151
|
Zanardini R, Ciani M, Benussi L, Ghidoni R. Molecular Pathways Bridging Frontotemporal Lobar Degeneration and Psychiatric Disorders. Front Aging Neurosci 2016; 8:10. [PMID: 26869919 PMCID: PMC4740789 DOI: 10.3389/fnagi.2016.00010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 01/12/2016] [Indexed: 12/12/2022] Open
Abstract
The overlap of symptoms between neurodegenerative and psychiatric diseases has been reported. Neuropsychiatric alterations are commonly observed in dementia, especially in the behavioral variant of frontotemporal dementia (bvFTD), which is the most common clinical FTD subtype. At the same time, psychiatric disorders, like schizophrenia (SCZ), can display symptoms of dementia, including features of frontal dysfunction with relative sparing of memory. In the present review, we discuss common molecular features in these pathologies with a special focus on FTD. Molecules like Brain Derived Neurotrophic Factor (BDNF) and progranulin are linked to the pathophysiology of both neurodegenerative and psychiatric diseases. In these brain-associated illnesses, the presence of disease-associated variants in BDNF and progranulin (GRN) genes cause a reduction of circulating proteins levels, through alterations in proteins expression or secretion. For these reasons, we believe that prevention and therapy of psychiatric and neurological disorders could be achieved enhancing both BDNF and progranulin levels thanks to drug discovery efforts.
Collapse
Affiliation(s)
- Roberta Zanardini
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio, Fatebenefratelli Brescia, Italy
| | - Miriam Ciani
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio, Fatebenefratelli Brescia, Italy
| | - Luisa Benussi
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio, Fatebenefratelli Brescia, Italy
| | - Roberta Ghidoni
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio, Fatebenefratelli Brescia, Italy
| |
Collapse
|
152
|
Pang PT, Nagappan G, Guo W, Lu B. Extracellular and intracellular cleavages of proBDNF required at two distinct stages of late-phase LTP. NPJ SCIENCE OF LEARNING 2016; 1:16003. [PMID: 30792890 PMCID: PMC6380376 DOI: 10.1038/npjscilearn.2016.3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 02/18/2016] [Accepted: 03/06/2016] [Indexed: 05/07/2023]
Abstract
Although late-phase long-term potentiation (L-LTP) is implicated in long-term memory, its molecular mechanisms are largely unknown. Here we provide evidence that L-LTP can be divided into two stages: an induction stage (I) and a maintenance stage (II). Both stages require mature brain-derived neurotrophic factor (mBDNF), but involve distinct underlying mechanisms. Stage I requires secretion of existing proBDNF followed by extracellular cleavage by tPA/plasmin. Stage II depends on newly synthesized BDNF. Surprisingly, mBDNF at stage II is derived from intracellular cleavage of proBDNF by furin/PC1. Moreover, stage I involves BDNF-TrkB signaling mainly through MAP kinase, whereas all three signaling pathways (phospholipase C-γ, PI3 kinase, and MAP kinase) are required for the maintenance of L-LTP at stage II. These results reveal the molecular basis for two temporally distinct stages in L-LTP, and provide insights on how BDNF modulates this long-lasting synaptic alternation at two critical time windows.
Collapse
Affiliation(s)
- Petti T Pang
- National Institute of Child Health and Human Development, Bethesda, MD, USA
- Sanofi-Genzyme, Framingham, MA, USA
| | - Guhan Nagappan
- National Institute of Child Health and Human Development, Bethesda, MD, USA
- GlaxoSmithKline, R&D China, Shanghai, China
| | - Wei Guo
- School of Medicine, Tsinghua Univ., Beijing, China
| | - Bai Lu
- National Institute of Child Health and Human Development, Bethesda, MD, USA
- School of Medicine, Tsinghua Univ., Beijing, China
- ()
| |
Collapse
|
153
|
Ma Q, Chen C, Deng P, Zhu G, Lin M, Zhang L, Xu S, He M, Lu Y, Duan W, Pi H, Cao Z, Pei L, Li M, Liu C, Zhang Y, Zhong M, Zhou Z, Yu Z. Extremely Low-Frequency Electromagnetic Fields Promote In Vitro Neuronal Differentiation and Neurite Outgrowth of Embryonic Neural Stem Cells via Up-Regulating TRPC1. PLoS One 2016; 11:e0150923. [PMID: 26950212 PMCID: PMC4780708 DOI: 10.1371/journal.pone.0150923] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 02/22/2016] [Indexed: 02/07/2023] Open
Abstract
Exposure to extremely low-frequency electromagnetic fields (ELF-EMFs) can enhance hippocampal neurogenesis in adult mice. However, little is focused on the effects of ELF-EMFs on embryonic neurogenesis. Here, we studied the potential effects of ELF-EMFs on embryonic neural stem cells (eNSCs). We exposed eNSCs to ELF-EMF (50 Hz, 1 mT) for 1, 2, and 3 days with 4 hours per day. We found that eNSC proliferation and maintenance were significantly enhanced after ELF-EMF exposure in proliferation medium. ELF-EMF exposure increased the ratio of differentiated neurons and promoted the neurite outgrowth of eNSC-derived neurons without influencing astrocyes differentiation and the cell apoptosis. In addition, the expression of the proneural genes, NeuroD and Ngn1, which are crucial for neuronal differentiation and neurite outgrowth, was increased after ELF-EMF exposure. Moreover, the expression of transient receptor potential canonical 1 (TRPC1) was significantly up-regulated accompanied by increased the peak amplitude of intracellular calcium level induced by ELF-EMF. Furthermore, silencing TRPC1 expression eliminated the up-regulation of the proneural genes and the promotion of neuronal differentiation and neurite outgrowth induced by ELF-EMF. These results suggest that ELF-EMF exposure promotes the neuronal differentiation and neurite outgrowth of eNSCs via up-regulation the expression of TRPC1 and proneural genes (NeuroD and Ngn1). These findings also provide new insights in understanding the effects of ELF-EMF exposure on embryonic brain development.
Collapse
Affiliation(s)
- Qinlong Ma
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Chunhai Chen
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
- Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education of China, Third Military Medical University, Chongqing, China
| | - Ping Deng
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Gang Zhu
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Min Lin
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Lei Zhang
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
- Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education of China, Third Military Medical University, Chongqing, China
| | - Shangcheng Xu
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
- Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education of China, Third Military Medical University, Chongqing, China
| | - Mindi He
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
- Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education of China, Third Military Medical University, Chongqing, China
| | - Yonghui Lu
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
- Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education of China, Third Military Medical University, Chongqing, China
| | - Weixia Duan
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Huifeng Pi
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Zhengwang Cao
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Liping Pei
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
- Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education of China, Third Military Medical University, Chongqing, China
| | - Min Li
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Chuan Liu
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Yanwen Zhang
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
- Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education of China, Third Military Medical University, Chongqing, China
| | - Min Zhong
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
- Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education of China, Third Military Medical University, Chongqing, China
| | - Zhou Zhou
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
- Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education of China, Third Military Medical University, Chongqing, China
- * E-mail: (ZY); (ZZ)
| | - Zhengping Yu
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
- Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education of China, Third Military Medical University, Chongqing, China
- * E-mail: (ZY); (ZZ)
| |
Collapse
|
154
|
Roumes H, Brossaud J, Lemelletier A, Moisan MP, Pallet V, Redonnet A, Corcuff JB. Retinoids and glucocorticoids have opposite effects on actin cytoskeleton rearrangement in hippocampal HT22 cells. Int J Biochem Cell Biol 2015; 71:102-110. [PMID: 26748244 DOI: 10.1016/j.biocel.2015.12.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 12/18/2015] [Accepted: 12/28/2015] [Indexed: 12/12/2022]
Abstract
A chronic excess of glucocorticoids elicits deleterious effects in the hippocampus. Conversely, retinoic acid plays a major role in aging brain plasticity. As synaptic plasticity depends on mechanisms related to cell morphology, we investigated the involvement of retinoic acid and glucocorticoids in the remodelling of the HT22 neurons actin cytoskeleton. Cells exhibited a significantly more elongated shape with retinoic acid and a rounder shape with dexamethasone; retinoic acid reversed the effects of dexamethasone. Actin expression and abundance were unchanged by retinoic acid or dexamethasone but F-actin organization was dramatically modified. Indeed, retinoic acid and dexamethasone increased (70 ± 7% and 176 ± 5%) cortical actin while retinoic acid suppressed the effect of dexamethasone (90 ± 6%). Retinoic acid decreased (-22 ± 9%) and dexamethasone increased (134 ± 16%) actin stress fibres. Retinoic acid also suppressed the effect of dexamethasone (-21 ± 7%). Spectrin is a key protein in the actin network remodelling. Its abundance was decreased by retinoic acid and increased by dexamethasone (-21 ± 11% and 52 ± 10%). However, retinoic acid did not modify the effect of dexamethasone (48 ± 7%). Calpain activity on spectrin was increased by retinoic acid and decreased by dexamethasone (26 ± 14% and -57 ± 5%); retinoic acid mildly but significantly modified the effect of dexamethasone (-44 ± 7%). The calpain inhibitor calpeptin suppressed the effects of retinoic acid and dexamethasone on cell shape and actin stress fibres remodelling but did not modify the effects on cortical actin. Retinoic acid and dexamethasone have a dramatic but mainly opposite effect on actin cytoskeleton remodelling. These effects originate, at least partly, from calpain activity.
Collapse
Affiliation(s)
- Hélène Roumes
- INRA, Nutrition et neurobiologie intégrée, UMR 1286, 33076 Bordeaux, France; University Bordeaux, Nutrition et neurobiologie intégrée, UMR 1286, 33076 Bordeaux, France
| | - Julie Brossaud
- University Bordeaux, Nutrition et neurobiologie intégrée, UMR 1286, 33076 Bordeaux, France; Laboratoire d'Hormonologie, Service de médecine nucléaire, CHU Bordeaux, 33604 Pessac, France
| | - Aloïs Lemelletier
- INRA, Nutrition et neurobiologie intégrée, UMR 1286, 33076 Bordeaux, France; University Bordeaux, Nutrition et neurobiologie intégrée, UMR 1286, 33076 Bordeaux, France
| | - Marie-Pierre Moisan
- INRA, Nutrition et neurobiologie intégrée, UMR 1286, 33076 Bordeaux, France; University Bordeaux, Nutrition et neurobiologie intégrée, UMR 1286, 33076 Bordeaux, France
| | - Véronique Pallet
- University Bordeaux, Nutrition et neurobiologie intégrée, UMR 1286, 33076 Bordeaux, France; IPB, Nutrition et neurobiologie intégrée, UMR 1286, 33076 Bordeaux, France
| | - Anabelle Redonnet
- INRA, Nutrition et neurobiologie intégrée, UMR 1286, 33076 Bordeaux, France; University Bordeaux, Nutrition et neurobiologie intégrée, UMR 1286, 33076 Bordeaux, France
| | - Jean-Benoît Corcuff
- University Bordeaux, Nutrition et neurobiologie intégrée, UMR 1286, 33076 Bordeaux, France; Laboratoire d'Hormonologie, Service de médecine nucléaire, CHU Bordeaux, 33604 Pessac, France.
| |
Collapse
|
155
|
Hu S, Cao Q, Xu P, Ji W, Wang G, Zhang Y. Rolipram stimulates angiogenesis and attenuates neuronal apoptosis through the cAMP/cAMP-responsive element binding protein pathway following ischemic stroke in rats. Exp Ther Med 2015; 11:1005-1010. [PMID: 26998028 DOI: 10.3892/etm.2015.2958] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 06/13/2014] [Indexed: 12/25/2022] Open
Abstract
Rolipram, a phosphodiesterase-4 inhibitor, can activate the cyclic adenosine monophosphate (cAMP)/cAMP-responsive element binding protein (CREB) pathway to facilitate functional recovery following ischemic stroke. However, to date, the effects of rolipram on angiogenesis and cerebral ischemia-induced neuronal apoptosis are yet to be fully elucidated. In this study, the aim was to reveal the effect of rolipram on the angiogenesis and neuronal apoptosis following brain cerebral ischemia. Rat models of ischemic stroke were established following transient middle cerebral artery occlusion and rolipram was administered for three, seven and 14 days. The results were examined using behavioral tests, triphenyl tetrazolium chloride staining, immunostaining and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) to evaluate the effects of rolipram therapy on functional outcome, angiogenesis and apoptosis. Western blot analysis was used to show the phosphorylated- (p-)CREB protein level in the ischemic hemisphere. The rolipram treatment group exhibited a marked reduction in infarct size and modified neurological severity score compared with the vehicle group, and rolipram treatment significantly promoted the microvessel density in the ischemic boundary region and increased p-CREB protein levels in the ischemic hemisphere. Furthermore, a significant reduction in the number of TUNEL-positive cells was observed in the rolipram group compared with the vehicle group. These findings suggest that rolipram has the ability to attenuate cerebral ischemic injury, stimulate angiogenesis and reduce neuronal apoptosis though the cAMP/CREB pathway.
