; .,Center for Human Genetics, Universitaire Ziekenhuizen and Leuven Research Institute for Neuroscience and Disease, KU-Leuven, Leuven, Belgium; and.,Institute of Neurology, University College London, London, United Kingdom
| |
Collapse
|
21
|
Abstract
The emerging complexity of the transcriptional landscape poses great challenges to our conventional preconceptions of how the genome regulates brain function and dysfunction. Non-protein-coding RNAs (ncRNAs) confer a high level of intricate and dynamic regulation of various molecular processes in the CNS and they have been implicated in neurodevelopment and brain ageing, as well as in synapse function and cognitive performance, in both health and disease. ncRNA-mediated processes may be involved in various aspects of the pathogenesis of neurodegenerative disorders. Understanding these events may help to develop novel diagnostic and therapeutic tools. Here, we provide an overview of the complex mechanisms that are affected by the diverse ncRNA classes that have been implicated in neurodegeneration.
Collapse
|
22
|
Neurons secrete miR-132-containing exosomes to regulate brain vascular integrity. Cell Res 2017; 27:882-897. [PMID: 28429770 PMCID: PMC5518987 DOI: 10.1038/cr.2017.62] [Citation(s) in RCA: 261] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 02/25/2017] [Accepted: 03/26/2017] [Indexed: 12/11/2022] Open
Abstract
Vascular integrity helps maintain brain microenvironment homeostasis, which is critical for the normal development and function of the central nervous system. It is known that neural cells can regulate brain vascular integrity. However, due to the high complexity of neurovascular interactions involved, understanding of the neural regulation of brain vascular integrity is still rudimentary. Using intact zebrafish larvae and cultured rodent brain cells, we find that neurons transfer miR-132, a highly conserved and neuron-enriched microRNA, via secreting exosomes to endothelial cells (ECs) to maintain brain vascular integrity. Following translocation to ECs through exosome internalization, miR-132 regulates the expression of vascular endothelial cadherin (VE-cadherin), an important adherens junction protein, by directly targeting eukaryotic elongation factor2kinase (eef2k). Disruption of neuronal miR-132 expression or exosome secretion, or overexpression of vascular eef2k impairs VE-cadherin expression and brain vascular integrity. Our study indicates that miR-132 acts as an intercellular signal mediating neural regulation of the brain vascular integrity and suggests that the neuronal exosome is a novel avenue for neurovascular communication.
Collapse
|
23
|
Salta E, Sierksma A, Vanden Eynden E, De Strooper B. miR-132 loss de-represses ITPKB and aggravates amyloid and TAU pathology in Alzheimer's brain. EMBO Mol Med 2016; 8:1005-18. [PMID: 27485122 PMCID: PMC5009807 DOI: 10.15252/emmm.201606520] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
microRNA‐132 (miR‐132) is involved in prosurvival, anti‐inflammatory and memory‐promoting functions in the nervous system and has been found consistently downregulated in Alzheimer's disease (AD). Whether and how miR‐132 deficiency impacts AD pathology remains, however, unaddressed. We show here that miR‐132 loss exacerbates both amyloid and TAU pathology via inositol 1,4,5‐trisphosphate 3‐kinase B (ITPKB) upregulation in an AD mouse model. This leads to increased ERK1/2 and BACE1 activity and elevated TAU phosphorylation. We confirm downregulation of miR‐132 and upregulation of ITPKB in three distinct human AD patient cohorts, indicating the pathological relevance of this pathway in AD.
Collapse
Affiliation(s)
- Evgenia Salta
- VIB Center for the Biology of Disease, Leuven, Belgium Center for Human Genetics, Universitaire ziekenhuizen and LIND, KU, Leuven, Belgium
| | - Annerieke Sierksma
- VIB Center for the Biology of Disease, Leuven, Belgium Center for Human Genetics, Universitaire ziekenhuizen and LIND, KU, Leuven, Belgium
| | - Elke Vanden Eynden
- VIB Center for the Biology of Disease, Leuven, Belgium Center for Human Genetics, Universitaire ziekenhuizen and LIND, KU, Leuven, Belgium
| | - Bart De Strooper
- VIB Center for the Biology of Disease, Leuven, Belgium Center for Human Genetics, Universitaire ziekenhuizen and LIND, KU, Leuven, Belgium Institute of Neurology, University College London, London, UK
| |
Collapse
|
24
|
Cai Z, Zhao B, Deng Y, Shangguan S, Zhou F, Zhou W, Li X, Li Y, Chen G. Notch signaling in cerebrovascular diseases (Review). Mol Med Rep 2016; 14:2883-98. [PMID: 27574001 PMCID: PMC5042775 DOI: 10.3892/mmr.2016.5641] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Accepted: 07/22/2016] [Indexed: 12/30/2022] Open
Abstract
The Notch signaling pathway is a crucial regulator of numerous fundamental cellular processes. Increasing evidence suggests that Notch signaling is involved in inflammation and oxidative stress, and thus in the progress of cerebrovascular diseases. In addition, Notch signaling in cerebrovascular diseases is associated with apoptosis, angiogenesis and the function of blood-brain barrier. Despite the contradictory results obtained to date as to whether Notch signaling is harmful or beneficial, the regulation of Notch signaling may provide a novel strategy for the treatment of cerebrovascular diseases.
