1
|
Brown JL, Hart DW, Boyle GE, Brown TG, LaCroix M, Baraibar AM, Pelzel R, Kim M, Sherman MA, Boes S, Sung M, Cole T, Lee MK, Araque A, Lesné SE. SNCA genetic lowering reveals differential cognitive function of alpha-synuclein dependent on sex. Acta Neuropathol Commun 2022; 10:180. [PMID: 36517890 PMCID: PMC9749314 DOI: 10.1186/s40478-022-01480-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 11/18/2022] [Indexed: 12/15/2022] Open
Abstract
Antisense oligonucleotide (ASO) therapy for neurological disease has been successful in clinical settings and its potential has generated hope for Alzheimer's disease (AD). We previously described that ablating SNCA encoding for α-synuclein (αSyn) in a mouse model of AD was beneficial. Here, we sought to demonstrate whether transient reduction of αSyn expression using ASOSNCA could be therapeutic in a mouse model of AD. The efficacy of the ASOSNCA was measured via immunocytochemistry, RT-qPCR and western blotting. To assess spatial learning and memory, ASOSNCA or PBS-injected APP and non-transgenic (NTG) mice, and separate groups of SNCA-null mice, were tested on the Barnes circular maze. Hippocampal slice electrophysiology and transcriptomic profiling were used to explore synaptic function and differential gene expression between groups. Reduction of SNCA transcripts alleviated cognitive deficits in male transgenic animals, but surprisingly, not in females. To determine the functional cause of this differential effect, we assessed memory function in SNCA-null mice. Learning and memory were intact in male mice but impaired in female animals, revealing that the role of αSyn on cognitive function is sex-specific. Transcriptional analyses identified a differentially expressed gene network centered around EGR1, a central modulator of learning and memory, in the hippocampi of SNCA-null mice. Thus, these novel results demonstrate that the function of αSyn on memory differs between male and female brains.
Collapse
Affiliation(s)
- Jennifer L Brown
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Institute for Translational Neuroscience, University of Minnesota, Wallin Medical Biosciences Building (Room 4-114), 2101 Sixth Street SE, CDC 2641, Minneapolis, MN, 55414, USA
| | - Damyan W Hart
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Institute for Translational Neuroscience, University of Minnesota, Wallin Medical Biosciences Building (Room 4-114), 2101 Sixth Street SE, CDC 2641, Minneapolis, MN, 55414, USA
| | - Gabriel E Boyle
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Institute for Translational Neuroscience, University of Minnesota, Wallin Medical Biosciences Building (Room 4-114), 2101 Sixth Street SE, CDC 2641, Minneapolis, MN, 55414, USA
- Graduate Program in Molecular and Cellular Biology, University of Washington, Seattle, WA, 98195, USA
| | - Taylor G Brown
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Medical Scientist Training Program, University of Minnesota, Minneapolis, MN, USA
- Institute for Translational Neuroscience, University of Minnesota, Wallin Medical Biosciences Building (Room 4-114), 2101 Sixth Street SE, CDC 2641, Minneapolis, MN, 55414, USA
| | - Michael LaCroix
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Institute for Translational Neuroscience, University of Minnesota, Wallin Medical Biosciences Building (Room 4-114), 2101 Sixth Street SE, CDC 2641, Minneapolis, MN, 55414, USA
- Medical Scientist Training Program, University of Texas Southwestern Medical School, Dallas, TX, 75390, USA
| | - Andrés M Baraibar
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Department of Neurosciences, University of the Basque Country UPV/EHU, Leioa, Spain
| | - Ross Pelzel
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Institute for Translational Neuroscience, University of Minnesota, Wallin Medical Biosciences Building (Room 4-114), 2101 Sixth Street SE, CDC 2641, Minneapolis, MN, 55414, USA
| | - Minwoo Kim
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Institute for Translational Neuroscience, University of Minnesota, Wallin Medical Biosciences Building (Room 4-114), 2101 Sixth Street SE, CDC 2641, Minneapolis, MN, 55414, USA
| | - Mathew A Sherman
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Institute for Translational Neuroscience, University of Minnesota, Wallin Medical Biosciences Building (Room 4-114), 2101 Sixth Street SE, CDC 2641, Minneapolis, MN, 55414, USA
| | - Samuel Boes
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Institute for Translational Neuroscience, University of Minnesota, Wallin Medical Biosciences Building (Room 4-114), 2101 Sixth Street SE, CDC 2641, Minneapolis, MN, 55414, USA
| | - Michelle Sung
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Institute for Translational Neuroscience, University of Minnesota, Wallin Medical Biosciences Building (Room 4-114), 2101 Sixth Street SE, CDC 2641, Minneapolis, MN, 55414, USA
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Tracy Cole
- Ionis Pharmaceuticals Inc., Carlsbad, CA, USA
- n-Lorem Foundation, Carlsbad, CA, 92010, USA
| | - Michael K Lee
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Institute for Translational Neuroscience, University of Minnesota, Wallin Medical Biosciences Building (Room 4-114), 2101 Sixth Street SE, CDC 2641, Minneapolis, MN, 55414, USA
| | - Alfonso Araque
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Sylvain E Lesné
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, USA.
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA.
- Institute for Translational Neuroscience, University of Minnesota, Wallin Medical Biosciences Building (Room 4-114), 2101 Sixth Street SE, CDC 2641, Minneapolis, MN, 55414, USA.
| |
Collapse
|
2
|
He L, Liu X, Li H, Dong R, Liang R, Wang R. Polyrhachis vicina Roger Alleviates Memory Impairment in a Rat Model of Alzheimer's Disease Through the EGR1/BACE1/APP Axis. ACS Chem Neurosci 2022; 13:1857-1867. [PMID: 35675207 DOI: 10.1021/acschemneuro.1c00193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Memory deficits and loss are the earliest and most prominent features of Alzheimer's disease (AD). This study was aimed to clarify the mechanistic basis of an active fraction of Polyrhachis vicina Roger (AFPR) on the memory abilities of AD rat models, which involves early growth response 1 (EGR1) expression and β-secretase 1 (BACE1)-mediated deposition of amyloid β peptide (Aβ). An AD rat model was developed by Aβ25-35, which was further treated with AFPR alone or in combination with lentiviral EGR1. The Morris water maze test and HE and Fluoro-Jade C staining were adopted to observe the memory behaviors, hippocampus neuron morphology, and Aβ deposition. Aβ25-35-induced SK-N-SH and HT22 neurons were subjected to AFPR for in vitro experiments on neuronal viability and apoptosis. AFPR improved the impaired memory function, preserved the neuron structure, and suppressed Aβ deposition in AD rat models. Further, the expression of APP pathway-related proteins was downregulated by AFPR in both rat and cellular models. Moreover, AFPR inhibited the Aβ25-35-induced neuronal apoptosis. AFPR suppressed the expression of EGR1, downregulated the BACE1 expression via impeding the binding of EGR1 to the BACE1 promoter, and thus blocked the activation of the APP signaling, ultimately protecting neurons. Notably, the aforementioned effects of AFPR were in a concentration-dependent manner; among three doses, 3.65, 15.6, and 30 mg/(kg·d), high-dose AFPR exhibited the most appreciable effects. In conclusion, AFPR inhibited the BACE1 expression by repressing the binding of EGR1 to the promoter of BACE1, thereby suppressing the Aβ deposition and improving the memory function of AD rats.
Collapse
Affiliation(s)
- Luyan He
- Department of Neurology, Cangzhou Central Hospital, Cangzhou 061000, P.R. China
| | - Xiaoman Liu
- Department of Neurology, Cangzhou Central Hospital, Cangzhou 061000, P.R. China
| | - Hualian Li
- Department of Neurology, Cangzhou Central Hospital, Cangzhou 061000, P.R. China
| | - Ruifang Dong
- Department of Neurology, Cangzhou Central Hospital, Cangzhou 061000, P.R. China
| | - Ruobing Liang
- Department of Neurology, Cangzhou Central Hospital, Cangzhou 061000, P.R. China
| | - Ruoxi Wang
- Department of Neurology, Cangzhou Central Hospital, Cangzhou 061000, P.R. China
| |
Collapse
|
3
|
Johnson AG, Webster JA, Hales CM. Glial profiling of human tauopathy brain demonstrates enrichment of astrocytic transcripts in tau-related frontotemporal degeneration. Neurobiol Aging 2022; 112:55-73. [PMID: 35051675 PMCID: PMC8976718 DOI: 10.1016/j.neurobiolaging.2021.12.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/10/2021] [Accepted: 12/17/2021] [Indexed: 01/17/2023]
Abstract
To understand how glia may be altered in frontotemporal degeneration with tau pathology (FTD-tau), we used a NanoString glial profiling panel to measure 770 transcripts related to glial biology in human control (n = 8), Alzheimer's disease (AD) (n = 8), and FTD-tau (n = 8) dorsolateral prefrontal cortex. Compared to control, 43 genes were upregulated and 86 genes were downregulated in the FTD-tau samples. Only 3 genes were upregulated and 2 were downregulated in AD. Pathway analysis revealed many astrocyte-, microglia-, and oligodendrocyte-related pathway scores increased in FTD-tau, while neuron-related pathway scores decreased. We compared these results to a previously published proteomic dataset containing many of the same samples and found that the targeted panel approach obtained measurements for genes whose proteins were not measured in the proteomics. Our results point to the utility of multiomic approaches and marked dysregulation of glia in FTD-tau.
Collapse
Affiliation(s)
- Ashlyn G Johnson
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA; Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA; Neuroscience Graduate Program, Emory University, Atlanta, GA, USA
| | - James A Webster
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA; Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Chadwick M Hales
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA; Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA; Neuroscience Graduate Program, Emory University, Atlanta, GA, USA.
| |
Collapse
|
4
|
Morin A, Mouzon B, Ferguson S, Paris D, Saltiel N, Browning M, Mullan M, Crawford F. A 3-month-delayed treatment with anatabine improves chronic outcomes in two different models of repetitive mild traumatic brain injury in hTau mice. Sci Rep 2021; 11:7900. [PMID: 33846461 PMCID: PMC8041866 DOI: 10.1038/s41598-021-87161-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 03/24/2021] [Indexed: 02/01/2023] Open
Abstract
To date, an overwhelming number of preclinical studies have addressed acute treatment in mild TBI (mTBI) and repetitive mTBI (r-mTBI), whereas, in humans, there often exists a significant time gap between the injury and the first medical intervention. Our study focused on a delayed treatment with anatabine, an anti-inflammatory compound, in hTau mice using two different models of r-mTBI. The rationale for using two models of the same impact but different frequencies (5 hit mTBI over 9 days and 24 hit mTBI over 90 days) was chosen to address the heterogeneity of r-mTBI in clinical population. Following the last injury in each model, three months elapsed before the initiation of treatment. Anatabine was administered in drinking water for 3 months thereafter. Our data demonstrated that a 3-month delayed treatment with anatabine mitigated astrogliosis in both TBI paradigms but improved cognitive functions only in more-frequently-injured mice (24 hit mTBI). We also found that anatabine decreased the phosphorylation of tau protein and NFκB, which were increased after r-mTBI in both models. The ability of anatabine to suppress these mechanisms suggests that delayed treatment can be effective for clinical population of r-mTBI. The discrepancy between the two models with regard to changes in cognitive performance suggests that r-mTBI heterogeneity may influence treatment efficiency and should be considered in therapeutic development.
Collapse
Affiliation(s)
- Alexander Morin
- The Roskamp Institute, 2040 Whitfield Ave., Sarasota, FL, 34243, USA.
- The Open University, Milton-Keynes, UK.