Collapse
Affiliation(s)
- Shouye Hu
- Department of Osteonecrosis and Joint Reconstruction, Xi'an Honghui Hospital, Xi'an, Shaanxi 710068, P.R. China; Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710068, P.R. China
| | - Qingwen Cao
- Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710068, P.R. China
| | - Peng Xu
- Department of Osteonecrosis and Joint Reconstruction, Xi'an Honghui Hospital, Xi'an, Shaanxi 710068, P.R. China; Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710068, P.R. China
| | - Wenchen Ji
- Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710068, P.R. China
| | - Gang Wang
- Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710068, P.R. China; Department of Neurosurgery, Shaanxi Province People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Yuelin Zhang
- Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710068, P.R. China; Department of Neurosurgery, Shaanxi Province People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| |
Collapse
|
156
|
Waller JA, Chen F, Sánchez C. Vortioxetine promotes maturation of dendritic spines in vitro: A comparative study in hippocampal cultures. Neuropharmacology 2015; 103:143-54. [PMID: 26702943 DOI: 10.1016/j.neuropharm.2015.12.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 11/03/2015] [Accepted: 12/13/2015] [Indexed: 10/22/2022]
Abstract
Cognitive dysfunction is prevalent in patients with major depressive disorder (MDD), and cognitive impairments can persist after relief of depressive symptoms. The multimodal-acting antidepressant vortioxetine is an antagonist at 5-HT3, 5-HT7, and 5-HT1D receptors, a partial agonist at 5-HT1B receptors, an agonist at 5-HT1A receptors, and an inhibitor of the serotonin (5-HT) transporter (SERT) and has pro-cognitive properties. In preclinical studies, vortioxetine enhances long-term potentiation (LTP), a cellular correlate of neuroplasticity, and enhances memory in various cognitive tasks. However, the molecular mechanisms by which vortioxetine augments LTP and memory remain unknown. Dendritic spines are specialized, actin-rich microdomains on dendritic shafts and are major sites of most excitatory synapses. Since dendritic spine remodeling is implicated in synaptic plasticity and spine size dictates the strength of synaptic transmission, we assessed if vortioxetine, relative to other antidepressants including ketamine, duloxetine, and fluoxetine, plays a role in the maintenance of dendritic spine architecture in vitro. We show that vortioxetine, ketamine, and duloxetine induce spine enlargement. However, only vortioxetine treatment increased the number of spines in contact with presynaptic terminals. In contrast, fluoxetine had no effect on spine remodeling. These findings imply that the various 5-HT receptor mechanisms of vortioxetine may play a role in its effect on spine dynamics and in increasing the proportion of potentially functional synaptic contacts.
Collapse
Affiliation(s)
- Jessica A Waller
- External Sourcing and Scientific Excellence, Lundbeck Research USA, Paramus, NJ 07652, USA
| | - Fenghua Chen
- Stereology and Electron Microscopy Laboratory, Centre for Stochastic Geometry and Advanced Bioimaging, Aarhus University Hospital, DK-8000 Aarhus C, Denmark; Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, DK-8240 Risskov, Denmark
| | - Connie Sánchez
- External Sourcing and Scientific Excellence, Lundbeck Research USA, Paramus, NJ 07652, USA.
| |
Collapse
|
157
|
Penazzi L, Bakota L, Brandt R. Microtubule Dynamics in Neuronal Development, Plasticity, and Neurodegeneration. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2015; 321:89-169. [PMID: 26811287 DOI: 10.1016/bs.ircmb.2015.09.004] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Neurons are the basic information-processing units of the nervous system. In fulfilling their task, they establish a structural polarity with an axon that can be over a meter long and dendrites with a complex arbor, which can harbor ten-thousands of spines. Microtubules and their associated proteins play important roles during the development of neuronal morphology, the plasticity of neurons, and neurodegenerative processes. They are dynamic structures, which can quickly adapt to changes in the environment and establish a structural scaffold with high local variations in composition and stability. This review presents a comprehensive overview about the role of microtubules and their dynamic behavior during the formation and maturation of processes and spines in the healthy brain, during aging and under neurodegenerative conditions. The review ends with a discussion of microtubule-targeted therapies as a perspective for the supportive treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
- Lorène Penazzi
- Department of Neurobiology, University of Osnabrück, Osnabrück, Germany
| | - Lidia Bakota
- Department of Neurobiology, University of Osnabrück, Osnabrück, Germany
| | - Roland Brandt
- Department of Neurobiology, University of Osnabrück, Osnabrück, Germany
| |
Collapse
|
158
|
Poddar R, Rajagopal S, Shuttleworth CW, Paul S. Zn2+-dependent Activation of the Trk Signaling Pathway Induces Phosphorylation of the Brain-enriched Tyrosine Phosphatase STEP: MOLECULAR BASIS FOR ZN2+-INDUCED ERK MAPK ACTIVATION. J Biol Chem 2015; 291:813-25. [PMID: 26574547 DOI: 10.1074/jbc.m115.663468] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Indexed: 11/06/2022] Open
Abstract
Excessive release of Zn(2+) in the brain is implicated in the progression of acute brain injuries. Although several signaling cascades have been reported to be involved in Zn(2+)-induced neurotoxicity, a potential contribution of tyrosine phosphatases in this process has not been well explored. Here we show that exposure to high concentrations of Zn(2+) led to a progressive increase in phosphorylation of the striatal-enriched phosphatase (STEP), a component of the excitotoxic-signaling pathway that plays a role in neuroprotection. Zn(2+)-mediated phosphorylation of STEP61 at multiple sites (hyperphosphorylation) was induced by the up-regulation of brain-derived neurotropic factor (BDNF), tropomyosin receptor kinase (Trk) signaling, and activation of cAMP-dependent PKA (protein kinase A). Mutational studies further show that differential phosphorylation of STEP61 at the PKA sites, Ser-160 and Ser-221 regulates the affinity of STEP61 toward its substrates. Consistent with these findings we also show that BDNF/Trk/PKA mediated signaling is required for Zn(2+)-induced phosphorylation of extracellular regulated kinase 2 (ERK2), a substrate of STEP that is involved in Zn(2+)-dependent neurotoxicity. The strong correlation between the temporal profile of STEP61 hyperphosphorylation and ERK2 phosphorylation indicates that loss of function of STEP61 through phosphorylation is necessary for maintaining sustained ERK2 phosphorylation. This interpretation is further supported by the findings that deletion of the STEP gene led to a rapid and sustained increase in ERK2 phosphorylation within minutes of exposure to Zn(2+). The study provides further insight into the mechanisms of regulation of STEP61 and also offers a molecular basis for the Zn(2+)-induced sustained activation of ERK2.
Collapse
Affiliation(s)
| | | | - C William Shuttleworth
- Neurosciences University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87131
| | - Surojit Paul
- From the Departments of Neurology and Neurosciences University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87131
| |
Collapse
|
159
|
Nakajima S, Numakawa T, Adachi N, Ooshima Y, Odaka H, Yoshimura A, Kunugi H. Self-amplified BDNF transcription is a regulatory system for synaptic maturation in cultured cortical neurons. Neurochem Int 2015; 91:55-61. [PMID: 26596846 DOI: 10.1016/j.neuint.2015.10.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 10/08/2015] [Accepted: 10/20/2015] [Indexed: 12/01/2022]
Abstract
Neuronal cell survival and synaptic plasticity are controlled through expression of various neurotrophic factors including brain-derived neurotrophic factor (BDNF). In the present study, we examined the mechanism behind BDNF-induced Bdnf mRNA production and the physiological role of its amplification system using cortical neurons. Exogenous BDNF was applied to the cultured cortical neurons at days in vitro (DIV) 3 and DIV 7 with or without inhibitors for intracellular signaling. Expression levels of total Bdnf and Bdnf variants (exon I, exon IV, and exon VI) were biphasically increased after the BDNF application in different developing stage of neurons. Inhibitor for extracellular signal-regulated kinase, calmodulin dependent protein kinase II, or protein kinase A repressed the BDNF-induced Bdnf mRNA expression. Furthermore, we found that application of TrkB-Fc, which scavenges produced endogenous BDNF, resulted in weakened BDNF/TrkB signaling and decreased expression of postsynaptic proteins, suggesting that newly synthesized BDNF induced by the self-amplification system contributes to the synaptic maturation and function.
Collapse
Affiliation(s)
- Shingo Nakajima
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan; Faculty of Health Science, Hokkaido University, Sapporo, Japan
| | - Tadahiro Numakawa
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan; Department of Cell Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan.
| | - Naoki Adachi
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan; Department of Biomedical Chemistry, Kwansei Gakuin University, Sanda, Japan
| | - Yoshiko Ooshima
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan; Department of Molecular Pharmacology, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| | - Haruki Odaka
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan; Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Aya Yoshimura
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| | - Hiroshi Kunugi
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| |
Collapse
|
160
|
PLD1 participates in BDNF-induced signalling in cortical neurons. Sci Rep 2015; 5:14778. [PMID: 26437780 PMCID: PMC4594037 DOI: 10.1038/srep14778] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 09/09/2015] [Indexed: 01/07/2023] Open
Abstract
The brain-derived neurotrophic factor BDNF plays a critical role in neuronal development and the induction of L-LTP at glutamatergic synapses in several brain regions. However, the cellular and molecular mechanisms underlying these BDNF effects have not been firmly established. Using in vitro cultures of cortical neurons from knockout mice for Pld1 and Rsk2, BDNF was observed to induce a rapid RSK2-dependent activation of PLD and to stimulate BDNF ERK1/2-CREB and mTor-S6K signalling pathways, but these effects were greatly reduced in Pld1(-/-) neurons. Furthermore, phospho-CREB did not accumulate in the nucleus, whereas overexpression of PLD1 amplified the BDNF-dependent nuclear recruitment of phospho-ERK1/2 and phospho-CREB. This BDNF retrograde signalling was prevented in cells silenced for the scaffolding protein PEA15, a protein which complexes with PLD1, ERK1/2, and RSK2 after BDNF treatment. Finally PLD1, ERK1/2, and RSK2 partially colocalized on endosomal structures, suggesting that these proteins are part of the molecular module responsible for BDNF signalling in cortical neurons.
