1
|
Zhou Q, Chen Y, Tang H, Zhang L, Ma Y, Bai D, Kong Y. Transcranial direct current stimulation alleviated ischemic stroke induced injury involving the BDNF-TrkB signaling axis in rats. Heliyon 2023; 9:e14946. [PMID: 37089354 PMCID: PMC10114158 DOI: 10.1016/j.heliyon.2023.e14946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 03/11/2023] [Accepted: 03/22/2023] [Indexed: 04/08/2023] Open
Abstract
Ischemic stroke causes a complicated sequence of apoptotic cascades leading to neuronal damage and functional impairments. Transcranial direct current stimulation (tDCS) is a non-invasive treatment technique that uses electrodes to deliver weak current to the head. It could influence brain activity and has a crucial role in neuronal survival and plasticity. The current study investigated the neuroprotective effects and potential mechanisms of tDCS by brain-derived neurotrophic factor (BDNF) and its related receptor tropomyosin-receptor kinase B (TrkB) against apoptosis following ischemic injury in vivo. The effect of consecutive treatment with tDCS for seven days on rats after Middle cerebral artery occlusion/reperfusion (MCAO/R) surgery was studied. Western blotting, immunofluorescent staining, TUNEL assay, and electron microscope were conducted seven days after tDCS treatment, and the motor function was assessed at 1, 3, and 7 days. Activities of BDNF-TrkB signaling axis and apoptosis-related proteins were determined in the cerebral cortex. At seven days after tDCS treatment, it increased BDNF levels and promoted the regeneration of axons compared with the MCAO/R group. There was also a reduction in neuronal apoptosis and improved functional deficits. Whereafter, a TrkB receptor inhibitor K252a was administrated to clarify whether the neuroprotection of tDCS is exerted via BDNF-TrkB signaling. The results depicted that K252a application significantly inhibited the neuroprotection impact of tDCS treatment. It was accompanied by a significant downregulation of phosphorylation of TrkB, PI3K, and Akt. Our study investigated the neuroprotective effects of tDCS against ischemic injury. The results indicate that upregulation of BDNF and its critical receptor TrkB, as well as its downstream PI3K/Akt pathway, were involved in the protective effects exerted by tDCS.
Collapse
|
2
|
Choi MS, Chung YY, Kim DJ, Kim ST, Jun YH. Immunoreactivity of MAPK Signaling in a Rat Model of Intrauterine Growth Retardation Induced by Uterine Artery Ligation. In Vivo 2020; 34:649-657. [PMID: 32111765 DOI: 10.21873/invivo.11819] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 12/21/2019] [Accepted: 01/05/2020] [Indexed: 11/10/2022]
Abstract
BACKGROUND/AIM Intrauterine growth retardation (IUGR) causes very low birth weight and is related to the morbidity and mortality of the newborn. In our previous study, expression of brain-derived neurotrophic factor (BDNF) was found reduced in the cerebral cortex and dentate gyrus of fetuses with IUGR. BDNF protected cortical neurons against hypoxic injury via activation of the extracellular signal-related kinase (ERK) pathway. The aim of the current study was to observe the immunoreactivity of ERK in mature neurons and proliferating cells. MATERIALS AND METHODS Uterine artery ligation was performed at 17 days of gestation (dg). Rat fetuses were obtained at 21 dg using cesarean section. Fetuses were designated either to the growth retardation (GR) group when removed from the horn with uterine artery ligation, or to the control group when removed from the other horn with the untied artery. Immunohistochemistry was performed with primary antibodies on paraffin-embedded forebrain sections. RESULTS The density and proportion of cells expressing PCNA, ERK, and phosphate ERK in the subventricular zone (SVZ) was not different between the control and GR group. The density and proportion of NeuN- and phosphate ERK-positive cells in the cerebral parietal cortex was lower in the GR group, compared to the control group. CONCLUSION Although IUGR had no effect on the proliferation of cells in the SVZ, it reduced neuronal survival in the cerebral parietal cortex, which was associated with the decrease of pERK-positive cell density and proportion in the cerebral cortex.