- James A Haley Veterans Administration, Tampa, FL, USA.
| | - Benoit Mouzon
- The Roskamp Institute, 2040 Whitfield Ave., Sarasota, FL, 34243, USA
- The Open University, Milton-Keynes, UK
- James A Haley Veterans Administration, Tampa, FL, USA
| | - Scott Ferguson
- The Roskamp Institute, 2040 Whitfield Ave., Sarasota, FL, 34243, USA
- The Open University, Milton-Keynes, UK
- James A Haley Veterans Administration, Tampa, FL, USA
| | - Daniel Paris
- The Roskamp Institute, 2040 Whitfield Ave., Sarasota, FL, 34243, USA
- The Open University, Milton-Keynes, UK
- James A Haley Veterans Administration, Tampa, FL, USA
| | - Nicole Saltiel
- The Roskamp Institute, 2040 Whitfield Ave., Sarasota, FL, 34243, USA
- James A Haley Veterans Administration, Tampa, FL, USA
| | | | - Mike Mullan
- The Roskamp Institute, 2040 Whitfield Ave., Sarasota, FL, 34243, USA
- The Open University, Milton-Keynes, UK
| | - Fiona Crawford
- The Roskamp Institute, 2040 Whitfield Ave., Sarasota, FL, 34243, USA
- The Open University, Milton-Keynes, UK
- James A Haley Veterans Administration, Tampa, FL, USA
| |
Collapse
|
5
|
Randez-Gil F, Bojunga L, Estruch F, Winderickx J, Del Poeta M, Prieto JA. Sphingolipids and Inositol Phosphates Regulate the Tau Protein Phosphorylation Status in Humanized Yeast. Front Cell Dev Biol 2020; 8:592159. [PMID: 33282871 PMCID: PMC7705114 DOI: 10.3389/fcell.2020.592159] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 10/21/2020] [Indexed: 01/08/2023] Open
Abstract
Hyperphosphorylation of protein tau is a hallmark of Alzheimer's disease (AD). Changes in energy and lipid metabolism have been correlated with the late onset of this neurological disorder. However, it is uncertain if metabolic dysregulation is a consequence of AD or one of the initiating factors of AD pathophysiology. Also, it is unclear whether variations in lipid metabolism regulate the phosphorylation state of tau. Here, we show that in humanized yeast, tau hyperphosphorylation is stimulated by glucose starvation in coincidence with the downregulation of Pho85, the yeast ortholog of CDK5. Changes in inositol phosphate (IP) signaling, which has a central role in energy metabolism, altered tau phosphorylation. Lack of inositol hexakisphosphate kinases Kcs1 and Vip1 (IP6 and IP7 kinases in mammals) increased tau hyperphosphorylation. Similar effects were found by mutation of IPK2 (inositol polyphosphate multikinase), or PLC1, the yeast phospholipase C gene. These effects may be explained by IP-mediated regulation of Pho85. Indeed, this appeared to be the case for plc1, ipk2, and kcs1. However, the effects of Vip1 on tau phosphorylation were independent of the presence of Pho85, suggesting additional mechanisms. Interestingly, kcs1 and vip1 strains, like pho85, displayed dysregulated sphingolipid (SL) metabolism. Moreover, genetic and pharmacological inhibition of SL biosynthesis stimulated the appearance of hyperphosphorylated forms of tau, while increased flux through the pathway reduced its abundance. Finally, we demonstrated that Sit4, the yeast ortholog of human PP2A protein phosphatase, is a downstream effector of SL signaling in mediating the tau phosphorylation state. Altogether, our results add new knowledge on the molecular effectors involved in tauopathies and identify new targets for pharmacological intervention.
Collapse
Affiliation(s)
- Francisca Randez-Gil
- Department of Biotechnology, Instituto de Agroquímica y Tecnología de los Alimentos, Consejo Superior de Investigaciones Científicas, Valencia, Spain
| | - Lino Bojunga
- Department of Biotechnology, Instituto de Agroquímica y Tecnología de los Alimentos, Consejo Superior de Investigaciones Científicas, Valencia, Spain
| | - Francisco Estruch
- Departament of Biochemistry and Molecular Biology, Universitat de València, Valencia, Spain
| | | | - Maurizio Del Poeta
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, NY, United States
- Veterans Administration Medical Center, Northport, NY, United States
| | - Jose A. Prieto
- Department of Biotechnology, Instituto de Agroquímica y Tecnología de los Alimentos, Consejo Superior de Investigaciones Científicas, Valencia, Spain
| |
Collapse
|
6
|
Hou TY, Zhou Y, Zhu LS, Wang X, Pang P, Wang DQ, Liuyang ZY, Man H, Lu Y, Zhu LQ, Liu D. Correcting abnormalities in miR-124/PTPN1 signaling rescues tau pathology in Alzheimer's disease. J Neurochem 2020; 154:441-457. [PMID: 31951013 DOI: 10.1111/jnc.14961] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 01/11/2020] [Accepted: 01/13/2020] [Indexed: 02/06/2023]
Abstract
MicroRNAs have been implicated in diverse physiological and pathological processes. We previously reported that aberrant microRNA-124 (miR-124)/non-receptor-type protein phosphatase 1 (PTPN1) signaling plays an important role in the synaptic disorders associated with Alzheimer's disease (AD). In this study, we further investigated the potential role of miR-124/PTPN1 in the tau pathology of AD. We first treated the mice with intra-hippocampal stereotactic injections. Then, we used quantitative real-time reverse transcription PCR (qRT-PCR) to detect the expression of microRNAs. Western blotting was used to measure the level of PTPN1, the level of tau protein, the phosphorylation of tau at AD-related sites, and alterations in the activity of glycogen synthase kinase 3β (GSK-3β) and protein phosphatase 2 (PP2A). Immunohistochemistry was also used to detect changes in tau phosphorylation levels at AD-related sites and somadendritic aggregation. Soluble and insoluble tau protein was separated by 70% formic acid (FA) extraction to examine tau solubility. Finally, behavioral experiments (including the Morris water maze, fear conditioning, and elevated plus maze) were performed to examine learning and memory ability and emotion-related behavior. We found that artificially replicating the abnormalities in miR-124/PTPN1 signaling induced AD-like tau pathology in the hippocampus of wild-type mice, including hyperphosphorylation at multiple sites, insolubility and somadendritic aggregation, as well as learning/memory deficits. We also found that disruption of miR-124/PTPN1 signaling was caused by the loss of RE1-silencing transcription factor protein, which can be initiated by Aβ insults or oxidative stress, as observed in the brains of P301S mice. Correcting the deregulation of miR-124/PTPN1 signaling rescued the tau pathology and learning/memory impairments in the P301S mice. We also found that miR-124/PTPN1 abnormalities induced activation of glycogen synthase kinase 3 (GSK-3) and inactivation of protein phosphatase 2A (PP2A) by promoting tyrosine phosphorylation, implicating an imbalance in tau kinase/phosphatase. Thus, targeting the miR-124/PTPN1 signaling pathway is a promising therapeutic strategy for AD.
Collapse
Affiliation(s)
- Tong-Yao Hou
- Department of Pathophysiology, Key Laboratory of Neurological Disorders of the Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China.,The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Yang Zhou
- Department of Pathophysiology, Key Laboratory of Neurological Disorders of the Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China.,The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Ling-Shuang Zhu
- Department of Pathophysiology, Key Laboratory of Neurological Disorders of the Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China.,The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Xiong Wang
- Department of Pathophysiology, Key Laboratory of Neurological Disorders of the Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Pei Pang
- Department of Pathophysiology, Key Laboratory of Neurological Disorders of the Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China.,The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Ding-Qi Wang
- Department of Pathophysiology, Key Laboratory of Neurological Disorders of the Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China.,The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Zhen-Yu Liuyang
- Department of Pathophysiology, Key Laboratory of Neurological Disorders of the Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Hengye Man
- Department of Biology, Boston University, Boston, MA, USA
| | - Youming Lu
- Department of Pathophysiology, Key Laboratory of Neurological Disorders of the Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China.,The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Ling-Qiang Zhu
- Department of Pathophysiology, Key Laboratory of Neurological Disorders of the Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China.,The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Dan Liu
- Department of Pathophysiology, Key Laboratory of Neurological Disorders of the Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China.,The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, P.R. China.,Department of Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| |
Collapse
|
7
|
Su D, Li W, Chi H, Yang H, She X, Wang K, Gao X, Ma K, Zhang M, Cui B. Transcriptome analysis of the hippocampus in environmental noise-exposed SAMP8 mice reveals regulatory pathways associated with Alzheimer's disease neuropathology. Environ Health Prev Med 2020; 25:3. [PMID: 31918655 PMCID: PMC6953163 DOI: 10.1186/s12199-019-0840-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 12/22/2019] [Indexed: 12/31/2022] Open
Abstract
Background Chronic noise exposure is one environmental hazard that is associated with genetic susceptibility factors that increase Alzheimer’s disease (AD) pathogenesis. However, the comprehensive understanding of the link between chronic noise stress and AD is limited. Herein, we investigated the effects of chronic noise exposure on AD-like changes in senescence-accelerated mouse prone 8 (SAMP8). Methods A total of 30 male SAMP8 mice were randomly divided into the noise-exposed group, the control group, and aging group (positive controls), and mice in the exposure group were exposed to 98 dB SPL white noise for 30 consecutive days. Transcriptome analysis and AD-like neuropathology of hippocampus were examined by RNA sequencing and immunoblotting. Enzyme-linked immunosorbent assay and real-time PCR were used to further determine the differential gene expression and explore the underlying mechanisms of chronic noise exposure in relation to AD at the genome level. Results Chronic noise exposure led to amyloid beta accumulation and increased the hyperphosphorylation of tau at the Ser202 and Ser404 sites in young SAMP8 mice; similar observations were noted in aging SAMP8 mice. We identified 21 protein-coding transcripts that were differentially expressed: 6 were downregulated and 15 were upregulated after chronic noise exposure; 8 genes were related to AD. qPCR results indicated that the expression of Arc, Egr1, Egr2, Fos, Nauk1, and Per2 were significantly high in the noise exposure group. These outcomes mirrored the results of the RNA sequencing data. Conclusions These findings further revealed that chronic noise exposure exacerbated aging-like impairment in the hippocampus of the SAMP8 mice and that the protein-coding transcripts discovered in the study may be key candidate regulators involved in environment-gene interactions.
Collapse
Affiliation(s)
- Donghong Su
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Wenlong Li
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Huimin Chi
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China.,School of Public Health and Management, Weifang Medical University, Weifang, China
| | - Honglian Yang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Xiaojun She
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Kun Wang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Xiujie Gao
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Kefeng Ma
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Ming Zhang
- Tianjin Centers for Disease Control and Prevention, Tianjin, China.
| | - Bo Cui
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China.
| |
Collapse
|
8
|
Lv X, Sun W, Zou S, Chen L, Mwacharo JM, Wang J. Characteristics of the BMP7 Promoter in Hu Sheep. Animals (Basel) 2019; 9:ani9110874. [PMID: 31661799 PMCID: PMC6912720 DOI: 10.3390/ani9110874] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 09/25/2019] [Accepted: 10/11/2019] [Indexed: 01/21/2023] Open
Abstract
Simple Summary Bone morphogenetic protein 7 (BMP7) is one of the largest secretory signal conductive molecules and is in the TGF-β superfamily. It plays an important role in the growth and development of hair follicles. We cloned the proximal promoter of the BMP7 gene for bioinformatics analysis. Dual-luciferase reporter system and overexpression were used to analyze the key regions and transcription factor binding sites. There was high activity between −758 bp and −545 bp in the core region of the gene and a possible binding site for transcription factors SP1 and EGR1. Abstract The BMP7 gene is involved in the growth and development of hair follicles but its regulation mechanism is unclear. We studied the regulation mechanism of the BMP7 promoter by cloning the proximal promoter of BMP7 for bioinformatics analysis. A series of missing vectors was then constructed for dual-fluorescein activity detection based on the bioinformatics analysis results. We tested transcription-factor binding-site mutations and transcription factor over-expression to analyze the transcriptional regulation principle of the BMP7 promoter region. The upstream transcriptional regulatory region of the BMP7 gene proximal promoter was predicted by bioinformatics. There were −1216 bp to −1166 bp and −632 bp to −582 bp transcription initiation sites in the upstream transcriptional regulatory region of the BMP7 gene proximal promoter. The CpG islands’ distribution showed that there were many CpG islands at −549 bp to 1 bp. A dual-luciferase assay revealed high activity between −758 bp and −545 bp in the core region and a possible binding site for transcription factors SP1 and EGR1. The transcriptional activity of BMP7 was significantly decreased in the transcriptional regulatory region of the BMP7 after EGR1 and SP1 mutation. Transcription was significantly enhanced by over expression of the EGR1 transcription factor, which strongly suggests that EGR1 and SP1 play important roles in BMP7 regulation.
Collapse
Affiliation(s)
- Xiaoyang Lv
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China.
| | - Wei Sun
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China.
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China.
| | - Shuangxia Zou
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China.
| | - Ling Chen
- Animal Science and Veterinary Medicine Bureau of Suzhou City, Suzhou 215200, China.
- Suzhou Stud Farm, Suzhou 215200, China.
| | - Joram M Mwacharo
- Small Ruminant Genomics Group, International Center for Agricultural Research in the Dry Areas (ICARDA), Addis Ababa P.O. BOX 5689, Ethiopia.
| | - Jinyu Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
9
|
Transcriptomics and proteomics analysis of Aβ (1-42)-induced neurotoxicity. Mol Cell Toxicol 2019. [DOI: 10.1007/s13273-019-0029-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
10
|
Zhao X, Wang X, Su G, Sun Q, Fu J, Zhang H, Teng J. The effect of early growth response 1 on levels of Amyloid-β 40 peptide in U87MG cells. J Cell Biochem 2018; 120:3514-3519. [PMID: 30548663 DOI: 10.1002/jcb.27627] [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: 06/15/2018] [Accepted: 08/14/2018] [Indexed: 11/11/2022]
Abstract
A recent study has shown that early growth response 1 (EGR1) plays a critical role in the β-amyloid cascade and tau hypotheses. In addition, evidence has suggested that EGR1 can regulate levels of amyloid-beta peptides, key molecules in the pathogenesis of Alzheimer's disease (AD). However, whether EGR1 is a deleterious or protective factor in the AD is still controversial. In this present study, we constructed an overexpression plasmid, CMV-EGFP-EGR1-Kanamycin, and transfected it into U87MG cells to investigate the effects of EGR1 expression on amyloid-β (1-40) peptide (Aβ40) levels. U87MG cells transfected by CMV-EGFP-EGR1-Kanamycin and CMV-EGFP-Kanamycin were assigned, respectively, to experimental and control groups. Fluorescence microscopy was used to observe transfection efficiencies of the plasmids after 6 hours. EGR1 messenger RNA levels were measured by quantitative reverse transcription polymerase chain reaction. Aβ40 secretion was analyzed by enzyme-linked immunosorbent assay. Expression of the amyloid precursor protein, beta-secretase enzyme, and presenilin 1 proteins were analyzed by Western blot analysis. The results showed that EGR1 overexpression increased Aβ40 secretion in vitro, possibly through increasing BACE1 expression. Based on these results, EGR1 might be a promising therapeutic target for the AD.