Collapse
|
161
|
Long ZM, Zhao L, Jiang R, Wang KJ, Luo SF, Zheng M, Li XF, He GQ. Valproic Acid Modifies Synaptic Structure and Accelerates Neurite Outgrowth Via the Glycogen Synthase Kinase-3β Signaling Pathway in an Alzheimer's Disease Model. CNS Neurosci Ther 2015; 21:887-97. [PMID: 26385876 DOI: 10.1111/cns.12445] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 07/28/2015] [Accepted: 07/29/2015] [Indexed: 12/20/2022] Open
Abstract
AIM Tau hyperphosphorylation and amyloid β-peptide overproduction, caused by altered localization or abnormal activation of glycogen synthase kinase-3β (GSK-3β), is a pathogenic mechanism in Alzheimer's disease (AD). Valproic acid (VPA) attenuates senile plaques and neuronal loss. Here, we confirmed that VPA treatment improved spatial memory in amyloid precursor protein (APP)/presenilin 1 (PS 1) double-transgenic mice and investigated the effect of VPA on synaptic structure and neurite outgrowth. METHODS We used ultrastructural analysis, immunocytochemistry, immunofluorescence staining, and Western blot analysis to assess the effect of VPA treatment in mice. RESULTS VPA treatment thickened the postsynaptic density, increased the number of presynaptic vesicles, and upregulated the expression of synaptic markers PSD-95 and GAP43. VPA increased neurite length of hippocampal neurons in vivo and in vitro. In VPA-treated AD mouse brain, inactivated GSK-3β (pSer9-GSK-3β) was markedly increased, while hyperphosphorylation of tau at Ser396 and Ser262 was decreased; total tau levels remained similar. VPA treatment notably improved pSer133-cAMP response element-binding protein (CREB) and brain-derived neurotrophic factor (BDNF) levels, which are associated with synaptic function and neurite outgrowth. CONCLUSION VPA improves behavioral deficits in AD, modifies synaptic structure, and accelerates neurite outgrowth, by inhibiting the activity of GSK-3β, decreasing hyperphosphorylated tau, enhancing CREB and BDNF expression.
Collapse
Affiliation(s)
- Zhi-Min Long
- Chongqing Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, China.,Department of Anatomy, Chongqing Medical University, Chongqing, China
| | - Lei Zhao
- Chongqing Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, China
| | - Rong Jiang
- Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, China
| | - Ke-Jian Wang
- Chongqing Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, China.,Department of Anatomy, Chongqing Medical University, Chongqing, China
| | - Shi-Fang Luo
- Chongqing Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, China.,Department of Anatomy, Chongqing Medical University, Chongqing, China
| | - Min Zheng
- Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, China
| | - Xiao-Feng Li
- Department of Neurology, The 2nd Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Gui-Qiong He
- Chongqing Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, China.,Department of Anatomy, Chongqing Medical University, Chongqing, China
| |
Collapse
|
162
|
In Vivo Two-Photon Imaging of Dendritic Spines in Marmoset Neocortex. eNeuro 2015; 2:eN-MNT-0019-15. [PMID: 26465000 PMCID: PMC4596018 DOI: 10.1523/eneuro.0019-15.2015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 07/03/2015] [Accepted: 07/27/2015] [Indexed: 11/21/2022] Open
Abstract
Two-photon microscopy in combination with a technique involving the artificial expression of fluorescent protein has enabled the direct observation of dendritic spines in living brains. However, the application of this method to primate brains has been hindered by the lack of appropriate labeling techniques for visualizing dendritic spines. Here, we developed an adeno-associated virus vector-based fluorescent protein expression system for visualizing dendritic spines in vivo in the marmoset neocortex. For the clear visualization of each spine, the expression of reporter fluorescent protein should be both sparse and strong. To fulfill these requirements, we amplified fluorescent signals using the tetracycline transactivator (tTA)–tetracycline-responsive element system and by titrating down the amount of Thy1S promoter-driven tTA for sparse expression. By this method, we were able to visualize dendritic spines in the marmoset cortex by two-photon microscopy in vivo and analyze the turnover of spines in the prefrontal cortex. Our results demonstrated that short spines in the marmoset cortex tend to change more frequently than long spines. The comparison of in vivo samples with fixed samples showed that we did not detect all existing spines by our method. Although we found glial cell proliferation, the damage of tissues caused by window construction was relatively small, judging from the comparison of spine length between samples with or without window construction. Our new labeling technique for two-photon imaging to visualize in vivo dendritic spines of the marmoset neocortex can be applicable to examining circuit reorganization and synaptic plasticity in primates.
Collapse
|
163
|
BONLAC: A combinatorial proteomic technique to measure stimulus-induced translational profiles in brain slices. Neuropharmacology 2015. [PMID: 26205778 DOI: 10.1016/j.neuropharm.2015.07.017] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Stimulus-triggered protein synthesis is critical for brain health and function. However, due to technical hurdles, de novo neuronal translation is predominantly studied in cultured cells, whereas electrophysiological and circuit analyses often are performed in brain slices. The different properties of these two experimental systems create an information gap about stimulus-induced alterations in the expression of new proteins in mature circuits. To address this, we adapted two existing techniques, BONCAT and SILAC, to a combined proteomic technique, BONLAC, for use in acute adult hippocampal slices. Using BDNF-induced protein synthesis as a proof of concept, we found alterations in expression of proteins involved in neurotransmission, trafficking, and cation binding that differed from those found in a similar screen in cultured neurons. Our results indicate important differences between cultured neurons and slices, and suggest that BONLAC could be used to dissect proteomic changes underlying synaptic events in adult circuits. This article is part of the Special Issue entitled 'Synaptopathy--from Biology to Therapy'.
Collapse
|
164
|
Marshall J, Wong KY, Rupasinghe CN, Tiwari R, Zhao X, Berberoglu ED, Sinkler C, Liu J, Lee I, Parang K, Spaller MR, Hüttemann M, Goebel DJ. Inhibition of N-Methyl-D-aspartate-induced Retinal Neuronal Death by Polyarginine Peptides Is Linked to the Attenuation of Stress-induced Hyperpolarization of the Inner Mitochondrial Membrane Potential. J Biol Chem 2015; 290:22030-48. [PMID: 26100636 DOI: 10.1074/jbc.m115.662791] [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: 05/11/2015] [Indexed: 02/02/2023] Open
Abstract
It is widely accepted that overactivation of NMDA receptors, resulting in calcium overload and consequent mitochondrial dysfunction in retinal ganglion neurons, plays a significant role in promoting neurodegenerative disorders such as glaucoma. Calcium has been shown to initiate a transient hyperpolarization of the mitochondrial membrane potential triggering a burst of reactive oxygen species leading to apoptosis. Strategies that enhance cell survival signaling pathways aimed at preventing this adverse hyperpolarization of the mitochondrial membrane potential may provide a novel therapeutic intervention in retinal disease. In the retina, brain-derived neurotrophic factor has been shown to be neuroprotective, and our group previously reported a PSD-95/PDZ-binding cyclic peptide (CN2097) that augments brain-derived neurotrophic factor-induced pro-survival signaling. Here, we examined the neuroprotective properties of CN2097 using an established retinal in vivo NMDA toxicity model. CN2097 completely attenuated NMDA-induced caspase 3-dependent and -independent cell death and PARP-1 activation pathways, blocked necrosis, and fully prevented the loss of long term ganglion cell viability. Although neuroprotection was partially dependent upon CN2097 binding to the PDZ domain of PSD-95, our results show that the polyarginine-rich transport moiety C-R(7), linked to the PDZ-PSD-95-binding cyclic peptide, was sufficient to mediate short and long term protection via a mitochondrial targeting mechanism. C-R(7) localized to mitochondria and was found to reduce mitochondrial respiration, mitochondrial membrane hyperpolarization, and the generation of reactive oxygen species, promoting survival of retinal neurons.
Collapse
Affiliation(s)
- John Marshall
- From the Department of Molecular Pharmacology, Physiology and Biotechnology, Brown University, Providence, Rhode Island, 02912
| | - Kwoon Y Wong
- the Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, Michigan 48105
| | - Chamila N Rupasinghe
- the Geisel School of Medicine at Dartmouth, Department of Pharmacology and Toxicology, and Norris Cotton Cancer Center, Lebanon, New Hampshire 03756
| | - Rakesh Tiwari
- the Chapman University School of Pharmacy, Irvine, California 92618
| | - Xiwu Zhao
- the Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, Michigan 48105
| | | | - Christopher Sinkler
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan 48201, and
| | - Jenney Liu
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan 48201, and
| | - Icksoo Lee
- the College of Medicine, Dankook University, Cheonan-si, Chungcheongnam-do, 330-714, Republic of Korea
| | - Keykavous Parang
- the Chapman University School of Pharmacy, Irvine, California 92618
| | - Mark R Spaller
- the Geisel School of Medicine at Dartmouth, Department of Pharmacology and Toxicology, and Norris Cotton Cancer Center, Lebanon, New Hampshire 03756
| | - Maik Hüttemann
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan 48201, and
| | | |
Collapse
|
165
|
Sun MK, Nelson TJ, Alkon DL. Towards universal therapeutics for memory disorders. Trends Pharmacol Sci 2015; 36:384-94. [DOI: 10.1016/j.tips.2015.04.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 04/07/2015] [Accepted: 04/08/2015] [Indexed: 12/22/2022]
|
166
|
Khan MZ, He L. The role of polyunsaturated fatty acids and GPR40 receptor in brain. Neuropharmacology 2015; 113:639-651. [PMID: 26005184 DOI: 10.1016/j.neuropharm.2015.05.013] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 04/02/2015] [Accepted: 05/08/2015] [Indexed: 01/15/2023]
Abstract
Polyunsaturated fatty acids (PUFAs) are found in abundance in the nervous system. They perform significant functions for example boosting synaptogenesis, neurogenesis, inducing antinociception, stimulating gene expression and neuronal activity, preventing apoptosis and neuroinflammation. G-protein-coupled receptor 40 (GPR40), also called free fatty acid receptor 1 (FFA1), is ubiquitously expressed in various regions of the human brain including the olfactory bulb, midbrain, medulla oblongata, hippocampus, hypothalamus, cerebral cortex, cerebellum and in the spinal cord. GPR40, when binding with polyunsaturated fatty acids (PUFAs) has shown promising therapeutic potential. This review presents current knowledge regarding the pharmacological properties of GPR40 and addresses its functions in brain, with a focus on neurodevelopment & neurogenesis. Furthermore, the demonstration of GPR40 involvement in several neuropathological conditions such as apoptosis, inflammatory pain, Alzheimer's disease and Parkinson's disease. Although the results are encouraging, further research is needed to clarify their role in the treatment of inflammatory pain, Alzheimer's disease and Parkinson's disease. This article is part of the Special Issue entitled 'Lipid Sensing G Protein-Coupled Receptors in the CNS'.
Collapse
Affiliation(s)
- Muhammad Zahid Khan
- Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China
| | - Ling He
- Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
167
|
Pickering C, Alsiö J, Morud J, Ericson M, Robbins TW, Söderpalm B. Ethanol impairment of spontaneous alternation behaviour and associated changes in medial prefrontal glutamatergic gene expression precede putative markers of dependence. Pharmacol Biochem Behav 2015; 132:63-70. [DOI: 10.1016/j.pbb.2015.02.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 02/17/2015] [Accepted: 02/21/2015] [Indexed: 12/21/2022]
|
168
|
Silva A, Naia L, Dominguez A, Ribeiro M, Rodrigues J, Vieira OV, Lessmann V, Rego AC. Overexpression of BDNF and Full-Length TrkB Receptor Ameliorate Striatal Neural Survival in Huntington's Disease. NEURODEGENER DIS 2015; 15:207-18. [PMID: 25896770 DOI: 10.1159/000375447] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 01/21/2015] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Several cellular mechanisms have been proposed to explain the pathogenesis of Huntington's disease (HD), including the lack of striatal brain-derived neurotrophic factor (BDNF). Thus, by preferentially binding to tropomyosin receptor kinase B (TrkB) receptor, BDNF is an important neurotrophin implicated in striatal neuronal survival. OBJECTIVE To study the influence of BDNF and TrkB receptors in intracellular signaling pathways and caspase-3 activation in HD striatal cells. METHODS HD mutant knockin and wild-type striatal cells were transduced with preproBDNF or full-length TrkB receptors to analyze BDNF processing, AKT and extracellular signal-regulated kinase (ERK) activation and the activity of caspase-3 in the absence or presence of staurosporine (STS). RESULTS HD mutant cells transduced with preproBDNF-mCherry (mCh) expressed similar levels of pro- and mature BDNF compared to WT cells, but HD cells released lower levels of pro- and mature BDNF. Despite this, BDNF-mCh overexpression rescued decreased AKT phosphorylation and reduced the caspase-3 activation observed in HD cells. Activated ERK was also enhanced in HD BDNF-mCh/TrkB-eGFP receptor co-cultures. Of relevance, overexpression of TrkB-eGFP in HD cells decreased caspase-3 activation, and stimulation of TrkB-eGFP-transduced mutant cells with recombinant human BDNF reduced both basal and STS-induced caspase-3 activation. CONCLUSION The results highlight the importance of BDNF-induced TrkB receptor signaling in rescuing HD-mediated apoptotic features in striatal cells.