Collapse
Affiliation(s)
- Zhiyou Cai
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Bin Zhao
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Yanqing Deng
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Shouqin Shangguan
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Faming Zhou
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Wenqing Zhou
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Xiaoli Li
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Yanfeng Li
- Department of Neurology, Peking Union Medical College Hospital, Beijing 100730, P.R. China
| | - Guanghui Chen
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| |
Collapse
|
25
|
microRNA-132/212 deficiency enhances Aβ production and senile plaque deposition in Alzheimer's disease triple transgenic mice. Sci Rep 2016; 6:30953. [PMID: 27484949 PMCID: PMC4971468 DOI: 10.1038/srep30953] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 07/11/2016] [Indexed: 01/02/2023] Open
Abstract
The abnormal regulation of amyloid-β (Aβ) metabolism (e.g., production, cleavage, clearance) plays a central role in Alzheimer’s disease (AD). Among endogenous factors believed to participate in AD progression are the small regulatory non-coding microRNAs (miRs). In particular, the miR-132/212 cluster is severely reduced in the AD brain. In previous studies we have shown that miR-132/212 deficiency in mice leads to impaired memory and enhanced Tau pathology as seen in AD patients. Here we demonstrate that the genetic deletion of miR-132/212 promotes Aβ production and amyloid (senile) plaque formation in triple transgenic AD (3xTg-AD) mice. Using RNA-Seq and bioinformatics, we identified genes of the miR-132/212 network with documented roles in the regulation of Aβ metabolism, including Tau, Mapk, and Sirt1. Consistent with these findings, we show that the modulation of miR-132, or its target Sirt1, can directly regulate Aβ production in cells. Finally, both miR-132 and Sirt1 levels correlated with Aβ load in humans. Overall, our results support the hypothesis that the miR-132/212 network, including Sirt1 and likely other target genes, contributes to abnormal Aβ metabolism and senile plaque deposition in AD. This study strengthens the importance of miR-dependent networks in neurodegenerative disorders, and opens the door to multifactorial drug targets of AD by targeting Aβ and Tau.
Collapse
|
26
|
Deng G, Ma L, Meng Q, Ju X, Jiang K, Jiang P, Yu Z. Notch signaling in the prostate: critical roles during development and in the hallmarks of prostate cancer biology. J Cancer Res Clin Oncol 2016; 142:531-47. [PMID: 25736982 DOI: 10.1007/s00432-015-1946-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 02/22/2015] [Indexed: 01/08/2023]
Abstract
PURPOSE This review aims to summarize the evidence that Notch signaling is associated with prostate development, tumorigenesis and prostate tumor progression. METHODS Studies in PubMed database were searched using the keywords of Notch signaling, prostate development and prostate cancer. Relevant literatures were identified and summarized. RESULTS The Notch pathway plays an important role in determining cell fate, proliferation, differentiation and apoptosis. Recent findings have highlighted the involvement of Notch signaling in prostate development and in the maintenance of adult prostate homeostasis. Aberrant Notch expression in tissues leads to dysregulation of Notch functions and promotes various neoplasms, including prostate cancer. High expression of Notch has been implicated in prostate cancer, and its expression increases with higher cancer grade. However, the precise role of Notch in prostate cancer has yet to be clearly defined. The roles of Notch either as an oncogene or tumor suppressor in prostate cancer hallmarks such as cell proliferation, apoptosis and anoikis, hypoxia, migration and invasion, angiogenesis as well as the correlation with metastasis are therefore discussed. CONCLUSIONS Notch signaling is a complicated signaling pathway in modulating prostate development and prostate cancer. Understanding and manipulating Notch signaling could therefore be of potential therapeutic value in combating prostate cancer.