Collapse
Affiliation(s)
- Xinyu Zhao
- Department of Neurology, The Frist Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaojie Wang
- Department of Neurology, The Frist Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Gang Su
- Department of Neurology, The Frist Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qi Sun
- Department of Neurology, The Frist Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jitong Fu
- Department of Neurology, The Frist Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Huili Zhang
- Department of Neurology, The Frist Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Junfang Teng
- Department of Neurology, The Frist Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
11
|
Rosa P, Zerbinati C, Crestini A, Canudas AM, Ragona G, Confaloni A, Iuliano L, Calogero A. Heme Oxygenase-1 and Brain Oxysterols Metabolism Are Linked to Egr-1 Expression in Aged Mice Cortex, but Not in Hippocampus. Front Aging Neurosci 2018; 10:363. [PMID: 30459596 PMCID: PMC6232516 DOI: 10.3389/fnagi.2018.00363] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 10/23/2018] [Indexed: 01/19/2023] Open
Abstract
Throughout life, stress stimuli act upon the brain leading to morphological and functional changes in advanced age, when it is likely to develop neurodegenerative disorders. There is an increasing need to unveil the molecular mechanisms underlying aging, in a world where populations are getting older. Egr-1 (early growth response 1), a transcriptional factor involved in cell survival, proliferation and differentiation – with a role also in memory, cognition and synaptic plasticity, can be implicated in the molecular mechanism of the aging process. Moreover, Heme Oxygenase-1a (HO), a 32 kDa heat-shock protein that converts heme to iron, carbon monoxide and biliverdin, is a key enzyme with neuroprotective properties. Several in vitro and in vivo studies reported that HO-1 could regulate the metabolism of oxysterols, oxidation products of cholesterol that include markers of oxidative stress. Recently, a link between Egr-1 and HO-1 has been demonstrated in mouse lung cells exposed to cigarette smoke. In view of these data, we wanted to investigate whether Egr-1 can be implicated also in the oxysterol metabolism during brain aging. Our results show that Egr-1 expression is differently expressed in the cortex and hippocampus of old mice, as well as the oxysterol profile between these two brain areas. In particular, we show that the cortex experiences in an age-dependent fashion increasing levels of the Egr-1 protein, and that these correlate with the level of HO-1 expression and oxysterol abundance. Such a situation was not observed in the hippocampus. These results are further strenghtened by our observations made with Egr-1 KO mice, confirming our hypothesis concerning the influence of Egr-1 on oxysterol production and accumulation via regulation of the expression of HO-1 in the cortex, but not the hippocampus, of old mice. It is important to notice that most of the oxysterols involved in this process are those usually stimulated by oxidative stress, which would then represent the triggering factor for this mechanism.
Collapse
Affiliation(s)
- Paolo Rosa
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Polo Pontino, Latina, Italy
| | - Chiara Zerbinati
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Polo Pontino, Latina, Italy.,Istituto Chirurgico Ortopedico Traumatologico, ICOT, Latina, Italy
| | - Alessio Crestini
- Department of Neuroscience, Istituto Superiore di Sanità, Rome, Italy
| | - Anna-Maria Canudas
- Department of Pharmacology, Toxicology and Therapeutic Chemistry (Pharmacology Section), Institute of Neuroscience, CIBERNED, University of Barcelona, Barcelona, Spain
| | - Giuseppe Ragona
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Polo Pontino, Latina, Italy.,Istituto Chirurgico Ortopedico Traumatologico, ICOT, Latina, Italy
| | | | - Luigi Iuliano
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Polo Pontino, Latina, Italy.,Istituto Chirurgico Ortopedico Traumatologico, ICOT, Latina, Italy
| | - Antonella Calogero
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Polo Pontino, Latina, Italy.,Istituto Chirurgico Ortopedico Traumatologico, ICOT, Latina, Italy
| |
Collapse
|
12
|
Dehghani R, Rahmani F, Rezaei N. MicroRNA in Alzheimer's disease revisited: implications for major neuropathological mechanisms. Rev Neurosci 2018; 29:161-182. [PMID: 28941357 DOI: 10.1515/revneuro-2017-0042] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 07/09/2017] [Indexed: 12/28/2022]
Abstract
Pathology of Alzheimer's disease (AD) goes far beyond neurotoxicity resulting from extracellular deposition of amyloid β (Aβ) plaques. Aberrant cleavage of amyloid precursor protein and accumulation of Aβ in the form of the plaque or neurofibrillary tangles are the known primary culprits of AD pathogenesis and target for various regulatory mechanisms. Hyper-phosphorylation of tau, a major component of neurofibrillary tangles, precipitates its aggregation and prevents its clearance. Lipid particles, apolipoproteins and lipoprotein receptors can act in favor or against Aβ and tau accumulation by altering neural membrane characteristics or dynamics of transport across the blood-brain barrier. Lipids also alter the oxidative/anti-oxidative milieu of the central nervous system (CNS). Irregular cell cycle regulation, mitochondrial stress and apoptosis, which follow both, are also implicated in AD-related neuronal loss. Dysfunction in synaptic transmission and loss of neural plasticity contribute to AD. Neuroinflammation is a final trail for many of the pathologic mechanisms while playing an active role in initiation of AD pathology. Alterations in the expression of microRNAs (miRNAs) in AD and their relevance to AD pathology have long been a focus of interest. Herein we focused on the precise pathomechanisms of AD in which miRNAs were implicated. We performed literature search through PubMed and Scopus using the search term: ('Alzheimer Disease') OR ('Alzheimer's Disease') AND ('microRNAs' OR 'miRNA' OR 'MiR') to reach for relevant articles. We show how a limited number of common dysregulated pathways and abnormal mechanisms are affected by various types of miRNAs in AD brain.
Collapse
Affiliation(s)
- Reihaneh Dehghani
- Molecular Immunology Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran 1419783151, Iran
| | - Farzaneh Rahmani
- Students Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Molecular Immunology Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran 1419783151, Iran
| |
Collapse
|
13
|
Li X, Wang Z, Tong H, Yan Y, Li S. Effects of COL8A1 on the proliferation of muscle-derived satellite cells. Cell Biol Int 2018; 42:1132-1140. [PMID: 29696735 DOI: 10.1002/cbin.10979] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Accepted: 04/21/2018] [Indexed: 11/10/2022]
Abstract
Collagen type VIII alpha 1 chain (COL8A1) is a component of the extracellular matrix. Our previous studies suggested that COL8A1 is associated with the proliferation of muscle-derived satellite cells (MDSCs). Additionally, it has been demonstrated that COL8A1 promotes the proliferation of smooth muscle cells and liver cancer cells. Therefore, we predicted that COL8A1 is associated with the proliferation of bovine MDSCs, which have potential applications in research. In this study, we constructed vectors to activate and repress COL8A1 in bovine MDSCs using the CRISPR/Cas9 technique and determined the effects of COL8A1 modulation by EdU labeling, Western blotting, and dual-luciferase reporter assays. The results showed that activation of COL8A1 increased the number of EdU-positive cells and expression of the proliferation markers cyclin B1 (CCNB1) and P-AKT. The expression of P-Akt was unchanged after addition of LY294002 (a protein kinase inhibitor capable of blocking the signal transduction pathway of the phosphoinositide 3-kinase). In contrast, repression of COL8A1 reduced the number of EdU-positive cells and expression of CCNB1 and P-AKT. We also observed upregulation and downregulation of COL8A1 following the overexpression and repression of EGR1, respectively. The dual-luciferase reporter assay revealed that EGR1 regulates the promoter activity of COL8A1. To our knowledge, this is the first study demonstrating that EGR1 positively regulates the expression of COL8A1, which in turn promotes the proliferation of bovine MDSCs via the PI3 K/AKT signaling pathway.
Collapse
Affiliation(s)
- Xiaofan Li
- Laboratory of Cellular and Developmental Biology, Life Science College, North-east Agricultural University, Harbin, 150030, China
| | - Zhao Wang
- Laboratory of Cellular and Developmental Biology, Life Science College, North-east Agricultural University, Harbin, 150030, China
| | - Huili Tong
- Laboratory of Cellular and Developmental Biology, Life Science College, North-east Agricultural University, Harbin, 150030, China
| | - Yunqin Yan
- Laboratory of Cellular and Developmental Biology, Life Science College, North-east Agricultural University, Harbin, 150030, China
| | - Shufeng Li
- Laboratory of Cellular and Developmental Biology, Life Science College, North-east Agricultural University, Harbin, 150030, China
| |
Collapse
|
14
|
Cystatin C promotes tau protein phosphorylation and causes microtubule instability by inhibiting intracellular turnover of GSK3β in neurons. Mol Cell Neurosci 2018; 89:1-8. [PMID: 29577984 DOI: 10.1016/j.mcn.2018.03.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 03/16/2018] [Accepted: 03/19/2018] [Indexed: 12/20/2022] Open
Abstract
In Alzheimer's disease (AD) tau protein hyperphosphorylation causes neurofibrillary tangle formation, microtubule instability and neurodegeneration. Determining the mechanism of tau hyperphosphorylation will provide a better understanding of AD pathology. Cystatin C (CysC) is a risk factor for late-onset AD and its level is upregulated in the brains of AD patients. The role of CysC is AD pathogenesis is not known. In this study, we found that CysC level is upregulated in 3xTg-AD mouse brain. We demonstrate that CysC does not affect cellular Aβ production. However, when overexpressed in neuron (NGF-differentiated PC12 cells), CysC inhibits turnover of GSK3β, promotes GSK3β-catalyzed tau phosphorylation at Ser396/404 and causes microtubule instability. Our data provide a novel insight into the role of CysC in AD pathogenesis.
Collapse
|
15
|
Wilkaniec A, Gąssowska-Dobrowolska M, Strawski M, Adamczyk A, Czapski GA. Inhibition of cyclin-dependent kinase 5 affects early neuroinflammatory signalling in murine model of amyloid beta toxicity. J Neuroinflammation 2018; 15:1. [PMID: 29301548 PMCID: PMC5753486 DOI: 10.1186/s12974-017-1027-y] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 12/07/2017] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Cyclin-dependent kinase 5 (Cdk5) belongs to the family of proline-directed serine/threonine kinases and plays a critical role in neuronal differentiation, migration, synaptogenesis, plasticity, neurotransmission and apoptosis. The deregulation of Cdk5 activity was observed in post mortem analysis of brain tissue of Alzheimer's disease (AD) patients, suggesting the involvement of Cdk5 in the pathomechanism of this neurodegenerative disease. However, our recent study demonstrated the important function of Cdk5 in regulating inflammatory reaction. METHODS Since the role of Cdk5 in regulation of inflammatory signalling in AD is unknown, we investigated the involvement of Cdk5 in neuroinflammation induced by single intracerebroventricular (icv) injection of amyloid beta protein (Aβ) oligomers in mouse. The brain tissue was analysed up to 35 days post injection. Roscovitine (intraperitoneal administration) was used as a potent Cdk5 inhibitor. The experiments were also performed on human neuroblastoma SH-SY5Y as well as mouse BV2 cell lines treated with exogenous oligomeric Aβ. RESULTS Our results demonstrated that single injection of Aβ oligomers induces long-lasting activation of microglia and astrocytes in the hippocampus. We observed also profound, early inflammatory response in the mice hippocampus, leading to the significant elevation of pro-inflammatory cytokines expression (e.g. TNF-α, IL-1β, IL-6). Moreover, Aβ oligomers elevated the formation of truncated protein p25 in mouse hippocampus and induced overactivation of Cdk5 in neuronal cells. Importantly, administration of roscovitine reduced the inflammatory processes evoked by Aβ in the hippocampus, leading to the significant decrease of cytokines level. CONCLUSIONS These studies clearly show the involvement of Cdk5 in modulation of brain inflammatory response induced by Aβ and may indicate this kinase as a novel target for pharmacological intervention in AD.