Collapse
Affiliation(s)
- Ana Silva
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | | | | | | | | | | | | | | |
Collapse
|
169
|
Shp2 in forebrain neurons regulates synaptic plasticity, locomotion, and memory formation in mice. Mol Cell Biol 2015; 35:1557-72. [PMID: 25713104 DOI: 10.1128/mcb.01339-14] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 02/17/2015] [Indexed: 11/20/2022] Open
Abstract
Shp2 (Src homology 2 domain-containing protein tyrosine phosphatase 2) regulates neural cell differentiation. It is also expressed in postmitotic neurons, however, and mutations of Shp2 are associated with clinical syndromes characterized by mental retardation. Here we show that conditional-knockout (cKO) mice lacking Shp2 specifically in postmitotic forebrain neurons manifest abnormal behavior, including hyperactivity. Novelty-induced expression of immediate-early genes and activation of extracellular-signal-regulated kinase (Erk) were attenuated in the cerebral cortex and hippocampus of Shp2 cKO mice, suggestive of reduced neuronal activity. In contrast, ablation of Shp2 enhanced high-K(+)-induced Erk activation in both cultured cortical neurons and synaptosomes, whereas it inhibited that induced by brain-derived growth factor in cultured neurons. Posttetanic potentiation and paired-pulse facilitation were attenuated and enhanced, respectively, in hippocampal slices from Shp2 cKO mice. The mutant mice also manifested transient impairment of memory formation in the Morris water maze. Our data suggest that Shp2 contributes to regulation of Erk activation and synaptic plasticity in postmitotic forebrain neurons and thereby controls locomotor activity and memory formation.
Collapse
|
170
|
Girdin phosphorylation is crucial for synaptic plasticity and memory: a potential role in the interaction of BDNF/TrkB/Akt signaling with NMDA receptor. J Neurosci 2015; 34:14995-5008. [PMID: 25378165 DOI: 10.1523/jneurosci.2228-14.2014] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Synaptic plasticity in hippocampal neurons has been thought to represent a variety of memories. Although accumulating evidence indicates a crucial role of BDNF/TrkB/Akt signaling in the synaptic plasticity of the hippocampus, the mechanism by which Akt, a serine/threonine kinase, controls activity-dependent neuronal plasticity remains unclear. Girdin (also known as APE, GIV, and HkRP1), an actin-binding protein involved both in the remodeling of the actin cytoskeleton and in cell migration, has been identified as a substrate of Akt. Previous studies have demonstrated that deficit of neuronal migration in the hippocampus of Girdin-deficient (Girdin(-/-)) mice is independent on serine phosphorylation of Girdin at S1416 (Girdin S1416) by Akt. In the present study, we focused on the role of Girdin S1416 phosphorylation in BDNF/TrkB/Akt signaling associated with synaptic plasticity. We found that Girdin in the hippocampus was phosphorylated at S1416 in an activity-dependent manner. Phosphorylation-deficient knock-in mice (Girdin(SA/SA) mice), in which S1416 is replaced with alanine, exhibited shrinkage of spines, deficit of hippocampal long-term potentiation, and memory impairment. These phenotypes of Girdin(SA/SA) mice resembled those of Girdin(+/-) mice, which have 50% loss of Girdin expression. Furthermore, Girdin interacted with Src kinase and NR2B subunit of NMDA receptor, leading to phosphorylation of the NR2B subunit and NMDA receptor activation. Our findings suggest that Girdin has two different functions in the hippocampus: Akt-independent neuronal migration and Akt-dependent NR2B phosphorylation through the interaction with Src, which is associated with synaptic plasticity in the hippocampus underlying memory formation.
Collapse
|
171
|
Liu H, Xue X, Shi H, Qi L, Gong D. Osthole Upregulates BDNF to Enhance Adult Hippocampal Neurogenesis in APP/PS1 Transgenic Mice. Biol Pharm Bull 2015; 38:1439-49. [DOI: 10.1248/bpb.b15-00013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Hong Liu
- Department of Neurology, Liaocheng Hospital
| | | | - Huijian Shi
- Department of Anesthesiology, Affilliated Hospital of TaiShan Medical College
| | - Lifeng Qi
- Department of Neurology, Liaocheng Hospital
| | | |
Collapse
|
172
|
Reimers JM, Loweth JA, Wolf ME. BDNF contributes to both rapid and homeostatic alterations in AMPA receptor surface expression in nucleus accumbens medium spiny neurons. Eur J Neurosci 2014; 39:1159-69. [PMID: 24712995 DOI: 10.1111/ejn.12422] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Revised: 09/30/2013] [Accepted: 10/12/2013] [Indexed: 12/15/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) plays a critical role in plasticity at glutamate synapses and in the effects of repeated cocaine exposure. We recently showed that intracranial injection of BDNF into the rat nucleus accumbens (NAc), a key region for cocaine addiction, rapidly increases α-amino-3-hyroxy-5-methyl-4-isoxazole-propionic acid receptor (AMPAR) surface expression. To further characterize BDNF's role in both rapid AMPAR trafficking and slower, homeostatic changes in AMPAR surface expression, we investigated the effects of acute (30 min) and long-term (24 h) treatment with BDNF on AMPAR distribution in NAc medium spiny neurons from postnatal rats co-cultured with mouse prefrontal cortex neurons to restore excitatory inputs. Immunocytochemical studies showed that acute BDNF treatment increased cell surface GluA1 and GluA2 levels, as well as their co-localization, on NAc neurons. This effect of BDNF, confirmed using a protein crosslinking assay, was dependent on ERK but not AKT signaling. In contrast, long-term BDNF treatment decreased AMPAR surface expression on NAc neurons. Based on this latter result, we tested the hypothesis that BDNF plays a role in AMPAR 'scaling down' in response to a prolonged increase in neuronal activity produced by bicuculline (24 h). Supporting this hypothesis, decreasing BDNF signaling with the extracellular BDNF scavenger TrkB-Fc prevented the scaling down of GluA1 and GluA2 surface levels in NAc neurons normally produced by bicuculline. In conclusion, BDNF exerts bidirectional effects on NAc AMPAR surface expression, depending on duration of exposure. Furthermore, BDNF's involvement in synaptic scaling in the NAc differs from its previously described role in the visual cortex.
Collapse
Affiliation(s)
- Jeremy M Reimers
- Department of Neuroscience, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064-3095, USA
| | | | | |
Collapse
|
173
|
Molecular regulation of synaptogenesis during associative learning and memory. Brain Res 2014; 1621:239-51. [PMID: 25485772 DOI: 10.1016/j.brainres.2014.11.054] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 11/24/2014] [Accepted: 11/25/2014] [Indexed: 01/06/2023]
Abstract
Synaptogenesis plays a central role in associative learning and memory. The biochemical pathways that underlie synaptogenesis are complex and incompletely understood. Nevertheless, research has so far identified three conceptually distinct routes to synaptogenesis: cell-cell contact mediated by adhesion proteins, cell-cell biochemical signaling from astrocytes and other cells, and neuronal signaling through classical ion channels and cell surface receptors. The cell adhesion pathways provide the physical substrate to the new synaptic connection, while cell-cell signaling may provide a global or regional signal, and the activity-dependent pathways provide the neuronal specificity that is required for the new synapses to produce functional neuronal networks capable of storing associative memories. These three aspects of synaptogenesis require activation of a variety of interacting biochemical pathways that converge on the actin cytoskeleton and strengthen the synapse in an information-dependent manner. This article is part of a Special Issue titled SI: Brain and Memory.
Collapse
|
174
|
Cholinergic differentiation of neural stem cells generated from cell aggregates-derived from Human Bone marrow stromal cells. Tissue Eng Regen Med 2014. [DOI: 10.1007/s13770-014-0019-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
|
175
|
Smith PA. BDNF: No gain without pain? Neuroscience 2014; 283:107-23. [DOI: 10.1016/j.neuroscience.2014.05.044] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 05/16/2014] [Accepted: 05/21/2014] [Indexed: 12/22/2022]
|
176
|
Adachi N, Numakawa T, Richards M, Nakajima S, Kunugi H. New insight in expression, transport, and secretion of brain-derived neurotrophic factor: Implications in brain-related diseases. World J Biol Chem 2014; 5:409-428. [PMID: 25426265 PMCID: PMC4243146 DOI: 10.4331/wjbc.v5.i4.409] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2014] [Revised: 06/10/2014] [Accepted: 08/31/2014] [Indexed: 02/05/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) attracts increasing attention from both research and clinical fields because of its important functions in the central nervous system. An adequate amount of BDNF is critical to develop and maintain normal neuronal circuits in the brain. Given that loss of BDNF function has been reported in the brains of patients with neurodegenerative or psychiatric diseases, understanding basic properties of BDNF and associated intracellular processes is imperative. In this review, we revisit the gene structure, transcription, translation, transport and secretion mechanisms of BDNF. We also introduce implications of BDNF in several brain-related diseases including Alzheimer’s disease, Huntington’s disease, depression and schizophrenia.
Collapse
|
177
|
Ding X, Cai J, Li S, Liu XD, Wan Y, Xing GG. BDNF contributes to the development of neuropathic pain by induction of spinal long-term potentiation via SHP2 associated GluN2B-containing NMDA receptors activation in rats with spinal nerve ligation. Neurobiol Dis 2014; 73:428-51. [PMID: 25447233 DOI: 10.1016/j.nbd.2014.10.025] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 10/16/2014] [Accepted: 10/31/2014] [Indexed: 12/30/2022] Open
Abstract
The pathogenic mechanisms underlying neuropathic pain still remain largely unknown. In this study, we investigated whether spinal BDNF contributes to dorsal horn LTP induction and neuropathic pain development by activation of GluN2B-NMDA receptors via Src homology-2 domain-containing protein tyrosine phosphatase-2 (SHP2) phosphorylation in rats following spinal nerve ligation (SNL). We first demonstrated that spinal BDNF participates in the development of long-lasting hyperexcitability of dorsal horn WDR neurons (i.e. central sensitization) as well as pain allodynia in both intact and SNL rats. Second, we revealed that BDNF induces spinal LTP at C-fiber synapses via functional up-regulation of GluN2B-NMDA receptors in the spinal dorsal horn, and this BDNF-mediated LTP-like state is responsible for the occlusion of spinal LTP elicited by subsequent high-frequency electrical stimulation (HFS) of the sciatic nerve in SNL rats. Finally, we validated that BDNF-evoked SHP2 phosphorylation is required for subsequent GluN2B-NMDA receptors up-regulation and spinal LTP induction, and also for pain allodynia development. Blockade of SHP2 phosphorylation in the spinal dorsal horn using a potent SHP2 protein tyrosine phosphatase inhibitor NSC-87877, or knockdown of spinal SHP2 by intrathecal delivery of SHP2 siRNA, not only prevents BDNF-mediated GluN2B-NMDA receptors activation as well as spinal LTP induction and pain allodynia elicitation in intact rats, but also reduces the SNL-evoked GluN2B-NMDA receptors up-regulation and spinal LTP occlusion, and ultimately alleviates pain allodynia in neuropathic rats. Taken together, these results suggest that the BDNF/SHP2/GluN2B-NMDA signaling cascade plays a vital role in the development of central sensitization and neuropathic pain after peripheral nerve injury.