Collapse
Affiliation(s)
- Gang Deng
- Department of Urology, The First People's Hospital of Hangzhou, 216 Huansha Road, Hangzhou, 310006, Zhejiang, China
| | - Libin Ma
- Department of Nephrology, The First People's Hospital of Hangzhou, 216 Huansha Road, Hangzhou, 310006, Zhejiang, China.
| | - Qi Meng
- Department of Urology, The First People's Hospital of Hangzhou, 216 Huansha Road, Hangzhou, 310006, Zhejiang, China
| | - Xiang Ju
- Department of Urology, The First People's Hospital of Hangzhou, 216 Huansha Road, Hangzhou, 310006, Zhejiang, China
| | - Kang Jiang
- Department of Urology, The First People's Hospital of Hangzhou, 216 Huansha Road, Hangzhou, 310006, Zhejiang, China
| | - Peiwu Jiang
- Department of Urology, The First People's Hospital of Hangzhou, 216 Huansha Road, Hangzhou, 310006, Zhejiang, China
| | - Zhijian Yu
- Department of Urology, The First People's Hospital of Hangzhou, 216 Huansha Road, Hangzhou, 310006, Zhejiang, China
| |
Collapse
|
27
|
Zhang G, Kang L, Chen J, Xue Y, Yang M, Qin B, Yang L, Zhang J, Lu H, Guan H. CtBP2 Regulates TGFβ2-Induced Epithelial-Mesenchymal Transition Through Notch Signaling Pathway in Lens Epithelial Cells. Curr Eye Res 2015; 41:1057-1063. [PMID: 26681554 DOI: 10.3109/02713683.2015.1092554] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND Epithelial-mesenchymal transition (EMT) of human lens epithelial cells (LECs) contributes to posterior capsule opacification (PCO). C-terminal binding protein 2 (CtBP2) has been reported to be essential in EMT and embryonic development. However, the function of CtBP2 in EMT of LECs is unknown. The goal of this study was to investigate the role of CtBP2 through Notch signaling in transforming growth factor β2 (TGFβ2)-induced EMT in LECs. METHODS The human LEC line SRA01/04 was cultured in the presence of TGFβ2 for different periods of time or with γ-Secretase Inhibitor IX (DAPT), a specific inhibitor of Notch receptor cleavage, for 24 h, utilizing plasmid-based method. The levels of protein expression of CtBP2, EMT markers, and Notch signaling molecules were measured by Western bolts. RESULTS Treatment of SRA01/04 cells with TGFβ2 induced typical molecular changes of EMT and increased the expression of CtBP2 in a time-dependent manner. Similarly, the expressions of Jagged1 and Notch1 were increased after TGFβ2 treatment. Knockdown of CtBP2 by specific siRNA inhibited TGFβ2-induced changes of Connexin 43 (CX43), α-smooth muscle actin (α-SMA), Notch1, and Notch intracellular domain (NICD). In addition, treatment of LECs with ectopic expression of CtBP2 changed the expressions of CX43, α-SMA, Notch1, and NICD, but blockade of Notch pathway with DAPT inhibited CtBP2-induced changes of α-SMA and CX43. CONCLUSION Our data suggest that CtBP2 plays a critical role in TGFβ2-induced EMT via the Jagged/Notch signaling pathway in human LECs and may contribute to the development of PCO.
Collapse
Affiliation(s)
- Guowei Zhang
- a Eye Institute , Affiliated Hospital of Nantong University , Nantong , Jiangsu Province , China
| | - Lihua Kang
- a Eye Institute , Affiliated Hospital of Nantong University , Nantong , Jiangsu Province , China
| | - Jia Chen
- a Eye Institute , Affiliated Hospital of Nantong University , Nantong , Jiangsu Province , China
| | - Ying Xue
- a Eye Institute , Affiliated Hospital of Nantong University , Nantong , Jiangsu Province , China
| | - Mei Yang
- a Eye Institute , Affiliated Hospital of Nantong University , Nantong , Jiangsu Province , China
| | - Bai Qin
- a Eye Institute , Affiliated Hospital of Nantong University , Nantong , Jiangsu Province , China
| | - Ling Yang
- a Eye Institute , Affiliated Hospital of Nantong University , Nantong , Jiangsu Province , China
| | - Junfang Zhang
- a Eye Institute , Affiliated Hospital of Nantong University , Nantong , Jiangsu Province , China
| | - Hong Lu
- a Eye Institute , Affiliated Hospital of Nantong University , Nantong , Jiangsu Province , China
| | - Huaijin Guan
- a Eye Institute , Affiliated Hospital of Nantong University , Nantong , Jiangsu Province , China
| |
Collapse
|
28
|
Hypoxia-Induced Upregulation of miR-132 Promotes Schwann Cell Migration After Sciatic Nerve Injury by Targeting PRKAG3. Mol Neurobiol 2015; 53:5129-39. [DOI: 10.1007/s12035-015-9449-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Accepted: 09/16/2015] [Indexed: 12/01/2022]
|
29
|
Campbell K, Booth SA. MicroRNA in neurodegenerative drug discovery: the way forward? Expert Opin Drug Discov 2014; 10:9-16. [DOI: 10.1517/17460441.2015.981254] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|