Collapse
Affiliation(s)
- Anna Wilkaniec
- Department of Cellular Signalling, Mossakowski Medical Research Centre Polish Academy of Sciences, Pawińskiego 5, 02-106, Warsaw, Poland
| | - Magdalena Gąssowska-Dobrowolska
- Department of Cellular Signalling, Mossakowski Medical Research Centre Polish Academy of Sciences, Pawińskiego 5, 02-106, Warsaw, Poland
| | - Marcin Strawski
- Laboratory of Electrochemistry, Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093, Warsaw, Poland
| | - Agata Adamczyk
- Department of Cellular Signalling, Mossakowski Medical Research Centre Polish Academy of Sciences, Pawińskiego 5, 02-106, Warsaw, Poland
| | - Grzegorz A Czapski
- Department of Cellular Signalling, Mossakowski Medical Research Centre Polish Academy of Sciences, Pawińskiego 5, 02-106, Warsaw, Poland.
| |
Collapse
|
16
|
Recabarren-Leiva D, Alarcón M. New insights into the gene expression associated to amyotrophic lateral sclerosis. Life Sci 2018; 193:110-123. [DOI: 10.1016/j.lfs.2017.12.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 12/01/2017] [Accepted: 12/10/2017] [Indexed: 12/11/2022]
|
17
|
Qin X, Wang Y, Paudel HK. Inhibition of Early Growth Response 1 in the Hippocampus Alleviates Neuropathology and Improves Cognition in an Alzheimer Model with Plaques and Tangles. THE AMERICAN JOURNAL OF PATHOLOGY 2017. [PMID: 28641077 DOI: 10.1016/j.ajpath.2017.04.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
A sporadic form of Alzheimer disease (AD) and vascular dementia share many risk factors, and their pathogenic mechanisms are suggested to be related. Transcription factor early growth response 1 (Egr-1) regulates various vascular pathologies and is up-regulated in both AD brains and AD mouse models; however, its role in AD pathogenesis is unclear. Herein, we report that silencing of Egr-1 in the hippocampus by shRNA reduces tau phosphorylation, lowers amyloid-β (Aβ) pathology, and improves cognition in the 3xTg-AD mouse model. Egr-1 silencing does not affect levels of cyclin-dependent protein kinase 5 (Cdk5), glycogen synthase kinase 3β, protein phosphatase 1, or protein phosphatase 2A, but reduces p35 subunit of Cdk5. Egr-1 silencing also reduces levels of β-secretase 1 (BACE-1) and BACE-1-cleaved amyloid precursor protein (APP) metabolites (secreted APPβ, C99, Aβ40, and Aβ42) but has no effect on presenilin 1 and presenilin 2. In hippocampal primary neurons, Egr-1 binds to BACE-1 and p35 promoters, enhances tau phosphorylation, activates Cdk5 and BACE-1, and accelerates amyloidogenic APP processing. Blocking Cdk5 action blocks Egr-1-induced tau phosphorylation but has no effect on BACE-1 activation and amyloidogenic APP processing. Blocking BACE-1 action, on the other hand, blocks Egr-1-induced amyloidogenic APP processing but does not affect tau phosphorylation. Egr-1 regulates tau phosphorylation and Aβ synthesis in the brain by respectively controlling activities of Cdk5 and BACE-1, suggesting that Egr-1 is a potential therapeutic candidate for the treatment of AD.
Collapse
Affiliation(s)
- Xike Qin
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Yunling Wang
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Hemant K Paudel
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada; Department of Neurology and Neurosurgery, McGill University, Montréal, Quebec, Canada.
| |
Collapse
|
18
|
Cho C, MacDonald R, Shang J, Cho MJ, Chalifour LE, Paudel HK. Early growth response-1-mediated down-regulation of drebrin correlates with loss of dendritic spines. J Neurochem 2017; 142:56-73. [DOI: 10.1111/jnc.14031] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 02/28/2017] [Accepted: 03/22/2017] [Indexed: 12/16/2022]
Affiliation(s)
- Chulmin Cho
- Department of Neurology and Neurosurgery; McGill University; Montreal Quebec Canada
- Lady Davis Institute for Medical Research; Jewish General Hospital; Montreal Quebec Canada
| | - Ryen MacDonald
- Department of Neurology and Neurosurgery; McGill University; Montreal Quebec Canada
- Lady Davis Institute for Medical Research; Jewish General Hospital; Montreal Quebec Canada
| | - Jijun Shang
- Lady Davis Institute for Medical Research; Jewish General Hospital; Montreal Quebec Canada
| | - Moon Jeong Cho
- Department of Neurology and Neurosurgery; McGill University; Montreal Quebec Canada
- Lady Davis Institute for Medical Research; Jewish General Hospital; Montreal Quebec Canada
| | - Lorraine E. Chalifour
- Lady Davis Institute for Medical Research; Jewish General Hospital; Montreal Quebec Canada
- Department of Medicine; McGill University; Montreal Quebec Canada
| | - Hemant K. Paudel
- Department of Neurology and Neurosurgery; McGill University; Montreal Quebec Canada
- Lady Davis Institute for Medical Research; Jewish General Hospital; Montreal Quebec Canada
- Department of Medicine; McGill University; Montreal Quebec Canada
| |
Collapse
|
19
|
Mukherjee S, Russell JC, Carr DT, Burgess JD, Allen M, Serie DJ, Boehme KL, Kauwe JSK, Naj AC, Fardo DW, Dickson DW, Montine TJ, Ertekin-Taner N, Kaeberlein MR, Crane PK. Systems biology approach to late-onset Alzheimer's disease genome-wide association study identifies novel candidate genes validated using brain expression data and Caenorhabditis elegans experiments. Alzheimers Dement 2017; 13:1133-1142. [PMID: 28242297 DOI: 10.1016/j.jalz.2017.01.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 12/27/2016] [Accepted: 01/12/2017] [Indexed: 01/08/2023]
Abstract
INTRODUCTION We sought to determine whether a systems biology approach may identify novel late-onset Alzheimer's disease (LOAD) loci. METHODS We performed gene-wide association analyses and integrated results with human protein-protein interaction data using network analyses. We performed functional validation on novel genes using a transgenic Caenorhabditis elegans Aβ proteotoxicity model and evaluated novel genes using brain expression data from people with LOAD and other neurodegenerative conditions. RESULTS We identified 13 novel candidate LOAD genes outside chromosome 19. Of those, RNA interference knockdowns of the C. elegans orthologs of UBC, NDUFS3, EGR1, and ATP5H were associated with Aβ toxicity, and NDUFS3, SLC25A11, ATP5H, and APP were differentially expressed in the temporal cortex. DISCUSSION Network analyses identified novel LOAD candidate genes. We demonstrated a functional role for four of these in a C. elegans model and found enrichment of differentially expressed genes in the temporal cortex.
Collapse
Affiliation(s)
| | - Joshua C Russell
- Department of Pathology, University of Washington, Seattle, Washington, USA
| | - Daniel T Carr
- Department of Pathology, University of Washington, Seattle, Washington, USA
| | - Jeremy D Burgess
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, Florida, USA
| | - Mariet Allen
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, Florida, USA
| | - Daniel J Serie
- Department of Health Sciences Research, Mayo Clinic Florida, Jacksonville, Florida, USA
| | - Kevin L Boehme
- Department of Biology, Brigham Young University, Provo, Utah, USA; Department of Neuroscience, Brigham Young University, Provo, Utah, USA
| | - John S K Kauwe
- Department of Biology, Brigham Young University, Provo, Utah, USA; Department of Neuroscience, Brigham Young University, Provo, Utah, USA
| | - Adam C Naj
- Department of Biostatistics and Epidemiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - David W Fardo
- Department of Biostatistics, University of Kentucky, Lexington, Kentucky, USA
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, Florida, USA
| | - Thomas J Montine
- Department of Pathology, University of Washington, Seattle, Washington, USA
| | - Nilufer Ertekin-Taner
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, Florida, USA; Department of Neurology, Mayo Clinic Florida, Jacksonville, Florida, USA
| | - Matt R Kaeberlein
- Department of Pathology, University of Washington, Seattle, Washington, USA
| | - Paul K Crane
- Department of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
20
|
Ramljak S, Herlyn H, Zerr I. Cellular Prion Protein (PrP c) and Hypoxia: True to Each Other in Good Times and in Bad, in Sickness, and in Health. Front Cell Neurosci 2016; 10:292. [PMID: 28066187 PMCID: PMC5165248 DOI: 10.3389/fncel.2016.00292] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 12/05/2016] [Indexed: 12/20/2022] Open
Abstract
The cellular prion protein (PrPc) and hypoxia appear to be tightly intertwined. Beneficial effects of PrPc on neuronal survival under hypoxic conditions such as focal cerebral ischemia are strongly supported. Conversely, increasing evidence indicates detrimental effects of increased PrPc expression on cancer progression, another condition accompanied by low oxygen tensions. A switch between anaerobic and aerobic metabolism characterizes both conditions. A cellular process that might unite both is glycolysis. Putative role of PrPc in stimulation of glycolysis in times of need is indeed thought provoking. A significance of astrocytic PrPc expression for neuronal survival under hypoxic conditions and possible association of PrPc with the astrocyte-neuron lactate shuttle is considered. We posit PrPc-induced lactate production via transactivation of lactate dehydrogenase A by hypoxia inducible factor 1α as an important factor for survival of both neurons and tumor cells in hypoxic microenvironment. Concomitantly, we discuss a cross-talk between Wnt/β-catenin and PI3K/Akt signaling pathways in executing PrPc-induced activation of glycolysis. Finally, we would like to emphasize that we see a great potential in joining expertise from both fields, neuroscience and cancer research in revealing the mechanisms underlying hypoxia-related pathologies. PrPc may prove focal point for future research.
Collapse
Affiliation(s)
| | - Holger Herlyn
- Institute of Anthropology, Johannes Gutenberg University of Mainz Mainz, Germany
| | - Inga Zerr
- Department of Neurology, University Medical Center Göttingen and German Center for Neurodegenerative Diseases Göttingen, Germany
| |
Collapse
|
21
|
Snow WM, Albensi BC. Neuronal Gene Targets of NF-κB and Their Dysregulation in Alzheimer's Disease. Front Mol Neurosci 2016; 9:118. [PMID: 27881951 PMCID: PMC5101203 DOI: 10.3389/fnmol.2016.00118] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 10/25/2016] [Indexed: 11/21/2022] Open
Abstract
Although, better known for its role in inflammation, the transcription factor nuclear factor kappa B (NF-κB) has more recently been implicated in synaptic plasticity, learning, and memory. This has been, in part, to the discovery of its localization not just in glia, cells that are integral to mediating the inflammatory process in the brain, but also neurons. Several effectors of neuronal NF-κB have been identified, including calcium, inflammatory cytokines (i.e., tumor necrosis factor alpha), and the induction of experimental paradigms thought to reflect learning and memory at the cellular level (i.e., long-term potentiation). NF-κB is also activated after learning and memory formation in vivo. In turn, activation of NF-κB can elicit either suppression or activation of other genes. Studies are only beginning to elucidate the multitude of neuronal gene targets of NF-κB in the normal brain, but research to date has confirmed targets involved in a wide array of cellular processes, including cell signaling and growth, neurotransmission, redox signaling, and gene regulation. Further, several lines of research confirm dysregulation of NF-κB in Alzheimer's disease (AD), a disorder characterized clinically by a profound deficit in the ability to form new memories. AD-related neuropathology includes the characteristic amyloid beta plaque formation and neurofibrillary tangles. Although, such neuropathological findings have been hypothesized to contribute to memory deficits in AD, research has identified perturbations at the cellular and synaptic level that occur even prior to more gross pathologies, including transcriptional dysregulation. Indeed, synaptic disturbances appear to be a significant correlate of cognitive deficits in AD. Given the more recently identified role for NF-κB in memory and synaptic transmission in the normal brain, the expansive network of gene targets of NF-κB, and its dysregulation in AD, a thorough understanding of NF-κB-related signaling in AD is warranted and may have important implications for uncovering treatments for the disease. This review aims to provide a comprehensive view of our current understanding of the gene targets of this transcription factor in neurons in the intact brain and provide an overview of studies investigating NF-κB signaling, including its downstream targets, in the AD brain as a means of uncovering the basic physiological mechanisms by which memory becomes fragile in the disease.
Collapse
Affiliation(s)
- Wanda M Snow
- Division of Neurodegenerative Disorders, St. Boniface Hospital ResearchWinnipeg, MB, Canada; Department of Pharmacology and Therapeutics, University of ManitobaWinnipeg, MB, Canada
| | - Benedict C Albensi
- Division of Neurodegenerative Disorders, St. Boniface Hospital ResearchWinnipeg, MB, Canada; Department of Pharmacology and Therapeutics, University of ManitobaWinnipeg, MB, Canada
| |
Collapse
|
22
|
Qin X, Wang Y, Paudel HK. Early Growth Response 1 (Egr-1) Is a Transcriptional Activator of β-Secretase 1 (BACE-1) in the Brain. J Biol Chem 2016; 291:22276-22287. [PMID: 27576688 DOI: 10.1074/jbc.m116.738849] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Indexed: 11/06/2022] Open
Abstract
Accumulation of amyloid-β peptide (Aβ) in the brain is regarded as central to Alzheimer's disease (AD) pathogenesis. Aβ is generated by a sequential cleavage of amyloid precursor protein (APP) by β-secretase 1 (BACE-1) followed by γ-secretase. BACE-1 cleavage of APP is the committed step in Aβ synthesis. Understanding the mechanism by which BACE-1 is activated leading to Aβ synthesis in the brain can provide better understanding of AD pathology and help to develop novel therapies. In this study, we found that the levels of Aβ and BACE-1 are significantly reduced in the brains of mice lacking transcription factor early growth response 1 (Egr-1) when compared with the WT. We demonstrate that in COS-7 cells, Egr-1 binds to the BACE-1 promoter and activates BACE-1 transcription. In rat hippocampal primary neurons, overexpression of Egr-1 induces BACE-1 expression, activates BACE-1, promotes amyloidogenic APP processing, and enhances Aβ synthesis. In mouse hippocampal primary neurons, knockdown of BACE-1 almost completely blocks Egr-1-induced amyloidogenic APP processing and Aβ synthesis. Our data indicate that Egr-1 promotes Aβ synthesis via transcriptional activation of BACE-1 and suggest that Egr-1 plays role in activation of BACE-1 and acceleration of Aβ synthesis in AD brain. Egr-1 is a potential therapeutic target for AD.