Collapse
Affiliation(s)
- Xu Ding
- Neuroscience Research Institute, Peking University, Beijing 100191, P.R. China; Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, P.R. China.
| | - Jie Cai
- Neuroscience Research Institute, Peking University, Beijing 100191, P.R. China; Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, P.R. China.
| | - Song Li
- Neuroscience Research Institute, Peking University, Beijing 100191, P.R. China; Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, P.R. China.
| | - Xiao-Dan Liu
- Neuroscience Research Institute, Peking University, Beijing 100191, P.R. China; Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, P.R. China.
| | - You Wan
- Neuroscience Research Institute, Peking University, Beijing 100191, P.R. China; Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, P.R. China.
| | - Guo-Gang Xing
- Neuroscience Research Institute, Peking University, Beijing 100191, P.R. China; Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, P.R. China; Key Laboratory for Neuroscience, Ministry of Education and Ministry of Health, Beijing 100191, P.R. China.
| |
Collapse
|
178
|
Brain-Derived Neurotrophic Factor in Alzheimer's Disease: Risk, Mechanisms, and Therapy. Mol Neurobiol 2014; 52:1477-1493. [PMID: 25354497 DOI: 10.1007/s12035-014-8958-4] [Citation(s) in RCA: 153] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2014] [Accepted: 10/21/2014] [Indexed: 12/19/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) has a neurotrophic support on neuron of central nervous system (CNS) and is a key molecule in the maintenance of synaptic plasticity and memory storage in hippocampus. However, changes of BDNF level and expression have been reported in the CNS as well as blood of Alzheimer's disease (AD) patients in the last decade, which indicates a potential role of BDNF in the pathogenesis of AD. Therefore, this review aims to summarize the latest progress in the field of BDNF and its biological roles in AD pathogenesis. We will discuss the interaction between BDNF and amyloid beta (Aβ) peptide, the effect of BDNF on synaptic repair in AD, and the association between BDNF polymorphism and AD risk. The most important is, enlightening the detailed biological ability and complicated mechanisms of action of BDNF in the context of AD would provide a future BDNF-related remedy for AD, such as increment in the production or release of endogenous BDNF by some drugs or BDNF mimics.
Collapse
|
179
|
Liu JH, You QL, Wei MD, Wang Q, Luo ZY, Lin S, Huang L, Li SJ, Li XW, Gao TM. Social Isolation During Adolescence Strengthens Retention of Fear Memories and Facilitates Induction of Late-Phase Long-Term Potentiation. Mol Neurobiol 2014; 52:1421-1429. [PMID: 25860250 PMCID: PMC4588096 DOI: 10.1007/s12035-014-8917-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2014] [Accepted: 09/28/2014] [Indexed: 12/23/2022]
Abstract
Social isolation during the vulnerable period of adolescence produces emotional dysregulation that often manifests as abnormal behavior in adulthood. The enduring consequence of isolation might be caused by a weakened ability to forget unpleasant memories. However, it remains unclear whether isolation affects unpleasant memories. To address this, we used a model of associative learning to induce the fear memories and evaluated the influence of isolation mice during adolescence on the subsequent retention of fear memories and its underlying cellular mechanisms. Following adolescent social isolation, we found that mice decreased their social interaction time and had an increase in anxiety-related behavior. Interestingly, when we assessed memory retention, we found that isolated mice were unable to forget aversive memories when tested 4 weeks after the original event. Consistent with this, we observed that a single train of high-frequency stimulation (HFS) enabled a late-phase long-term potentiation (L-LTP) in the hippocampal CA1 region of isolated mice, whereas only an early-phase LTP was observed with the same stimulation in the control mice. Social isolation during adolescence also increased brain-derived neurotrophic factor (BDNF) expression in the hippocampus, and application of a tropomyosin-related kinase B (TrkB) receptor inhibitor ameliorated the facilitated L-LTP seen after isolation. Together, our results suggest that adolescent isolation may result in mental disorders during adulthood and that this may stem from an inability to forget the unpleasant memories via BDNF-mediated synaptic plasticity. These findings may give us a new strategy to prevent mental disorders caused by persistent unpleasant memories.
Collapse
Affiliation(s)
- Ji-Hong Liu
- State Key Laboratory of Organ Failure Research, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.,Key Laboratory of Psychiatric Disorders of Guangdong Province, Southern Medical University, Guangzhou, 510515, China
| | - Qiang-Long You
- State Key Laboratory of Organ Failure Research, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.,Key Laboratory of Psychiatric Disorders of Guangdong Province, Southern Medical University, Guangzhou, 510515, China
| | - Mei-Dan Wei
- Department of Pharmacy, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, China
| | - Qian Wang
- State Key Laboratory of Organ Failure Research, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.,Key Laboratory of Psychiatric Disorders of Guangdong Province, Southern Medical University, Guangzhou, 510515, China
| | - Zheng-Yi Luo
- State Key Laboratory of Organ Failure Research, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.,Key Laboratory of Psychiatric Disorders of Guangdong Province, Southern Medical University, Guangzhou, 510515, China
| | - Song Lin
- State Key Laboratory of Organ Failure Research, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.,Key Laboratory of Psychiatric Disorders of Guangdong Province, Southern Medical University, Guangzhou, 510515, China
| | - Lang Huang
- State Key Laboratory of Organ Failure Research, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.,Key Laboratory of Psychiatric Disorders of Guangdong Province, Southern Medical University, Guangzhou, 510515, China
| | - Shu-Ji Li
- State Key Laboratory of Organ Failure Research, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.,Key Laboratory of Psychiatric Disorders of Guangdong Province, Southern Medical University, Guangzhou, 510515, China
| | - Xiao-Wen Li
- State Key Laboratory of Organ Failure Research, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.,Key Laboratory of Psychiatric Disorders of Guangdong Province, Southern Medical University, Guangzhou, 510515, China
| | - Tian-Ming Gao
- State Key Laboratory of Organ Failure Research, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China. .,Key Laboratory of Psychiatric Disorders of Guangdong Province, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
180
|
Leal G, Afonso PM, Salazar IL, Duarte CB. Regulation of hippocampal synaptic plasticity by BDNF. Brain Res 2014; 1621:82-101. [PMID: 25451089 DOI: 10.1016/j.brainres.2014.10.019] [Citation(s) in RCA: 292] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 10/10/2014] [Accepted: 10/13/2014] [Indexed: 01/01/2023]
Abstract
The neurotrophin brain-derived neurotrophic factor (BDNF) has emerged as a major regulator of activity-dependent plasticity at excitatory synapses in the mammalian central nervous system. In particular, much attention has been given to the role of the neurotrophin in the regulation of hippocampal long-term potentiation (LTP), a sustained enhancement of excitatory synaptic strength believed to underlie learning and memory processes. In this review we summarize the evidence pointing to a role for BDNF in generating functional and structural changes at synapses required for both early- and late phases of LTP in the hippocampus. The available information regarding the pre- and/or postsynaptic release of BDNF and action of the neurotrophin during LTP will be also reviewed. Finally, we discuss the effects of BDNF on the synaptic proteome, either by acting on the protein synthesis machinery and/or by regulating protein degradation by calpains and possibly by the ubiquitin-proteasome system (UPS). This fine-tuned control of the synaptic proteome rather than a simple upregulation of the protein synthesis may play a key role in BDNF-mediated synaptic potentiation. This article is part of a Special Issue entitled SI: Brain and Memory.
Collapse
Affiliation(s)
- Graciano Leal
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| | - Pedro M Afonso
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Ivan L Salazar
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal; PhD Programme in Experimental Biology and Biomedicine (PDBEB) and Institute for Interdisciplinary Research, University of Coimbra (IIIUC), 3030-789 Coimbra, Portugal
| | - Carlos B Duarte
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal; Department of Life Sciences, University of Coimbra, 3004-517 Coimbra, Portugal.
| |
Collapse
|
181
|
Yu Y, Cao DQ, Xu HY, Sun M, Huang ZL, Yung WH, Lu N, Huang Y. 5-HT3A receptors are required in long-term depression and AMPA receptor internalization. Neuroscience 2014; 278:105-12. [DOI: 10.1016/j.neuroscience.2014.07.070] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2014] [Revised: 07/11/2014] [Accepted: 07/24/2014] [Indexed: 11/28/2022]
|
182
|
Non-coding RNA regulation of synaptic plasticity and memory: implications for aging. Ageing Res Rev 2014; 17:34-42. [PMID: 24681292 DOI: 10.1016/j.arr.2014.03.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 03/14/2014] [Accepted: 03/18/2014] [Indexed: 12/31/2022]
Abstract
Advancing age is associated with the loss of cognitive ability and vulnerability to debilitating mental diseases. Although much is known about the development of cognitive processes in the brain, the study of the molecular mechanisms governing memory decline with aging is still in its infancy. Recently, it has become apparent that most of the human genome is transcribed into non-coding RNAs (ncRNAs) rather than protein-coding mRNAs. Multiple types of ncRNAs are enriched in the central nervous system, and this large group of molecules may regulate the molecular complexity of the brain, its neurons, and synapses. Here, we review the current knowledge on the role of ncRNAs in synaptic plasticity, learning, and memory in the broader context of the aging brain and associated memory loss. We also discuss future directions to study the role of ncRNAs in the aging process.
Collapse
|
183
|
Dong Y, Nestler EJ. The neural rejuvenation hypothesis of cocaine addiction. Trends Pharmacol Sci 2014; 35:374-83. [PMID: 24958329 DOI: 10.1016/j.tips.2014.05.005] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Revised: 05/27/2014] [Accepted: 05/27/2014] [Indexed: 12/16/2022]
Abstract
A leading hypothesis guiding current molecular and cellular research into drug addiction conceptualizes key aspects of addiction as a form of memory in which common neuroplasticity mechanisms that mediate normal learning and memory processes are 'hijacked' by exposure to drugs of abuse to produce pathologic addiction-related memories. Such addiction-related memories are particularly robust and long-lasting and once formed are less amenable to updating. Here we propose a neural rejuvenation hypothesis of cocaine addiction. According to this hypothesis, repeated exposure to drugs of abuse induces some plasticity mechanisms normally associated with brain development within the reward circuitry that mediate the highly efficient and unusually stable memory abnormalities that characterize addiction.
Collapse
Affiliation(s)
- Yan Dong
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA15260, USA.
| | - Eric J Nestler
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
184
|
Chang PKY, Prenosil GA, Verbich D, Gill R, McKinney RA. Prolonged ampakine exposure prunes dendritic spines and increases presynaptic release probability for enhanced long-term potentiation in the hippocampus. Eur J Neurosci 2014; 40:2766-76. [PMID: 24925283 DOI: 10.1111/ejn.12638] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Revised: 04/24/2014] [Accepted: 04/28/2014] [Indexed: 01/19/2023]
Abstract
CX 546, an allosteric positive modulator of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid-type ionotropic glutamate receptors (AMPARs), belongs to a drug class called ampakines. These compounds have been shown to enhance long-term potentiation (LTP), a cellular model of learning and memory, and improve animal learning task performance, and have augmented cognition in neurodegenerative patients. However, the chronic effect of CX546 on synaptic structures has not been examined. The structure and integrity of dendritic spines are thought to play a role in learning and memory, and their abnormalities have been implicated in cognitive disorders. In addition, their structural plasticity has been shown to be important for cognitive function, such that dendritic spine remodeling has been proposed as the morphological correlate for LTP. Here, we tested the effect of CX546 on dendritic spine remodeling following long-term treatment. We found that, with prolonged CX546 treatment, organotypic hippocampal slice cultures showed a significant reduction in CA3-CA1 excitatory synapse and spine density. Electrophysiological approaches revealed that the CA3-CA1 circuitry compensates for this synapse loss by increasing synaptic efficacy through enhancement of presynaptic release probability. CX546-treated slices showed prolonged and enhanced potentiation upon LTP induction. Furthermore, structural plasticity, namely spine head enlargement, was also more pronounced after CX546 treatment. Our results suggest a concordance of functional and structural changes that is enhanced with prolonged CX546 exposure. Thus, the improved cognitive ability of patients receiving ampakine treatment may result from the priming of synapses through increases in the structural plasticity and functional reliability of hippocampal synapses.
Collapse
Affiliation(s)
- Philip K-Y Chang
- Department of Pharmacology & Therapeutics, McGill University, Bellini Life Science Complex, Room 167, 3649 Promenade Sir-William-Osler, Montreal, QC, H3G 0B1, Canada
| | | | | | | | | |
Collapse
|
185
|
Li J, Gu J, Wang B, Xie M, Huang L, Liu Y, Zhang L, Xue J, Guo F, Zhang L, Zhang L. Activation of Dopamine D1 Receptors Regulates Dendritic Morphogenesis Through Rac1 and RhoA in Prefrontal Cortex Neurons. Mol Neurobiol 2014; 51:1024-37. [PMID: 24915967 DOI: 10.1007/s12035-014-8762-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 05/22/2014] [Indexed: 12/22/2022]
Abstract
Dopamine (DA) is an important regulator of neuronal plasticity in the prefrontal cortex (PFC) and plays a critical role in addiction-related neuroadaptation. The Rho GTPases, including Rac1, RhoA and Cdc42, are key regulators of actin cytoskeleton rearrangement that play important roles in dendritic morphogenesis. The goal of the current study was to use cultures of primary PFC neurons to gain a better understanding of the molecular mechanisms underlying DA-induced dendritic morphogenesis, a phenomenon that mimics the increase in DA synaptic transmission observed in the PFC of in vivo cocaine administration. We investigated the effects of repeated DA treatments on dendritic morphology changes in PFC neurons, and identified Rac1 and RhoA as downstream effectors of D1 receptors during the regulation of dendritic morphogenesis. Importantly, we found that D1 receptor-regulated Rac1 and RhoA have distinct roles in the regulation of dendritic morphogenesis after repeated DA treatments. Our data provide the first evidence that Rac1 and RhoA are effectors of D1 receptor signaling during dendritic morphogenesis and represent new signaling molecules involved in long-lasting neuroadaptation in the PFC.
Collapse
Affiliation(s)
- Juan Li
- Key Laboratory of Functional Proteomics of Guangdong Province, Department of Pathophysiology, Southern Medical University, Guangzhou, 510515, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
186
|
Exposure to 1800 MHz radiofrequency radiation impairs neurite outgrowth of embryonic neural stem cells. Sci Rep 2014; 4:5103. [PMID: 24869783 PMCID: PMC4037711 DOI: 10.1038/srep05103] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 05/09/2014] [Indexed: 01/31/2023] Open
Abstract
A radiofrequency electromagnetic field (RF-EMF) of 1800 MHz is widely used in mobile communications. However, the effects of RF-EMFs on cell biology are unclear. Embryonic neural stem cells (eNSCs) play a critical role in brain development. Thus, detecting the effects of RF-EMF on eNSCs is important for exploring the effects of RF-EMF on brain development. Here, we exposed eNSCs to 1800 MHz RF-EMF at specific absorption rate (SAR) values of 1, 2, and 4 W/kg for 1, 2, and 3 days. We found that 1800 MHz RF-EMF exposure did not influence eNSC apoptosis, proliferation, cell cycle or the mRNA expressions of related genes. RF-EMF exposure also did not alter the ratio of eNSC differentiated neurons and astrocytes. However, neurite outgrowth of eNSC differentiated neurons was inhibited after 4 W/kg RF-EMF exposure for 3 days. Additionally, the mRNA and protein expression of the proneural genes Ngn1 and NeuroD, which are crucial for neurite outgrowth, were decreased after RF-EMF exposure. The expression of their inhibitor Hes1 was upregulated by RF-EMF exposure. These results together suggested that 1800 MHz RF-EMF exposure impairs neurite outgrowth of eNSCs. More attention should be given to the potential adverse effects of RF-EMF exposure on brain development.
Collapse
|
187
|
Goldental A, Guberman S, Vardi R, Kanter I. A computational paradigm for dynamic logic-gates in neuronal activity. Front Comput Neurosci 2014; 8:52. [PMID: 24808856 PMCID: PMC4010740 DOI: 10.3389/fncom.2014.00052] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 04/07/2014] [Indexed: 01/12/2023] Open
Abstract
In 1943 McCulloch and Pitts suggested that the brain is composed of reliable logic-gates similar to the logic at the core of today's computers. This framework had a limited impact on neuroscience, since neurons exhibit far richer dynamics. Here we propose a new experimentally corroborated paradigm in which the truth tables of the brain's logic-gates are time dependent, i.e., dynamic logic-gates (DLGs). The truth tables of the DLGs depend on the history of their activity and the stimulation frequencies of their input neurons. Our experimental results are based on a procedure where conditioned stimulations were enforced on circuits of neurons embedded within a large-scale network of cortical cells in-vitro. We demonstrate that the underlying biological mechanism is the unavoidable increase of neuronal response latencies to ongoing stimulations, which imposes a non-uniform gradual stretching of network delays. The limited experimental results are confirmed and extended by simulations and theoretical arguments based on identical neurons with a fixed increase of the neuronal response latency per evoked spike. We anticipate our results to lead to better understanding of the suitability of this computational paradigm to account for the brain's functionalities and will require the development of new systematic mathematical methods beyond the methods developed for traditional Boolean algebra.
Collapse
Affiliation(s)
- Amir Goldental
- Department of Physics, Bar-Ilan UniversityRamat-Gan, Israel
| | - Shoshana Guberman
- Department of Physics, Bar-Ilan UniversityRamat-Gan, Israel
- The Goodman Faculty of Life Sciences, Gonda Interdisciplinary Brain Research Center, Bar-Ilan UniversityRamat-Gan, Israel
| | - Roni Vardi
- The Goodman Faculty of Life Sciences, Gonda Interdisciplinary Brain Research Center, Bar-Ilan UniversityRamat-Gan, Israel
| | - Ido Kanter
- Department of Physics, Bar-Ilan UniversityRamat-Gan, Israel
- The Goodman Faculty of Life Sciences, Gonda Interdisciplinary Brain Research Center, Bar-Ilan UniversityRamat-Gan, Israel
| |
Collapse
|
188
|
Soltész F, Suckling J, Lawrence P, Tait R, Ooi C, Bentley G, Dodds CM, Miller SR, Wille DR, Byrne M, McHugh SM, Bellgrove MA, Croft RJ, Lu B, Bullmore ET, Nathan PJ. Identification of BDNF sensitive electrophysiological markers of synaptic activity and their structural correlates in healthy subjects using a genetic approach utilizing the functional BDNF Val66Met polymorphism. PLoS One 2014; 9:e95558. [PMID: 24760076 PMCID: PMC3997566 DOI: 10.1371/journal.pone.0095558] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 03/28/2014] [Indexed: 12/18/2022] Open
Abstract
Increasing evidence suggests that synaptic dysfunction is a core pathophysiological hallmark of neurodegenerative disorders. Brain-derived neurotropic factor (BDNF) is key synaptogenic molecule and targeting synaptic repair through modulation of BDNF signalling has been suggested as a potential drug discovery strategy. The development of such "synaptogenic" therapies depend on the availability of BDNF sensitive markers of synaptic function that could be utilized as biomarkers for examining target engagement or drug efficacy in humans. Here we have utilized the BDNF Val66Met genetic polymorphism to examine the effect of the polymorphism and genetic load (i.e. Met allele load) on electrophysiological (EEG) markers of synaptic activity and their structural (MRI) correlates. Sixty healthy adults were prospectively recruited into the three genetic groups (Val/Val, Val/Met, Met/Met). Subjects also underwent fMRI, tDCS/TMS, and cognitive assessments as part of a larger study. Overall, some of the EEG markers of synaptic activity and brain structure measured with MRI were the most sensitive markers of the polymorphism. Met carriers showed decreased oscillatory activity and synchrony in the neural network subserving error-processing, as measured during a flanker task (ERN); and showed increased slow-wave activity during resting. There was no evidence for a Met load effect on the EEG measures and the polymorphism had no effects on MMN and P300. Met carriers also showed reduced grey matter volume in the anterior cingulate and in the (left) prefrontal cortex. Furthermore, anterior cingulate grey matter volume, and oscillatory EEG power during the flanker task predicted subsequent behavioural adaptation, indicating a BDNF dependent link between brain structure, function and behaviour associated with error processing and monitoring. These findings suggest that EEG markers such as ERN and resting EEG could be used as BDNF sensitive functional markers in early clinical development to examine target engagement or drug related efficacy of synaptic repair therapies in humans.
Collapse
Affiliation(s)
- Fruzsina Soltész
- Clinical Unit Cambridge, GlaxoSmithKline, Cambridge, United Kingdom
| | - John Suckling
- Brain Mapping Unit, Department of Psychiatry, University of Cambridge, United Kingdom
| | - Phil Lawrence
- Clinical Unit Cambridge, GlaxoSmithKline, Cambridge, United Kingdom
| | - Roger Tait
- Brain Mapping Unit, Department of Psychiatry, University of Cambridge, United Kingdom
| | - Cinly Ooi
- Brain Mapping Unit, Department of Psychiatry, University of Cambridge, United Kingdom
| | - Graham Bentley
- Clinical Unit Cambridge, GlaxoSmithKline, Cambridge, United Kingdom
| | - Chris M. Dodds
- Department of Psychology, University of Exeter, Exeter, United Kingdom
| | - Sam R. Miller
- Clinical Unit Cambridge, GlaxoSmithKline, Cambridge, United Kingdom
| | - David R. Wille
- Clinical Unit Cambridge, GlaxoSmithKline, Cambridge, United Kingdom
| | - Misha Byrne
- Queensland Brain Institute, University of Queensland, Queensland, Australia
| | - Simon M. McHugh
- Clinical Unit Cambridge, GlaxoSmithKline, Cambridge, United Kingdom
| | - Mark A. Bellgrove
- School of Psychology and Psychiatry, Monash University, Melbourne, Australia
| | - Rodney J. Croft
- Queensland Brain Institute, University of Queensland, Queensland, Australia
| | - Bai Lu
- Tsinghua University Medical School, Beijing, China
| | - Edward T. Bullmore
- Brain Mapping Unit, Department of Psychiatry, University of Cambridge, United Kingdom
| | - Pradeep J. Nathan
- Brain Mapping Unit, Department of Psychiatry, University of Cambridge, United Kingdom
- School of Psychology and Psychiatry, Monash University, Melbourne, Australia
- New Medicines, UCB Pharma, Brussels, Belgium
| |
Collapse
|
189
|
Garrett CA, Barri M, Kuta A, Soura V, Deng W, Fisher EMC, Schiavo G, Hafezparast M. DYNC1H1 mutation alters transport kinetics and ERK1/2-cFos signalling in a mouse model of distal spinal muscular atrophy. ACTA ACUST UNITED AC 2014; 137:1883-93. [PMID: 24755273 DOI: 10.1093/brain/awu097] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Mutations in the gene encoding the heavy chain subunit (DYNC1H1) of cytoplasmic dynein cause spinal muscular atrophy with lower extremity predominance, Charcot-Marie-Tooth disease and intellectual disability. We used the legs at odd angles (Loa) (DYNC1H1(F580Y)) mouse model for spinal muscular atrophy with lower extremity predominance and a combination of live-cell imaging and biochemical assays to show that the velocity of dynein-dependent microtubule minus-end (towards the nucleus) movement of EGF and BDNF induced signalling endosomes is significantly reduced in Loa embryonic fibroblasts and motor neurons. At the same time, the number of the plus-end (towards the cell periphery) moving endosomes is increased in the mutant cells. As a result, the extracellular signal-regulated kinases (ERK) 1/2 activation and c-Fos expression are altered in both mutant cell types, but the motor neurons exhibit a strikingly abnormal ERK1/2 and c-Fos response to serum-starvation induced stress. These data highlight the cell-type specific ERK1/2 response as a possible contributory factor in the neuropathological nature of Dync1h1 mutations, despite generic aberrant kinetics in both cell types, providing an explanation for how mutations in the ubiquitously expressed DYNC1H1 cause neuron-specific disease.