Collapse
Affiliation(s)
- Xike Qin
- From the Lady Davis Institute for Medical Research, Jewish General Hospital, and
| | - Yunling Wang
- From the Lady Davis Institute for Medical Research, Jewish General Hospital, and
| | - Hemant K Paudel
- From the Lady Davis Institute for Medical Research, Jewish General Hospital, and the Department of Neurology and Neurosurgery, McGill University, Montréal, Quebec H4H 1R3, Canada
| |
Collapse
|
23
|
Li T, Paudel HK. 14-3-3ζ Mediates Tau Aggregation in Human Neuroblastoma M17 Cells. PLoS One 2016; 11:e0160635. [PMID: 27548710 PMCID: PMC4993442 DOI: 10.1371/journal.pone.0160635] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 07/22/2016] [Indexed: 12/20/2022] Open
Abstract
Microtubule-associated protein tau is the major component of paired helical filaments (PHFs) associated with the neuropathology of Alzheimer’s disease (AD). Tau in the normal brain binds and stabilizes microtubules. Tau isolated from PHFs is hyperphosphorylated, which prevents it from binding to microtubules. Tau phosphorylation has been suggested to be involved in the development of NFT pathology in the AD brain. Recently, we showed that 14-3-3ζ is bound to tau in the PHFs and when incubated in vitro with 14-3-3ζ, tau formed amorphous aggregates, single-stranded straight filaments, double stranded ribbon-like filaments and PHF-like filaments that displayed close resemblance with corresponding ultrastructures of AD brain. Surprisingly however, phosphorylated and non-phosphorylated tau aggregated in a similar manner, indicating that tau phosphorylation does not affect in vitro tau aggregation (Qureshi et al (2013) Biochemistry 52, 6445–6455). In this study, we have examined the role of tau phosphorylation in tau aggregation in cellular level. We have found that in human M17 neuroblastoma cells, tau phosphorylation by GSK3β or PKA does not cause tau aggregation, but promotes 14-3-3ζ-induced tau aggregation by destabilizing microtubules. Microtubule disrupting drugs also promoted 14-3-3ζ-induced tau aggregation without changing tau phosphorylation in M17 cell. In vitro, when incubated with 14-3-3ζ and microtubules, nonphosphorylated tau bound to microtubules and did not aggregate. Phosphorylated tau on the other hand did not bind to microtubules and aggregated. Our data indicate that microtubule-bound tau is resistant to 14-3-3ζ-induced tau aggregation and suggest that tau phosphorylation promotes tau aggregation in the brain by detaching tau from microtubules and thus making it accessible to 14-3-3ζ.
Collapse
Affiliation(s)
- Tong Li
- The Bloomfield Center for Research in Aging, Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada
| | - Hemant K Paudel
- The Bloomfield Center for Research in Aging, Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada.,The Department of Neurology and Neurosurgery, McGill University, Montreal, Canada
| |
Collapse
|
24
|
Zhu QB, Unmehopa U, Bossers K, Hu YT, Verwer R, Balesar R, Zhao J, Bao AM, Swaab D. MicroRNA-132 and early growth response-1 in nucleus basalis of Meynert during the course of Alzheimer's disease. Brain 2016; 139:908-21. [PMID: 26792551 DOI: 10.1093/brain/awv383] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 11/02/2015] [Indexed: 12/21/2022] Open
Abstract
The cholinergic nucleus basalis of Meynert, which is important for memory functions, shows neuronal activation ('up-phase') during the early stages of Alzheimer's disease and neurodegeneration ('down-phase') in later stages of Alzheimer's disease. MicroRNA-132 (miR-132) and the transcription factor early growth response-1 (EGR1) were proposed as possible candidate molecules regulating such an up-down activity pattern of the nucleus basalis of Meynert during the course of Alzheimer's disease, as they both show this up-down pattern of expression in the prefrontal cortex during the course of Alzheimer's disease. Not only do these two molecules stimulate synaptic activity and plasticity, they are also involved in Alzheimer's disease pathology and might, in addition, affect cholinergic function. In the nucleus basalis of Meynert, we investigated the expression of miR-132 and EGR1 along the entire course of Alzheimer's disease. Forty-nine post-mortem nucleus basalis of Meynert samples were studied, ranging from non-demented controls (Braak stage = 0) to late Alzheimer's disease patients (Braak stage = VI), and from clinical Reisberg scale 1 to 7. Each Braak stage contained seven samples, that were all well matched for confounding factors, i.e. age (range 58-91), sex, post-mortem delay, cerebrospinal fluid pH (as a measure for agonal state), APOE genotype, clock time of death, tissue fixation time, and tissue storage time. The alterations of these two molecules were studied over the course of Alzheimer's disease in relation to the expression of 4G8-stained amyloid-β, hyperphosphorylated tau stained by antibody AT8, neuronal fibrillary tangles and neuropil threads stained by silver, and in relation to alterations in choline acetyltransferase. We found that the expression of miR-132 and EGR1 in the nucleus basalis of Meynert was quite stable during the early stages of Alzheimer's disease and decreased significantly only during late Alzheimer's disease stages. In addition, miR-132 and EGR1 showed a significant positive correlation with choline acetyltransferase expression (r = 0.49, P < 0.001 and r = 0.61, P < 0.001), while choline acetyltransferase expression showed a significantly negative correlation with hyperphosphorylated tau (r = -0.33, P = 0.021) but no correlation with 4G8-stained amyloid-β. From the functional changes of miR-132 and EGR1 along the course of Alzheimer's disease we conclude: (i) that these two molecules may play a role in keeping the cholinergic function intact in early Alzheimer's disease stages; and (ii) that these molecules may contribute to the late neurodegeneration of this cholinergic nucleus.
Collapse
Affiliation(s)
- Qiong-Bin Zhu
- 1 Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, P.R. China
| | - Unga Unmehopa
- 2 Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands
| | - Koen Bossers
- 2 Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands
| | - Yu-Ting Hu
- 1 Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, P.R. China
| | - Ronald Verwer
- 2 Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands
| | - Rawien Balesar
- 2 Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands
| | - Juan Zhao
- 2 Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands
| | - Ai-Min Bao
- 1 Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, P.R. China
| | - Dick Swaab
- 1 Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, P.R. China 2 Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands
| |
Collapse
|
25
|
Qin X, Jiang Y, Tse YC, Wang Y, Wong TP, Paudel HK. Early Growth Response 1 (Egr-1) Regulates N-Methyl-d-aspartate Receptor (NMDAR)-dependent Transcription of PSD-95 and α-Amino-3-hydroxy-5-methyl-4-isoxazole Propionic Acid Receptor (AMPAR) Trafficking in Hippocampal Primary Neurons. J Biol Chem 2015; 290:29603-16. [PMID: 26475861 DOI: 10.1074/jbc.m115.668889] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Indexed: 01/07/2023] Open
Abstract
The N-methyl-d-aspartate receptor (NMDAR) controls synaptic plasticity and memory function and is one of the major inducers of transcription factor Egr-1 in the hippocampus. However, how Egr-1 mediates the NMDAR signal in neurons has remained unclear. Here, we show that the hippocampus of mice lacking Egr-1 displays electrophysiology properties and ultrastructure that are similar to mice overexpressing PSD-95, a major scaffolding protein of postsynaptic density involved in synapse formation, synaptic plasticity, and synaptic targeting of AMPA receptors (AMPARs), which mediate the vast majority of excitatory transmission in the CNS. We demonstrate that Egr-1 is a transcription repressor of the PSD-95 gene and is recruited to the PSD-95 promoter in response to NMDAR activation. Knockdown of Egr-1 in rat hippocampal primary neurons blocks NMDAR-induced PSD-95 down-regulation and AMPAR endocytosis. Likewise, overexpression of Egr-1 in rat hippocampal primary neurons causes reduction in PSD-95 protein level and promotes AMPAR endocytosis. Our data indicate that Egr-1 is involved in NMDAR-mediated PSD-95 down-regulation and AMPAR endocytosis, a process important in the expression of long term depression.
Collapse
Affiliation(s)
- Xike Qin
- From The Bloomfield Center for Research in Aging, Lady Davis Institute for Medical Research, Jewish General Hospital, and
| | - Yongjun Jiang
- From The Bloomfield Center for Research in Aging, Lady Davis Institute for Medical Research, Jewish General Hospital, and Department of Neurology and Neurosurgery
| | - Yiu Chung Tse
- Douglas Mental Health University Institute, and Department of Psychiatry, McGill University, Montréal, Quebec H4H 1R3, Canada
| | - Yunling Wang
- From The Bloomfield Center for Research in Aging, Lady Davis Institute for Medical Research, Jewish General Hospital, and
| | - Tak Pan Wong
- Douglas Mental Health University Institute, and Department of Psychiatry, McGill University, Montréal, Quebec H4H 1R3, Canada
| | - Hemant K Paudel
- From The Bloomfield Center for Research in Aging, Lady Davis Institute for Medical Research, Jewish General Hospital, and Department of Neurology and Neurosurgery,
| |
Collapse
|
26
|
Wilkaniec A, Czapski GA, Adamczyk A. Cdk5 at crossroads of protein oligomerization in neurodegenerative diseases: facts and hypotheses. J Neurochem 2015; 136:222-33. [PMID: 26376455 DOI: 10.1111/jnc.13365] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 09/02/2015] [Accepted: 09/03/2015] [Indexed: 02/06/2023]
Abstract
Cyclin-dependent kinase 5 (Cdk5) is involved in proper neurodevelopment and brain function and serves as a switch between neuronal survival and death. Overactivation of Cdk5 is associated with many neurodegenerative disorders such as Alzheimer's or Parkinson's diseases. It is believed that in those diseases Cdk5 may be an important link between disease-initiating factors and cell death effectors. A common hallmark of neurodegenerative disorders is incorrect folding of specific proteins, thus leading to their intra- and extracellular accumulation in the nervous system. Abnormal Cdk5 signaling contributes to dysfunction of individual proteins and has a substantial role in either direct or indirect interactions of proteins common to, and critical in, different neurodegenerative diseases. While the roles of Cdk5 in α-synuclein (ASN) - tau or β-amyloid peptide (Aβ) - tau interactions are well documented, its contribution to many other pertinent interactions, such as that of ASN with Aβ, or interactions of the Aβ - ASN - tau triad with prion proteins, did not get beyond plausible hypotheses and remains to be proven. Understanding of the exact position of Cdk5 in the deleterious feed-forward loop critical for development and progression of neurodegenerative diseases may help designing successful therapeutic strategies of several fatal neurodegenerative diseases. Cyclin-dependent kinase 5 (Cdk5) is associated with many neurodegenerative disorders such as Alzheimer's or Parkinson's diseases. It is believed that in those diseases Cdk5 may be an important factor involved in protein misfolding, toxicity and interaction. We suggest that Cdk5 may contribute to the vicious circle of neurotoxic events involved in the pathogenesis of different neurodegenerative diseases.