Collapse
Affiliation(s)
- Caroline A Garrett
- 1 School of Life Sciences, University of Sussex, Falmer, Brighton, BN1 9QG, UK
| | - Muruj Barri
- 1 School of Life Sciences, University of Sussex, Falmer, Brighton, BN1 9QG, UK
| | - Anna Kuta
- 2 Department of Neurodegenerative Disease, Institute of Neurology, University College London, Queen Square, London WC1N 3BG, UK
| | - Violetta Soura
- 1 School of Life Sciences, University of Sussex, Falmer, Brighton, BN1 9QG, UK
| | - Wenhan Deng
- 1 School of Life Sciences, University of Sussex, Falmer, Brighton, BN1 9QG, UK
| | - Elizabeth M C Fisher
- 2 Department of Neurodegenerative Disease, Institute of Neurology, University College London, Queen Square, London WC1N 3BG, UK
| | - Giampietro Schiavo
- 3 Sobell Department of Motor Neuroscience and Movement Disorders, Institute of Neurology, University College London, Queen Square, London WC1N 3BG, UK
| | - Majid Hafezparast
- 1 School of Life Sciences, University of Sussex, Falmer, Brighton, BN1 9QG, UK
| |
Collapse
|
190
|
Vermehren-Schmaedick A, Krueger W, Jacob T, Ramunno-Johnson D, Balkowiec A, Lidke KA, Vu TQ. Heterogeneous intracellular trafficking dynamics of brain-derived neurotrophic factor complexes in the neuronal soma revealed by single quantum dot tracking. PLoS One 2014; 9:e95113. [PMID: 24732948 PMCID: PMC3986401 DOI: 10.1371/journal.pone.0095113] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 03/23/2014] [Indexed: 01/19/2023] Open
Abstract
Accumulating evidence underscores the importance of ligand-receptor dynamics in shaping cellular signaling. In the nervous system, growth factor-activated Trk receptor trafficking serves to convey biochemical signaling that underlies fundamental neural functions. Focus has been placed on axonal trafficking but little is known about growth factor-activated Trk dynamics in the neuronal soma, particularly at the molecular scale, due in large part to technical hurdles in observing individual growth factor-Trk complexes for long periods of time inside live cells. Quantum dots (QDs) are intensely fluorescent nanoparticles that have been used to study the dynamics of ligand-receptor complexes at the plasma membrane but the value of QDs for investigating ligand-receptor intracellular dynamics has not been well exploited. The current study establishes that QD conjugated brain-derived neurotrophic factor (QD-BDNF) binds to TrkB receptors with high specificity, activates TrkB downstream signaling, and allows single QD tracking capability for long recording durations deep within the soma of live neurons. QD-BDNF complexes undergo internalization, recycling, and intracellular trafficking in the neuronal soma. These trafficking events exhibit little time-synchrony and diverse heterogeneity in underlying dynamics that include phases of sustained rapid motor transport without pause as well as immobility of surprisingly long-lasting duration (several minutes). Moreover, the trajectories formed by dynamic individual BDNF complexes show no apparent end destination; BDNF complexes can be found meandering over long distances of several microns throughout the expanse of the neuronal soma in a circuitous fashion. The complex, heterogeneous nature of neuronal soma trafficking dynamics contrasts the reported linear nature of axonal transport data and calls for models that surpass our generally limited notions of nuclear-directed transport in the soma. QD-ligand probes are poised to provide understanding of how the molecular mechanisms underlying intracellular ligand-receptor trafficking shape cell signaling under conditions of both healthy and dysfunctional neurological disease models.
Collapse
Affiliation(s)
- Anke Vermehren-Schmaedick
- Department of Biomedical Engineering and Center for Spatial Systems Biomedicine, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Wesley Krueger
- Department of Physics & Astronomy, University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Thomas Jacob
- Department of Biomedical Engineering and Center for Spatial Systems Biomedicine, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Damien Ramunno-Johnson
- Department of Biomedical Engineering and Center for Spatial Systems Biomedicine, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Agnieszka Balkowiec
- Department of Integrative Biosciences, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Keith A. Lidke
- Department of Physics & Astronomy, University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Tania Q. Vu
- Department of Biomedical Engineering and Center for Spatial Systems Biomedicine, Oregon Health & Science University, Portland, Oregon, United States of America
- * E-mail:
| |
Collapse
|
191
|
Zhang K, Duan L, Ong Q, Lin Z, Varman PM, Sung K, Cui B. Light-mediated kinetic control reveals the temporal effect of the Raf/MEK/ERK pathway in PC12 cell neurite outgrowth. PLoS One 2014; 9:e92917. [PMID: 24667437 PMCID: PMC3965503 DOI: 10.1371/journal.pone.0092917] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2013] [Accepted: 02/26/2014] [Indexed: 11/29/2022] Open
Abstract
It has been proposed that differential activation kinetics allows cells to use a common set of signaling pathways to specify distinct cellular outcomes. For example, nerve growth factor (NGF) and epidermal growth factor (EGF) induce different activation kinetics of the Raf/MEK/ERK signaling pathway and result in differentiation and proliferation, respectively. However, a direct and quantitative linkage between the temporal profile of Raf/MEK/ERK activation and the cellular outputs has not been established due to a lack of means to precisely perturb its signaling kinetics. Here, we construct a light-gated protein-protein interaction system to regulate the activation pattern of the Raf/MEK/ERK signaling pathway. Light-induced activation of the Raf/MEK/ERK cascade leads to significant neurite outgrowth in rat PC12 pheochromocytoma cell lines in the absence of growth factors. Compared with NGF stimulation, light stimulation induces longer but fewer neurites. Intermittent on/off illumination reveals that cells achieve maximum neurite outgrowth if the off-time duration per cycle is shorter than 45 min. Overall, light-mediated kinetic control enables precise dissection of the temporal dimension within the intracellular signal transduction network.
Collapse
Affiliation(s)
- Kai Zhang
- Department of Chemistry, Stanford University, Stanford, California, United States of America
| | - Liting Duan
- Department of Chemistry, Stanford University, Stanford, California, United States of America
| | - Qunxiang Ong
- Department of Chemistry, Stanford University, Stanford, California, United States of America
| | - Ziliang Lin
- Department of Applied Physics, Stanford University, Stanford, California, United States of America
| | - Pooja Mahendra Varman
- Department of Chemistry, Stanford University, Stanford, California, United States of America
| | - Kijung Sung
- Biophysics Program, Stanford University, Stanford, California, United States of America
| | - Bianxiao Cui
- Department of Chemistry, Stanford University, Stanford, California, United States of America
- * E-mail:
| |
Collapse
|
192
|
Kellner Y, Gödecke N, Dierkes T, Thieme N, Zagrebelsky M, Korte M. The BDNF effects on dendritic spines of mature hippocampal neurons depend on neuronal activity. Front Synaptic Neurosci 2014; 6:5. [PMID: 24688467 PMCID: PMC3960490 DOI: 10.3389/fnsyn.2014.00005] [Citation(s) in RCA: 117] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 03/01/2014] [Indexed: 01/05/2023] Open
Abstract
The fine tuning of neural networks during development and learning relies upon both functional and structural plastic processes. Changes in the number as well as in the size and shape of dendritic spines are associated to long-term activity-dependent synaptic plasticity. However, the molecular mechanisms translating functional into structural changes are still largely unknown. In this context, neurotrophins, like Brain-Derived Neurotrophic Factor (BDNF), are among promising candidates. Specifically BDNF-TrkB receptor signaling is crucial for activity-dependent strengthening of synapses in different brain regions. BDNF application has been shown to positively modulate dendritic and spine architecture in cortical and hippocampal neurons as well as structural plasticity in vitro. However, a global BDNF deprivation throughout the central nervous system (CNS) resulted in very mild structural alterations of dendritic spines, questioning the relevance of the endogenous BDNF signaling in modulating the development and the mature structure of neurons in vivo. Here we show that a loss-of-function approach, blocking BDNF results in a significant reduction in dendritic spine density, associated with an increase in spine length and a decrease in head width. These changes are associated with a decrease in F-actin levels within spine heads. On the other hand, a gain-of-function approach, applying exogenous BDNF, could not reproduce the increase in spine density or the changes in spine morphology previously described. Taken together, we show here that the effects exerted by BDNF on the dendritic architecture of hippocampal neurons are dependent on the neuron's maturation stage. Indeed, in mature hippocampal neurons in vitro as shown in vivo BDNF is specifically required for the activity-dependent maintenance of the mature spine phenotype.
Collapse
Affiliation(s)
- Yves Kellner
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig Braunschweig, Germany
| | - Nina Gödecke
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig Braunschweig, Germany
| | - Tobias Dierkes
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig Braunschweig, Germany
| | - Nils Thieme
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig Braunschweig, Germany
| | - Marta Zagrebelsky
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig Braunschweig, Germany
| | - Martin Korte
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig Braunschweig, Germany
| |
Collapse
|
193
|
Guo W, Ji Y, Wang S, Sun Y, Lu B. Neuronal activity alters BDNF-TrkB signaling kinetics and downstream functions. J Cell Sci 2014; 127:2249-60. [PMID: 24634513 DOI: 10.1242/jcs.139964] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Differential kinetics of the same signaling pathway might elicit different cellular outcomes. Here, we show that high-frequency neuronal activity converts BDNF-induced TrkB (also known as NTRK2) signaling from a transient to a sustained mode. A prior depolarization (15 mM KCl, 1 hour) resulted in a long-lasting (>24 hours) activation of the TrkB receptor and its downstream signaling, which otherwise lasts less than an hour. The long-term potentiation (LTP)-inducing theta-burst stimulation but not the long-term depression (LTD)-inducing low-frequency stimulation also induced sustained activation of TrkB. This sustained signaling facilitated dendritic branching and rescued neuronal apoptosis induced by glutamate. The change in TrkB signaling kinetics is mediated by Ca(2+) elevation and CaMKII activation, leading to an increase in TrkB expression on the neuronal surface. Physical exercise also alters the kinetics of TrkB phosphorylation induced by exogenous BDNF. Sustained TrkB signaling might serve as a key mechanism underlying the synergistic effects of neuronal activity and BDNF.
Collapse
Affiliation(s)
- Wei Guo
- Tsinghua-Peking Center for Life Sciences, Beijing, China School of Medicine, Tsinghua University, 1 Qinghuayuan Road, Beijing, 100084, China School of Life Sciences, Tsinghua University, 1 Qinghuayuan Road, Beijing, 100084, China
| | - Yuanyuan Ji
- GlaxoSmithKline, R&D China, Building 3, 898 Halei Road, Zhangjiang Hi-tech Park, Pudong, Shanghai, 201203, China
| | - Shudan Wang
- Tsinghua-Peking Center for Life Sciences, Beijing, China School of Life Sciences, Tsinghua University, 1 Qinghuayuan Road, Beijing, 100084, China
| | - Yun Sun
- Tsinghua-Peking Center for Life Sciences, Beijing, China National Institute of Biological Sciences, Beijing, 102206, China School of Life Sciences, Peking University, Beijing, 100871, China
| | - Bai Lu
- Tsinghua-Peking Center for Life Sciences, Beijing, China School of Medicine, Tsinghua University, 1 Qinghuayuan Road, Beijing, 100084, China
| |
Collapse
|
194
|
Tian N, Cao Z, Zhang Y. MiR-206 decreases brain-derived neurotrophic factor levels in a transgenic mouse model of Alzheimer's disease. Neurosci Bull 2014; 30:191-7. [PMID: 24604632 DOI: 10.1007/s12264-013-1419-7] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 11/06/2013] [Indexed: 01/01/2023] Open
Abstract
MicroRNA alterations have been reported in patients with Alzheimer's disease (AD) and AD mouse models. We now report that miR-206 is upregulated in the hippocampal tissue, cerebrospinal fluid, and plasma of embryonic APP/PS1 transgenic mice. The increased miR-206 downregulates the expression of brain-derived neurotrophic factor (BDNF). BDNF is neuroprotective against cell death after various insults, but in embryonic and newborn APP/PS1 mice it is decreased. Thus, a specific microRNA alteration may contribute to AD pathology by downregulating BDNF.
Collapse
Affiliation(s)
- Ning Tian
- Beijing Obstetrics & Gynecology Hospital, Capital Medical University, Beijing, 100026, China
| | | | | |
Collapse
|
195
|
Fon D, Zhou K, Ercole F, Fehr F, Marchesan S, Minter MR, Crack PJ, Finkelstein DI, Forsythe JS. Nanofibrous scaffolds releasing a small molecule BDNF-mimetic for the re-direction of endogenous neuroblast migration in the brain. Biomaterials 2014; 35:2692-712. [PMID: 24406218 DOI: 10.1016/j.biomaterials.2013.12.016] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 12/10/2013] [Indexed: 01/19/2023]
Abstract
Brain tissue engineering has the potential to harness existing elements of neurogenesis within the adult brain to overcome a microenvironment that is otherwise inhibitory to regeneration, especially following severe tissue damage. This study investigates the ability of electrospun poly ε-caprolactone (PCL) to re-direct the migratory pathway of endogenous neuroblasts from the disrupted subventricular zone (SVZ). A small molecule non-peptide ligand (BDNF-mimetic) that mimicked the trophic properties of brain-derived neurotrophic factor (BDNF) was incorporated into electrospun PCL scaffolds to improve neuroblast survival and promote neuroblast migration towards the implant. PCL scaffolds were able to support neuroblast infiltration and migration along the implant tract. In the presence of the BDNF-mimetic, neuroblasts were able to migrate towards the implant via the parenchyma, and their persistence within the implants was prolonged. In addition, the BDNF-mimetic improved implant integration and increased local neuronal plasticity by increasing neurite sprouting at the tissue-implant interface. SMI32+ neurites were observed inside scaffolds at 21 days but not 8 days post implantation, indicating that at least some of the infiltrated neuroblasts had differentiated into neurons.
Collapse
Affiliation(s)
- Deniece Fon
- Department of Materials Engineering, Monash University, Clayton, VIC 3800, Australia; Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Kun Zhou
- Department of Materials Engineering, Monash University, Clayton, VIC 3800, Australia
| | - Francesca Ercole
- Department of Materials Engineering, Monash University, Clayton, VIC 3800, Australia
| | - Friederike Fehr
- CSIRO Materials Science and Engineering, Clayton, VIC 3053, Australia
| | - Silvia Marchesan
- CSIRO Materials Science and Engineering, Clayton, VIC 3053, Australia
| | - Myles R Minter
- Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Peter J Crack
- Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, VIC 3010, Australia
| | - David I Finkelstein
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3010, Australia.
| | - John S Forsythe
- Department of Materials Engineering, Monash University, Clayton, VIC 3800, Australia.
| |
Collapse
|
196
|
Higa GSV, de Sousa E, Walter LT, Kinjo ER, Resende RR, Kihara AH. MicroRNAs in neuronal communication. Mol Neurobiol 2014; 49:1309-26. [PMID: 24385256 DOI: 10.1007/s12035-013-8603-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Accepted: 12/05/2013] [Indexed: 12/28/2022]
Abstract
MicroRNAs (miRNAs) are short nucleotides sequences that regulate the expression of genes in different eukaryotic cell types. A tremendous amount of knowledge on miRNAs has rapidly accumulated over the last few years, revealing the growing interest in this field of research. On the other hand, clarifying the physiological regulation of gene expression in the central nervous system is important for establishing a reference for comparison to the diseased state. It is well known that the fine tuning of neuronal networks relies on intricate molecular mechanisms, such as the adjustment of the synaptic transmission. As determined by recent studies, regulation of neuronal interactions by miRNAs has critical consequences in the development, adaptation to ambient demands, and degeneration of the nervous system. In contrast, activation of synaptic receptors triggers downstream signaling cascades that generate a vast array of effects, which includes the regulation of novel genes involved in the control of the miRNA life cycle. In this review, we have examined the hot topics on miRNA gene-regulatory activities in the broad field of neuronal communication-related processes. Furthermore, in addition to indicating the newly described effect of miRNAs on the regulation of specific neurotransmitter systems, we have pointed out how these systems affect the expression, transport, and stability of miRNAs. Moreover, we discuss newly described and under-investigation mechanisms involving the intercellular transfer of miRNAs, aided by exosomes and gap junctions. Thus, in the current review, we were able to highlight recent findings related to miRNAs that indisputably contributed towards the understanding of the nervous system in health and disease.
Collapse
Affiliation(s)
- Guilherme Shigueto Vilar Higa
- Núcleo de Cognição e Sistemas Complexos, Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, Av. Atlântica 420, 09060-000, Santo André, SP, Brazil
| | | | | | | | | | | |
Collapse
|
197
|
Form follows function: BDNF and its involvement in sculpting the function and structure of synapses. Neuropharmacology 2014; 76 Pt C:628-38. [DOI: 10.1016/j.neuropharm.2013.05.029] [Citation(s) in RCA: 160] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 05/13/2013] [Accepted: 05/15/2013] [Indexed: 12/31/2022]
|
198
|
Lu B, Nagappan G, Lu Y. BDNF and synaptic plasticity, cognitive function, and dysfunction. Handb Exp Pharmacol 2014; 220:223-50. [PMID: 24668475 DOI: 10.1007/978-3-642-45106-5_9] [Citation(s) in RCA: 671] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Among all neurotrophins, brain-derived neurotrophic factor (BDNF) stands out for its high level of expression in the brain and its potent effects on synapses. It is now widely accepted that the main function of BDNF in the adult brain is to regulate synapses, with structural and functional effects ranging from short-term to long-lasting, on excitatory or inhibitory synapses, in many brain regions. The diverse effects of BDNF on brain synapses stem from its complex downstream signaling cascades, as well as the diametrically opposing effects of the pro- and mature form through distinct receptors, TrkB and p75(NTR). Many aspects of BDNF cell biology are regulated by neuronal activity. The synergistic interactions between neuronal activity and synaptic plasticity by BDNF make it an ideal and essential regulator of cellular processes that underlie cognition and other complex behaviors. Indeed, numerous studies firmly established that BDNF plays a critical role in hippocampal long-term potentiation (LTP), a long-term enhancement of synaptic efficacy thought to underlie learning and memory. Converging evidence now strongly suggest that deficits in BDNF signaling contribute to the pathogenesis of several major diseases and disorders such as Huntington's disease, Alzheimer's disease, and depression. Thus, manipulating BDNF pathways represents a viable treatment approach to a variety of neurological and psychiatric disorders.
Collapse
Affiliation(s)
- B Lu
- GlaxoSmithKline, R&D China, Building 3, 898 Halei Road, Zhangjiang Hi-tech Park, Pudong, Shanghai, 201203, China,
| | | | | |
Collapse
|
199
|
Protective effect of Homer 1a on tumor necrosis factor-α with cycloheximide-induced apoptosis is mediated by mitogen-activated protein kinase pathways. Apoptosis 2013; 17:975-88. [PMID: 22660975 DOI: 10.1007/s10495-012-0736-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Although Homer 1, of the postsynaptic density, regulates apoptosis, the signaling mechanisms are not fully elucidated. In this study, we found that tumor necrosis factor-α (TNF-α)/cycloheximide (CHX) treatment transiently increased Homer 1a (the short variant of Homer 1), but did not affect Homer 1b/c (the long variant of Homer 1). Overexpression of Homer 1a blocked TNF-α/CHX-induced apoptotic cell death, whereas inhibition of Homer 1a induction enhanced the pro-apoptotic effect of TNF-α/CHX treatment. Moreover, brain-derived neurotrophic factor, as a potential activator of endogenous Homer 1a, inhibited apoptotic cell death after TNF-α/CHX treatment through induction of Homer 1a. Since three major mitogen-activated protein kinase (MAPK) pathways have important roles in apoptosis, we examined if Homer 1a is involved in the effects of MAPK pathways on apoptosis. It was shown that inhibition of the ERK1/2 pathway increased the expression and the protective effect of Homer 1a, but inhibition of the p38 pathway produced the opposite effect. Cross-talk among MAPK pathways was also associated with the regulation of Homer 1a during apoptotic cell death. Blocking the p38 pathway increased the activity in the ERK1/2 pathway, while inhibition of ERK1/2 pathway abolished the effect of p38 inhibitor on Homer 1a. Furthermore, Homer 1a reversely affected the activation of MAPK pathways. These findings suggest that Homer 1a plays an important role in the prevention of apoptotic cell death and contributes to distinct regulatory effects of MAPK pathways on apoptotic cell death.
Collapse
|
200
|
Su B, Ji YS, Sun XL, Liu XH, Chen ZY. Brain-derived neurotrophic factor (BDNF)-induced mitochondrial motility arrest and presynaptic docking contribute to BDNF-enhanced synaptic transmission. J Biol Chem 2013; 289:1213-26. [PMID: 24302729 DOI: 10.1074/jbc.m113.526129] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Appropriate mitochondrial transport and distribution are essential for neurons because of the high energy and Ca(2+) buffering requirements at synapses. Brain-derived neurotrophic factor (BDNF) plays an essential role in regulating synaptic transmission and plasticity. However, whether and how BDNF can regulate mitochondrial transport and distribution are still unclear. Here, we find that in cultured hippocampal neurons, application of BDNF for 15 min decreased the percentage of moving mitochondria in axons, a process dependent on the activation of the TrkB receptor and its downstream PI3K and phospholipase-Cγ signaling pathways. Moreover, the BDNF-induced mitochondrial stopping requires the activation of transient receptor potential canonical 3 and 6 (TRPC3 and TRPC6) channels and elevated intracellular Ca(2+) levels. The Ca(2+) sensor Miro1 plays an important role in this process. Finally, the BDNF-induced mitochondrial stopping leads to the accumulation of more mitochondria at presynaptic sites. Mutant Miro1 lacking the ability to bind Ca(2+) prevents BDNF-induced mitochondrial presynaptic accumulation and synaptic transmission, suggesting that Miro1-mediated mitochondrial motility is involved in BDNF-induced mitochondrial presynaptic docking and neurotransmission. Together, these data suggest that mitochondrial transport and distribution play essential roles in BDNF-mediated synaptic transmission.
Collapse
Affiliation(s)
- Bo Su
- From the Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, School of Medicine, Shandong University, Jinan, Shandong 250012, China
| | | | | | | | | |
Collapse
|