Collapse
Affiliation(s)
- Anna Wilkaniec
- Department of Cellular Signalling, Mossakowski Medical Research Centre Polish Academy of Sciences, Warsaw, Poland
| | - Grzegorz A Czapski
- Department of Cellular Signalling, Mossakowski Medical Research Centre Polish Academy of Sciences, Warsaw, Poland
| | - Agata Adamczyk
- Department of Cellular Signalling, Mossakowski Medical Research Centre Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
27
|
Expression of early growth response 1 affects miR-106a/signal transducer and activator of transcription 3 regulating cognitive impairment in ovariectomized mice. Menopause 2015; 21:1143-50. [PMID: 24686449 DOI: 10.1097/gme.0000000000000234] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
OBJECTIVE This study aims to investigate the effects of early growth response 1 (Egr1) on miR-106a/signal transducer and activator of transcription 3 (STAT3) regulating cognitive impairment in an ovariectomy model. METHODS Using the Morris water maze test, we assessed escape latency and time spent in a quadrant among mice at 6, 8, and 12 weeks after ovariectomy and their age-matched controls (n = 15 each group). Egr1, miR-106a, and STAT3 messenger RNA expression (n = 7) in the hippocampus and cortex of mice at 6, 8, and 12 weeks after ovariectomy was detected by quantitative real-time polymerase chain reaction, whereas Egr1, phospho-STAT3 (p-STAT3), and STAT3 protein expression (n = 8) was evaluated by Western blot analysis. Moreover, alterations in miR-106a and STAT3 expression were investigated in neuroblastoma (SH-SY5Y) cells transfected with a human Egr1 interference fragment (si-Egr1) or an Egr1-overexpressing plasmid (GV141-Egr1), respectively. RESULTS Escape latency was significantly increased and time spent in a platform quadrant was reduced in mice at 12 weeks after ovariectomy compared with age-matched controls. Egr1 and miR-106a expression was obviously increased in the hippocampus and cortex at 12 weeks after ovariectomy, whereas STAT3 levels were decreased compared with 12-week controls. After SH-SY5Y cell transfection with the si-Egr1 fragment, miR-106a levels decreased and STAT3/p-STAT3 levels increased, whereas cotransfection of the miR-106a mimic caused a significant decrease in STAT3 levels. MiR-106a messenger RNA expression was significantly increased and STAT3/p-STAT3 protein levels were decreased by Egr1 overexpression, whereas simultaneous transfection with the miR-106a inhibitor inhibited alterations in STAT3 levels. CONCLUSIONS This study suggests that Egr1 decreases STAT3 expression via miR-106a in ovariectomized mice with cognitive impairment, indicating that Egr1 represents a potential target for therapeutic intervention in postmenopausal cognitive decline.
Collapse
|
28
|
AC1MMYR2 impairs high dose paclitaxel-induced tumor metastasis by targeting miR-21/CDK5 axis. Cancer Lett 2015; 362:174-82. [PMID: 25827073 DOI: 10.1016/j.canlet.2015.03.038] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 03/24/2015] [Accepted: 03/24/2015] [Indexed: 12/21/2022]
Abstract
Paclitaxel (taxol) is a widely used chemo-drug for many solid tumors, while continual taxol treatment is revealed to stimulate tumor dissemination. We previously found that a small molecule inhibitor of miR-21, termed AC1MMYR2, had the potential to impair tumorigenesis and metastasis. The aim of this study was to investigate whether combining AC1MMYR2 with taxol could be explored as a means to limit tumor metastasis. Here we showed that abnormal activation of miR-21/CDK5 axis was associated with breast cancer lymph node metastasis, which was also contribute to high dose taxol-induced invasion and epithelial mesenchymal transition (EMT) in both breast cancer cell line MDA-MB-231 and glioblastoma cell line U87VIII. AC1MMYR2 attenuated CDK5 activity by functional targeting CDK5RAP1, CDK5 activator p39 and target p-FAK(ser732). A series of in vitro assays indicated that treatment of AC1MMYR2 combined with taxol suppressed tumor migration and invasion ability in both MDA-MB-231 and U87VIII cell. More importantly, combination therapy impaired high-dose taxol induced invadopodia, and EMT markers including β-catenin, E-cadherin and vimentin. Strikingly, a significant reduction of lung metastasis in mice was observed in the AC1MMYR2 plus taxol treatment. Taken together, our work demonstrated that AC1MMYR2 appeared to be a promising strategy in combating taxol induced cancer metastasis by targeting miR-21/CDK5 axis, which highlighted the potential for development of therapeutic modalities for better clinic taxol application.
Collapse
|
29
|
Xue M, Zhu L, Zhang J, Qiu J, Du G, Qiao Z, Jin G, Gao F, Zhang Q. Low dose nicotine attenuates Aβ neurotoxicity through activation early growth response gene 1 pathway. PLoS One 2015; 10:e0120267. [PMID: 25815723 PMCID: PMC4376385 DOI: 10.1371/journal.pone.0120267] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 01/21/2015] [Indexed: 02/06/2023] Open
Abstract
Epidemiological studies indicate that smoking is negatively correlated with the incidence and development of Alzheimer's disease (AD). Nicotine was reported to be the active factor. However, the detailed mechanisms still remain to be fully elucidated. Early growth response gene 1 (EGR-1) plays important roles in several important biological processes such as promoting cell growth, differentiation, anti oxidative stress, and apoptosis, but few in the pathogenesis of AD. In the present study, we show that nicotine can activate the MAPK/ERK/EGR-1 signaling pathway partially through α7 nAChR. In addition, the up-regulation of EGR-1 by nicotine can also increase the phosphorylation of CyclinD1 which contributes to the attenuation of amyloid-β (Aβ(25-35)) -induced neurotoxicity. Although nicotine and Aβ(25-35) can activate EGR-1, the expression of EGR-1 is down-regulated following treatment with nicotine and Aβ(25-35). This study demonstrates that low dose nicotine attenuates Aβ(25-35)-induced neurotoxicity in vitro and in vivo through activating EGR-1 pathway.
Collapse
Affiliation(s)
- Maoqiang Xue
- College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian, 361005, P.R.China
- Institute of Biomedical Engineering, Department of Biomaterials, College of Materials, Xiamen University, Xiamen, Fujian, 361005, P.R.China
- Department of Basic Medical Science, Medical College, Xiamen University, Xiamen, Fujian, 361005, P.R.China
| | - Liuwei Zhu
- Department of Basic Medical Science, Medical College, Xiamen University, Xiamen, Fujian, 361005, P.R.China
| | - Jie Zhang
- Department of Basic Medical Science, Medical College, Xiamen University, Xiamen, Fujian, 361005, P.R.China
| | - Jinhua Qiu
- Department of Basic Medical Science, Medical College, Xiamen University, Xiamen, Fujian, 361005, P.R.China
| | - Guicheng Du
- Department of Basic Medical Science, Medical College, Xiamen University, Xiamen, Fujian, 361005, P.R.China
| | - Zhiliang Qiao
- Department of Basic Medical Science, Medical College, Xiamen University, Xiamen, Fujian, 361005, P.R.China
| | - Guanghui Jin
- Department of Basic Medical Science, Medical College, Xiamen University, Xiamen, Fujian, 361005, P.R.China
| | - Fengguang Gao
- Department of Basic Medical Science, Medical College, Xiamen University, Xiamen, Fujian, 361005, P.R.China
| | - Qiqing Zhang
- College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian, 361005, P.R.China
- Institute of Biomedical Engineering, Department of Biomaterials, College of Materials, Xiamen University, Xiamen, Fujian, 361005, P.R.China
- Institute of Biomedical Engineering, Chinese Academy of Medical Science & Peking Union Medical College, The Key Laboratory of Biomedical Material of Tianjin, Tianjin, 300192, P.R.China
- * E-mail:
| |
Collapse
|
30
|
Over-expressed EGR1 may exaggerate ischemic injury after experimental stroke by decreasing BDNF expression. Neuroscience 2015; 290:509-17. [PMID: 25637490 DOI: 10.1016/j.neuroscience.2015.01.020] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 01/03/2015] [Accepted: 01/18/2015] [Indexed: 11/23/2022]
Abstract
PURPOSE This study aimed to clarify whether ischemia-induced early growth response 1 (EGR1) influenced the outcomes of experimental stroke by regulating brain-derived neurotrophic factor (BDNF) expression. METHODS AND RESULTS To mimic ischemia, mice were subjected to middle cerebral artery occlusion, and neurons challenged with oxygen-glucose deprivation. The expression of EGR1 was increased immediately and reached the peak 24h after reperfusion. To increase and to decrease EGR1 expressions, two types of recombinant lentiviruses were constructed. EGR1 over-expression induced by recombinant lentiviruses expanded infarct volumes and increased the numbers of terminal deoxynucleoitidyl transferase-mediated dUTP nick end labeling (TUNEL) and Fluoro-Jade C-positive cells; while decreased EGR1 expression induced by recombinant lentiviruses diminished infarct volumes and decreased the numbers of TUNEL- and Fluoro-Jade C-positive cells. Both in vitro and in vivo, increasing EGR1 expression with recombinant lentiviruses lead to decreased BDNF expressions; while silencing EGR1 expression with recombinant lentiviruses lead to increased BDNF expressions. Results from electrophoretic mobility shift assay indicated that EGR1 influenced the BDNF expression by binding to its promoter. CONCLUSION Ischemia-induced EGR1 expression may exaggerate brain injury by reducing BDNF expression. Inhibiting EGR1 may become a potential treatment for improving outcomes of ischemic stroke.
Collapse
|
31
|
Jérôme M, Paudel HK. 14-3-3ζ regulates nuclear trafficking of protein phosphatase 1α (PP1α) in HEK-293 cells. Arch Biochem Biophys 2014; 558:28-35. [DOI: 10.1016/j.abb.2014.06.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 06/09/2014] [Accepted: 06/11/2014] [Indexed: 12/24/2022]
|
32
|
Gatta V, D'Aurora M, Granzotto A, Stuppia L, Sensi SL. Early and sustained altered expression of aging-related genes in young 3xTg-AD mice. Cell Death Dis 2014; 5:e1054. [PMID: 24525730 PMCID: PMC3944230 DOI: 10.1038/cddis.2014.11] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 12/12/2013] [Accepted: 01/07/2014] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is a multifactorial neurological condition associated with a genetic profile that is still not completely understood. In this study, using a whole gene microarray approach, we investigated age-dependent gene expression profile changes occurring in the hippocampus of young and old transgenic AD (3xTg-AD) and wild-type (WT) mice. The aim of the study was to assess similarities between aging- and AD-related modifications of gene expression and investigate possible interactions between the two processes. Global gene expression profiles of hippocampal tissue obtained from 3xTg-AD and WT mice at 3 and 12 months of age (m.o.a.) were analyzed by hierarchical clustering. Interaction among transcripts was then studied with the Ingenuity Pathway Analysis (IPA) software, a tool that discloses functional networks and/or pathways associated with sets of specific genes of interest. Cluster analysis revealed the selective presence of hundreds of upregulated and downregulated transcripts. Functional analysis showed transcript involvement mainly in neuronal death and autophagy, mitochondrial functioning, intracellular calcium homeostasis, inflammatory response, dendritic spine formation, modulation of synaptic functioning, and cognitive decline. Thus, overexpression of AD-related genes (such as mutant APP, PS1, and hyperphosphorylated tau, the three genes that characterize our model) appears to favor modifications of additional genes that are involved in AD development and progression. The study also showed overlapping changes in 3xTg-AD at 3 m.o.a. and WT mice at 12 m.o.a., thereby suggesting altered expression of aging-related genes that occurs earlier in 3xTg-AD mice.
Collapse
Affiliation(s)
- V Gatta
- Functional Genetics Unit, Center of Excellence on Aging (CeSI), Chieti, Italy
- Department of Psychological Sciences, ‘G. d'Annunzio' University, Chieti, Italy
| | - M D'Aurora
- Functional Genetics Unit, Center of Excellence on Aging (CeSI), Chieti, Italy
- Department of Neuroscience and Imaging, ‘G. d'Annunzio' University, Chieti, Italy
| | - A Granzotto
- Molecular Neurology Unit, Center of Excellence on Aging (CeSI), Chieti, Italy
| | - L Stuppia
- Functional Genetics Unit, Center of Excellence on Aging (CeSI), Chieti, Italy
- Department of Psychological Sciences, ‘G. d'Annunzio' University, Chieti, Italy
| | - S L Sensi
- Department of Neuroscience and Imaging, ‘G. d'Annunzio' University, Chieti, Italy
- Molecular Neurology Unit, Center of Excellence on Aging (CeSI), Chieti, Italy
- Departments of Neurology and Pharmacology, University of California-Irvine, Irvine, CA, USA
- Institute for Memory Impairments and Neurological Disorders, University of California-Irvine, Irvine, CA, USA
| |
Collapse
|
33
|
Veyrac A, Besnard A, Caboche J, Davis S, Laroche S. The transcription factor Zif268/Egr1, brain plasticity, and memory. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 122:89-129. [PMID: 24484699 DOI: 10.1016/b978-0-12-420170-5.00004-0] [Citation(s) in RCA: 133] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
The capacity to remember our past experiences and organize our future draws on a number of cognitive processes that allow our brain to form and store neural representations that can be recalled and updated at will. In the brain, these processes require mechanisms of neural plasticity in the activated circuits, brought about by cellular and molecular changes within the neurons activated during learning. At the cellular level, a wealth of experimental data accumulated in recent years provides evidence that signaling from synapses to nucleus and the rapid regulation of the expression of immediate early genes encoding inducible, regulatory transcription factors is a key step in the mechanisms underlying synaptic plasticity and the modification of neural networks required for the laying down of memories. In the activated neurons, these transcriptional events are thought to mediate the activation of selective gene programs and subsequent synthesis of proteins, leading to stable functional and structural remodeling of the activated networks, so that the memory can later be reactivated upon recall. Over the past few decades, novel insights have been gained in identifying key transcriptional regulators that can control the genomic response of synaptically activated neurons. Here, as an example of this approach, we focus on one such activity-dependent transcription factor, Zif268, known to be implicated in neuronal plasticity and memory formation. We summarize current knowledge about the regulation and function of Zif268 in different types of brain plasticity and memory processes.
Collapse
Affiliation(s)
- Alexandra Veyrac
- CNRS, Centre de Neurosciences Paris-Sud, UMR 8195, Orsay, France; Centre de Neurosciences Paris-Sud, Univ Paris-Sud, UMR 8195, Orsay, France
| | - Antoine Besnard
- Harvard Stem Cell Institute, Harvard Medical School, Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jocelyne Caboche
- INSERM, UMRS 952, Physiopathologie des Maladies du Système Nerveux Central, Paris, France; CNRS, UMR7224, Physiopathologie des Maladies du Système Nerveux Central, Paris, France; UPMC University Paris 6, Paris, France
| | - Sabrina Davis
- CNRS, Centre de Neurosciences Paris-Sud, UMR 8195, Orsay, France; Centre de Neurosciences Paris-Sud, Univ Paris-Sud, UMR 8195, Orsay, France
| | - Serge Laroche
- CNRS, Centre de Neurosciences Paris-Sud, UMR 8195, Orsay, France; Centre de Neurosciences Paris-Sud, Univ Paris-Sud, UMR 8195, Orsay, France
| |
Collapse
|
34
|
Killick R, Ribe EM, Al-Shawi R, Malik B, Hooper C, Fernandes C, Dobson R, Nolan PM, Lourdusamy A, Furney S, Lin K, Breen G, Wroe R, To AWM, Leroy K, Causevic M, Usardi A, Robinson M, Noble W, Williamson R, Lunnon K, Kellie S, Reynolds CH, Bazenet C, Hodges A, Brion JP, Stephenson J, Paul Simons J, Lovestone S. Clusterin regulates β-amyloid toxicity via Dickkopf-1-driven induction of the wnt-PCP-JNK pathway. Mol Psychiatry 2014; 19:88-98. [PMID: 23164821 PMCID: PMC3873038 DOI: 10.1038/mp.2012.163] [Citation(s) in RCA: 174] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Revised: 10/02/2012] [Accepted: 10/09/2012] [Indexed: 02/01/2023]
Abstract
Although the mechanism of Aβ action in the pathogenesis of Alzheimer's disease (AD) has remained elusive, it is known to increase the expression of the antagonist of canonical wnt signalling, Dickkopf-1 (Dkk1), whereas the silencing of Dkk1 blocks Aβ neurotoxicity. We asked if clusterin, known to be regulated by wnt, is part of an Aβ/Dkk1 neurotoxic pathway. Knockdown of clusterin in primary neurons reduced Aβ toxicity and DKK1 upregulation and, conversely, Aβ increased intracellular clusterin and decreased clusterin protein secretion, resulting in the p53-dependent induction of DKK1. To further elucidate how the clusterin-dependent induction of Dkk1 by Aβ mediates neurotoxicity, we measured the effects of Aβ and Dkk1 protein on whole-genome expression in primary neurons, finding a common pathway suggestive of activation of wnt-planar cell polarity (PCP)-c-Jun N-terminal kinase (JNK) signalling leading to the induction of genes including EGR1 (early growth response-1), NAB2 (Ngfi-A-binding protein-2) and KLF10 (Krüppel-like factor-10) that, when individually silenced, protected against Aβ neurotoxicity and/or tau phosphorylation. Neuronal overexpression of Dkk1 in transgenic mice mimicked this Aβ-induced pathway and resulted in age-dependent increases in tau phosphorylation in hippocampus and cognitive impairment. Furthermore, we show that this Dkk1/wnt-PCP-JNK pathway is active in an Aβ-based mouse model of AD and in AD brain, but not in a tau-based mouse model or in frontotemporal dementia brain. Thus, we have identified a pathway whereby Aβ induces a clusterin/p53/Dkk1/wnt-PCP-JNK pathway, which drives the upregulation of several genes that mediate the development of AD-like neuropathologies, thereby providing new mechanistic insights into the action of Aβ in neurodegenerative diseases.
Collapse
Affiliation(s)
- R Killick
- King's College London, Institute of Psychiatry, London, UK
| | - E M Ribe
- King's College London, Institute of Psychiatry, London, UK
| | - R Al-Shawi
- Division of Medicine and Centre for Biomedical Science, University College London, London, UK
| | - B Malik
- King's College London, Institute of Psychiatry, London, UK
| | - C Hooper
- King's College London, Institute of Psychiatry, London, UK
| | - C Fernandes
- King's College London, Institute of Psychiatry, London, UK
| | - R Dobson
- King's College London, Institute of Psychiatry, London, UK
| | - P M Nolan
- MRC Harwell, Mammalian Genetics Unit, Harwell Science and Innovation Campus, Oxfordshire, UK
| | - A Lourdusamy
- King's College London, Institute of Psychiatry, London, UK
| | - S Furney
- King's College London, Institute of Psychiatry, London, UK
| | - K Lin
- King's College London, Institute of Psychiatry, London, UK
| | - G Breen
- King's College London, Institute of Psychiatry, London, UK
| | - R Wroe
- King's College London, Institute of Psychiatry, London, UK
| | - A W M To
- King's College London, Institute of Psychiatry, London, UK
| | - K Leroy
- Université Libre de Bruxelles, Faculté de Médecine, Brussels, Belgium
| | - M Causevic
- King's College London, Institute of Psychiatry, London, UK
| | - A Usardi
- King's College London, Institute of Psychiatry, London, UK
| | - M Robinson
- King's College London, Institute of Psychiatry, London, UK
| | - W Noble
- King's College London, Institute of Psychiatry, London, UK
| | - R Williamson
- King's College London, Institute of Psychiatry, London, UK
| | - K Lunnon
- Division of Medicine and Centre for Biomedical Science, University College London, London, UK
| | - S Kellie
- University of Queensland, School of Chemistry and Molecular Biosciences, Brisbane, Queensland, Australia
| | - C H Reynolds
- King's College London, Institute of Psychiatry, London, UK
| | - C Bazenet
- King's College London, Institute of Psychiatry, London, UK
| | - A Hodges
- King's College London, Institute of Psychiatry, London, UK
| | - J-P Brion
- Université Libre de Bruxelles, Faculté de Médecine, Brussels, Belgium
| | - J Stephenson
- King's College London, Institute of Psychiatry, London, UK
| | - J Paul Simons
- Division of Medicine and Centre for Biomedical Science, University College London, London, UK
| | | |
Collapse
|
35
|
Overexpression of 14-3-3z promotes tau phosphorylation at Ser262 and accelerates proteosomal degradation of synaptophysin in rat primary hippocampal neurons. PLoS One 2013; 8:e84615. [PMID: 24367683 PMCID: PMC3868614 DOI: 10.1371/journal.pone.0084615] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Accepted: 11/15/2013] [Indexed: 01/09/2023] Open
Abstract
β-amyloid peptide accumulation, tau hyperphosphorylation, and synapse loss are characteristic neuropathological symptoms of Alzheimer’s disease (AD). Tau hyperphosphorylation is suggested to inhibit the association of tau with microtubules, making microtubules unstable and causing neurodegeneration. The mechanism of tau phosphorylation in AD brain, therefore, is of considerable significance. Although PHF-tau is phosphorylated at over 40 Ser/Thr sites, Ser262 phosphorylation was shown to mediate β-amyloid neurotoxicity and formation of toxic tau lesions in the brain. In vitro, PKA is one of the kinases that phosphorylates tau at Ser262, but the mechanism by which it phosphorylates tau in AD brain is not very clear. 14-3-3ζ is associated with neurofibrillary tangles and is upregulated in AD brain. In this study, we show that 14-3-3ζ promotes tau phosphorylation at Ser262 by PKA in differentiating neurons. When overexpressed in rat hippocampal primary neurons, 14-3-3ζ causes an increase in Ser262 phosphorylation, a decrease in the amount of microtubule-bound tau, a reduction in the amount of polymerized microtubules, as well as microtubule instability. More importantly, the level of pre-synaptic protein synaptophysin was significantly reduced. Downregulation of synaptophysin in 14-3-3ζ overexpressing neurons was mitigated by inhibiting the proteosome, indicating that 14-3-3ζ promotes proteosomal degradation of synaptophysin. When 14-3-3ζ overexpressing neurons were treated with the microtubule stabilizing drug taxol, tau Ser262 phosphorylation decreased and synaptophysin level was restored. Our data demonstrate that overexpression of 14-3-3ζ accelerates proteosomal turnover of synaptophysin by promoting the destabilization of microtubules. Synaptophysin is involved in synapse formation and neurotransmitter release. Our results suggest that 14-3-3ζ may cause synaptic pathology by reducing synaptophysin levels in the brains of patients suffering from AD.
Collapse
|
36
|
Ramanan VK, Saykin AJ. Pathways to neurodegeneration: mechanistic insights from GWAS in Alzheimer's disease, Parkinson's disease, and related disorders. AMERICAN JOURNAL OF NEURODEGENERATIVE DISEASE 2013; 2:145-175. [PMID: 24093081 PMCID: PMC3783830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 08/25/2013] [Indexed: 06/02/2023]
Abstract
The discovery of causative genetic mutations in affected family members has historically dominated our understanding of neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), frontotemporal dementia (FTD), and amyotrophic lateral sclerosis (ALS). Nevertheless, most cases of neurodegenerative disease are not explained by Mendelian inheritance of known genetic variants, but instead are thought to have a complex etiology with numerous genetic and environmental factors contributing to susceptibility. Although unbiased genome-wide association studies (GWAS) have identified novel associations to neurodegenerative diseases, most of these hits explain only modest fractions of disease heritability. In addition, despite the substantial overlap of clinical and pathologic features among major neurodegenerative diseases, surprisingly few GWAS-implicated variants appear to exhibit cross-disease association. These realities suggest limitations of the focus on individual genetic variants and create challenges for the development of diagnostic and therapeutic strategies, which traditionally target an isolated molecule or mechanistic step. Recently, GWAS of complex diseases and traits have focused less on individual susceptibility variants and instead have emphasized the biological pathways and networks revealed by genetic associations. This new paradigm draws on the hypothesis that fundamental disease processes may be influenced on a personalized basis by a combination of variants - some common and others rare, some protective and others deleterious - in key genes and pathways. Here, we review and synthesize the major pathways implicated in neurodegeneration, focusing on GWAS from the most prevalent neurodegenerative disorders, AD and PD. Using literature mining, we also discover a novel regulatory network that is enriched with AD- and PD-associated genes and centered on the SP1 and AP-1 (Jun/Fos) transcription factors. Overall, this pathway- and network-driven model highlights several potential shared mechanisms in AD and PD that will inform future studies of these and other neurodegenerative disorders. These insights also suggest that biomarker and treatment strategies may require simultaneous targeting of multiple components, including some specific to disease stage, in order to assess and modulate neurodegeneration. Pathways and networks will provide ideal vehicles for integrating relevant findings from GWAS and other modalities to enhance clinical translation.
Collapse
Affiliation(s)
- Vijay K Ramanan
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of MedicineIndianapolis, IN, USA
- Department of Medical and Molecular Genetics, Indiana University School of MedicineIndianapolis, IN, USA
- Medical Scientist Training Program, Indiana University School of MedicineIndianapolis, IN, USA
| | - Andrew J Saykin
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of MedicineIndianapolis, IN, USA
- Department of Medical and Molecular Genetics, Indiana University School of MedicineIndianapolis, IN, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of MedicineIndianapolis, IN, USA
- Indiana Alzheimer Disease Center, Indiana University School of MedicineIndianapolis, IN, USA
| |
Collapse
|
37
|
Epigenetic induction of EGR-1 expression by the amyloid precursor protein during exposure to novelty. PLoS One 2013; 8:e74305. [PMID: 24066134 PMCID: PMC3774615 DOI: 10.1371/journal.pone.0074305] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Accepted: 07/30/2013] [Indexed: 11/19/2022] Open
Abstract
Following transcriptome comparison of primary cultures isolated from brain of mice expressing or not the amyloid precursor protein APP, we found transcription of the EGR-1 gene to be regulated by APP. In primary cultures of cortical neurons, APP significantly down regulated EGR-1 expression at both mRNA and protein levels in a γ-secretase independent manner. The intracellular domain of APP did not interact with EGR-1 gene promoter, but enrichment of acetylated histone H4 at the EGR-1 promoter region was measured in APP-/- neurons, as well as in brain of APP-/- mice, in which increase in EGR-1 expression was also measured. These results argue for an important function of APP in the epigenetic regulation of EGR-1 gene transcription both in vitro and in vivo. In APP-/- mice, constitutive overexpression of EGR-1 in brain impaired epigenetic induction of this early transcriptional regulator during exposure to novelty. Altogether, these results indicate an important function of APP in the epigenetic regulation of the transcription of EGR-1, known to be important for memory formation.
Collapse
|
38
|
Qureshi HY, Li T, MacDonald R, Cho CM, Leclerc N, Paudel HK. Interaction of 14-3-3ζ with microtubule-associated protein tau within Alzheimer's disease neurofibrillary tangles. Biochemistry 2013; 52:6445-55. [PMID: 23962087 DOI: 10.1021/bi400442d] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Alzheimer's disease (AD) is characterized by the presence of abnormal, straight filaments and paired helical filaments (PHFs) that are coated with amorphous aggregates. When PHFs are treated with alkali, they untwist and form filaments with a ribbonlike morphology. Tau protein is the major component of all of these ultrastructures. 14-3-3ζ is present in NFTs and is significantly upregulated in AD brain. The molecular basis of the association of 14-3-3ζ within NFTs and the pathological significance of its association are not known. In this study, we have found that 14-3-3ζ is copurified and co-immunoprecipitates with tau from NFTs of AD brain extract. In vitro, tau binds to both phosphorylated and nonphosphorylated tau. When incubated with 14-3-3ζ, tau forms amorphous aggregates, single-stranded, straight filaments, ribbonlike filaments, and PHF-like filaments, all of which resemble the corresponding ultrastructures found in AD brain. Immuno-electron microscopy determined that both tau and 14-3-3ζ are present in these ultrastructures and that they are formed in an incubation time-dependent manner. Amorphous aggregates are formed first. As the incubation time increases, the size of amorphous aggregates increases and they are incorporated into single-stranded filaments. Single-stranded filaments laterally associate to form double-stranded, ribbonlike, and PHF-like filaments. Both tau and phosphorylated tau aggregate in a similar manner when they are incubated with 14-3-3ζ. Our data suggest that 14-3-3ζ has a role in the fibrillization of tau in AD brain, and that tau phosphorylation does not affect 14-3-3ζ-induced tau aggregation.
Collapse
Affiliation(s)
- Hamid Y Qureshi
- The Bloomfield Center for Research in Aging, Lady Davis Institute for Medical Research, Jewish General Hospital , 3755 Côte-Sainte-Catherine Road, Montreal, Quebec, Canada H3T 1E2
| | | | | | | | | | | |
Collapse
|
39
|
Twine NA, Janitz C, Wilkins MR, Janitz M. Sequencing of hippocampal and cerebellar transcriptomes provides new insights into the complexity of gene regulation in the human brain. Neurosci Lett 2013; 541:263-8. [PMID: 23454452 DOI: 10.1016/j.neulet.2013.02.034] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2012] [Revised: 02/13/2013] [Accepted: 02/17/2013] [Indexed: 11/28/2022]
Abstract
The hippocampus and cerebellum represent anatomically and functionally distinct parts of the human brain. The RNA-Seq technique makes it possible to investigate the human transcriptome with unprecedented resolution, allowing identification of differential mRNA splicing and promoter usage on a genome-wide scale. We undertook whole-mRNA sequencing of samples from the human hippocampus and cerebellum. A bioinformatic analysis revealed distinct expression patterns of genes related to the molecular physiology of neurons and glial cells. Upregulated genes in hippocampal tissue included serpin peptidase inhibitor, clade A (SERPINA3), lymphocyte antigen 6 complex, locus H (LY6H) and transthyretin (TTR). In cerebellum, the cerebellin 3 precursor (CLBN3) and Zic family member 4 (ZIC4) genes were significantly upregulated. These changes were validated in independent donor samples by qRT-PCR. The hippocampus and the cerebellum showed striking differences in splicing patterns and promoter usage. A notable example of this was the gene for NGFI-A binding protein 2 (NAB2), which displayed tissue-specific isoforms which may affect its function as a transcriptional repressor.
Collapse
Affiliation(s)
- Natalie A Twine
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | | | | | | |
Collapse
|
40
|
Hashiguchi M, Hashiguchi T. Kinase–Kinase Interaction and Modulation of Tau Phosphorylation. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2013; 300:121-60. [DOI: 10.1016/b978-0-12-405210-9.00004-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
41
|
Aso E, Lomoio S, López‐González I, Joda L, Carmona M, Fernández‐Yagüe N, Moreno J, Juvés S, Pujol A, Pamplona R, Portero‐Otin M, Martín V, Díaz M, Ferrer I. Amyloid generation and dysfunctional immunoproteasome activation with disease progression in animal model of familial Alzheimer's disease. Brain Pathol 2012; 22:636-53. [PMID: 22188425 PMCID: PMC8057644 DOI: 10.1111/j.1750-3639.2011.00560.x] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2011] [Accepted: 12/12/2011] [Indexed: 12/16/2022] Open
Abstract
Double-transgenic amyloid precursor protein/presenilin 1 (APP/PS1) mice express a chimeric mouse/human APP bearing the Swedish mutation (Mo/HuAPP695swe) and a mutant human PS1-dE9 both causative of familial Alzheimer's disease (FAD). Transgenic mice show impaired memory and learning performance from the age of 6 months onwards. Double-transgenic APP/PS1 mice express altered APP and PS1 mRNAs and proteins, reduced β-secretase 1 (BACE1) mRNA and normal BACE1 protein, all of which suggest a particular mechanism of amyloidogenesis when compared with sporadic AD. The first β-amyloid plaques in APP/PS1 mice appear at 3 months, and they increase in number and distribution with disease progression in parallel with increased levels of brain soluble β-amyloid 1-42 and 1-40, but also with reduced 1-42/1-40 ratio with age. Amyloid deposition in plaques is accompanied by altered mitochondria and increased oxidative damage, post-translational modifications and accumulation of altered proteins at the dystrophic neurites surrounding plaques. Degradation pathways are also modified with disease progression including activation of the immunoproteasome together with variable alterations of the different protease activities of the ubiquitin-proteasome system. Present observations show modifications in the production of β-amyloid and activation and malfunction of the subcellular degradation pathways that have general implications in the pathogenesis of AD and more particularly in specificities of FAD amyloidogenesis.
Collapse
MESH Headings
- Age Factors
- Alzheimer Disease/complications
- Alzheimer Disease/genetics
- Alzheimer Disease/pathology
- Amyloid beta-Peptides/metabolism
- Amyloid beta-Protein Precursor/genetics
- Animals
- Aspartic Acid Endopeptidases/genetics
- Aspartic Acid Endopeptidases/metabolism
- Avoidance Learning/physiology
- Brain/metabolism
- Brain/pathology
- Brain/ultrastructure
- Cognition Disorders/etiology
- Cysteine Endopeptidases/metabolism
- Disease Models, Animal
- Disease Progression
- Gene Expression Regulation/genetics
- Humans
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Microscopy, Electron, Transmission
- Microscopy, Immunoelectron
- Mitogen-Activated Protein Kinases
- Mutation/genetics
- Neuropsychological Tests
- Plaque, Amyloid/metabolism
- Plaque, Amyloid/pathology
- Plaque, Amyloid/ultrastructure
- Presenilin-1/genetics
- Proteasome Endopeptidase Complex/genetics
- Proteasome Endopeptidase Complex/metabolism
- RNA, Messenger/metabolism
- Recognition, Psychology/physiology
- Signal Transduction/genetics
- Superoxide Dismutase/metabolism
- Superoxide Dismutase-1
- Ubiquitin Thiolesterase/metabolism
- alpha-Synuclein/metabolism
- tau Proteins/metabolism
Collapse
Affiliation(s)
- Ester Aso
- Institut de Neuropatologia, IDIBELL‐Hospital Universitari de Bellvitge, Universitat de Barcelona, Hospitalet de Llobregat; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Selene Lomoio
- Institut de Neuropatologia, IDIBELL‐Hospital Universitari de Bellvitge, Universitat de Barcelona, Hospitalet de Llobregat; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
- Dipartimento di Biologia Animale, Laboratorio di Biologia Cellulare e Neurobiologia, Università di Pavia, Pavia, Italy
| | - Irene López‐González
- Institut de Neuropatologia, IDIBELL‐Hospital Universitari de Bellvitge, Universitat de Barcelona, Hospitalet de Llobregat; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Laura Joda
- Institut de Neuropatologia, IDIBELL‐Hospital Universitari de Bellvitge, Universitat de Barcelona, Hospitalet de Llobregat; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Margarita Carmona
- Institut de Neuropatologia, IDIBELL‐Hospital Universitari de Bellvitge, Universitat de Barcelona, Hospitalet de Llobregat; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Núria Fernández‐Yagüe
- Institut de Neuropatologia, IDIBELL‐Hospital Universitari de Bellvitge, Universitat de Barcelona, Hospitalet de Llobregat; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Jesús Moreno
- Institut de Neuropatologia, IDIBELL‐Hospital Universitari de Bellvitge, Universitat de Barcelona, Hospitalet de Llobregat; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Salvador Juvés
- Institut de Neuropatologia, IDIBELL‐Hospital Universitari de Bellvitge, Universitat de Barcelona, Hospitalet de Llobregat; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Aurora Pujol
- Catalan Institution of Research and Advanced Studies (ICREA), Centre de Genètica Mèdica i Molecular, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Reinald Pamplona
- Departament de Medicina Experimental, Universitat de Lleida‐IRBLleida, Lleida, Spain
| | - Manuel Portero‐Otin
- Departament de Medicina Experimental, Universitat de Lleida‐IRBLleida, Lleida, Spain
| | - Virginia Martín
- Laboratorio de Fisiología y Biofísica de Membranas, Departamento de Biología Animal e Instituto de Tecnologías Biomédicas, Universidad de La Laguna, Tenerife, Spain
| | - Mario Díaz
- Laboratorio de Fisiología y Biofísica de Membranas, Departamento de Biología Animal e Instituto de Tecnologías Biomédicas, Universidad de La Laguna, Tenerife, Spain
| | - Isidro Ferrer
- Institut de Neuropatologia, IDIBELL‐Hospital Universitari de Bellvitge, Universitat de Barcelona, Hospitalet de Llobregat; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| |
Collapse
|
42
|
c-Jun induction is independent of early growth response factor during cerebellar granule neuron apoptosis. Neuroreport 2012; 23:67-72. [DOI: 10.1097/wnr.0b013e32834e7d69] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
43
|
Friocourt G, Parnavelas JG. Identification of Arx targets unveils new candidates for controlling cortical interneuron migration and differentiation. Front Cell Neurosci 2011; 5:28. [PMID: 22355284 PMCID: PMC3280452 DOI: 10.3389/fncel.2011.00028] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2011] [Accepted: 12/08/2011] [Indexed: 11/13/2022] Open
Abstract
Mutations in the homeobox transcription factor ARX have been found to be responsible for a wide spectrum of disorders extending from phenotypes with severe neuronal migration defects, such as lissencephaly, to mild forms of intellectual disabilities without apparent brain abnormalities, but with associated features of dystonia and epilepsy. Arx expression is mainly restricted to populations of GABA-containing neurons. Studies of the effects of ARX loss of function, either in humans or mutant mice, revealed varying defects, suggesting multiple roles of this gene in brain patterning, neuronal proliferation and migration, cell maturation and differentiation, as well as axonal outgrowth and connectivity. However, to date, little is known about how Arx functions as a transcription factor or which genes it binds and regulates. Recently, we combined chromatin immunoprecipitation and mRNA expression with microarray analysis and identified approximately 1000 gene promoters bound by Arx in transfected neuroblastoma N2a cells and mouse embryonic brain. To narrow the analysis of Arx targets to those most likely to control cortical interneuron migration and/or differentiation, we compare here our data to previously published studies searching for genes enriched or down-regulated in cortical interneurons between E13.5 and E15.5. We thus identified 14 Arx-target genes enriched (Cxcr7, Meis1, Ppap2a, Slc 12a5, Ets2, Phlda1, Egr1, Igf1, Lmo3, Sema6, Lgi1, Alk, Tgfb3, and Napb) and 5 genes specifically down-regulated (Hmgn3, Lmo1, Ebf3, Rasgef1b, and Slit2) in cortical migrating neurons. In this review, we present these genes and discuss how their possible regulation by Arx may lead to the dysfunction of GABAergic neurons, resulting in mental retardation and epilepsy.
Collapse
Affiliation(s)
- Gaëlle Friocourt
- Laboratory of Molecular Genetics and Histocompatibility Inserm U613, Brest, France
| | | |
Collapse
|
44
|
Cheung ZH, Ip NY. Cdk5: a multifaceted kinase in neurodegenerative diseases. Trends Cell Biol 2011; 22:169-75. [PMID: 22189166 DOI: 10.1016/j.tcb.2011.11.003] [Citation(s) in RCA: 184] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2011] [Revised: 11/09/2011] [Accepted: 11/14/2011] [Indexed: 10/14/2022]
Abstract
Since the identification of cyclin-dependent kinase-5 (Cdk5) as a tau kinase and member of the Cdk family almost 20 years ago, deregulation of Cdk5 activity has been linked to an array of neurodegenerative diseases. As knowledge on the etiopathological mechanisms of these diseases evolved through the years, Cdk5 has also been implicated in additional cellular events that are affected under these pathological conditions. From the role of Cdk5 in the regulation of synaptic functions to its involvement in autophagy deregulation, significant insights have been obtained regarding the role of Cdk5 as a key regulator of neurodegeneration. Here, we summarize recent findings on the involvement of Cdk5 in the pathophysiological mechanisms underlying various neurodegenerative diseases.
Collapse
Affiliation(s)
- Zelda H Cheung
- Division of Life Science, Molecular Neuroscience Center and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | | |
Collapse
|