Collapse
Affiliation(s)
- Min Seon Choi
- Department of Pediatrics, Chosun University Hospital, Gwang-ju, Republic of Korea
| | - Yoon Young Chung
- Department of Anatomy, School of Medicine, Chosun University, Gwang-ju, Republic of Korea
| | - Dong-Joon Kim
- Department of Anesthesiology and Pain Medicine, Chosun University Hospital, Gwang-ju, Republic of Korea
| | - Seong Taeck Kim
- Department of Ophthalmology, Chosun University Hospital, Gwang-ju, Republic of Korea
| | - Yong Hyun Jun
- Department of Anatomy, School of Medicine, Chosun University, Gwang-ju, Republic of Korea
| |
Collapse
|
3
|
Kumar V, Ali Shariati M, Mesentier-Louro L, Jinsook Oh A, Russano K, Goldberg JL, Liao YJ. Dual Specific Phosphatase 14 Deletion Rescues Retinal Ganglion Cells and Optic Nerve Axons after Experimental Anterior Ischemic Optic Neuropathy. Curr Eye Res 2020; 46:710-718. [PMID: 33107352 PMCID: PMC8291381 DOI: 10.1080/02713683.2020.1826976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PURPOSE Understanding molecular changes is essential for designing effective treatments for nonarteritic anterior ischemic optic neuropathy (AION), the most common acute optic neuropathy in adults older than 50 years. We investigated changes in the mitogen-activated protein kinase (MAPK) pathway after experimental AION and focused on dual specificity phosphatase 14 (Dusp14), an atypical MAPK phosphatase that is downstream of Krüppel-like transcription factor (KLF) 9-mediated inhibition of retinal ganglion cell (RGC) survival and axonal regeneration. MATERIALS AND METHODS We induced severe AION in a photochemical thrombosis model in adult C57BL/6 wild-type and Dusp14 knockout mice. For comparison, some studies were performed using an optic nerve crush model. We assessed changes in MAPK pathway molecules using Western blot and immunohistochemistry, measured retinal thickness using optical coherence tomography (OCT), and quantified RGCs and axons using histologic methods. RESULTS Three days after severe AION, there was no change in the retinal protein levels of MAPK ERK1/2, phosphorylated-ERK1/2 (pERK1/2), downstream effector Elk-1 and phosphatase Dusp14 on Western blot. Western blot analysis of purified RGCs after a more severe model using optic nerve crush also showed no change in Dusp14 protein expression. Because of the known importance of the Dusp14 and MAPK pathway in RGCs, we examined changes after AION in Dusp14 knockout mice. Three days after AION, Dusp14 knockout mice had significantly increased pERK1/2+, Brn3A+ RGCs on immunohistochemistry. Three weeks after AION, Dusp14 knockout mice had significantly greater preservation of retinal thickness, increased number of Brn3A+ RGCs on whole mount preparation, and increased number of optic nerve axons compared with wild-type mice. CONCLUSIONS Genetic deletion of Dusp14, a MAPK phosphatase important in KFL9-mediated inhibition of RGC survival, led to increased activation of MAPK ERK1/2 and greater RGC and axonal survival after experimental AION. Inhibiting Dusp14 or activating the MAPK pathway should be examined further as a potential therapeutic approach to treatment of AION. Abbreviations: AION: anterior ischemic optic neuropathy; Dusp14: dual specific phosphatase 14; ERK1/2: extracellular signal-regulated kinases 1/2; Elk-1: ETS Like-1 protein; GCC: ganglion cell complex; GCL: ganglion cell layer; inner nuclear layer; KO: knockout; MAPK: mitogen-activated phosphokinase; OCT: optical coherence tomography; RGC: retinal ganglion cell; RNFL: retinal nerve fiber layer.
Collapse
Affiliation(s)
- Varun Kumar
- Spencer Center for Vision Research, Byers Eye Institute, Palo Alto, California, USA.,Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Harvard University, Boston, Massachusetts, USA
| | | | | | - Angela Jinsook Oh
- Spencer Center for Vision Research, Byers Eye Institute, Palo Alto, California, USA
| | - Kristina Russano
- Spencer Center for Vision Research, Byers Eye Institute, Palo Alto, California, USA
| | - Jeffrey L Goldberg
- Spencer Center for Vision Research, Byers Eye Institute, Palo Alto, California, USA
| | - Yaping Joyce Liao
- Spencer Center for Vision Research, Byers Eye Institute, Palo Alto, California, USA.,Department of Neurology, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
4
|
Jun YH, Kim ST. Brain-derived neurotrophic factor in non-proliferative diabetic retinopathy with diabetic macular edema. Eur J Ophthalmol 2020; 31:1915-1919. [PMID: 32686489 DOI: 10.1177/1120672120944801] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
PURPOSE To investigate aqueous humor (AH) and serum levels of brain-derived neurotrophic factor (BDNF) in non-proliferative diabetic retinopathy (NPDR) patients with diabetic macular edema (DME). METHODS The prospective study consists of 20 patients with DME NPDR, 20 patients with no-DME NPDR, and 20 healthy control subjects. Serum and AH samples were obtained during cataract surgery and intravitreal injection. Serum and AH levels of BDNF were measured by enzyme-linked immunosorbent assay. RESULTS The mean serum levels of BDNF were lower in both NPDR groups compared to the control group (DME NPDR group, p = 0.015; no-DME NPDR group, p = 0.024). Furthermore, the mean serum level of BDNF was lower in the DME NPDR group compared to the no-DME NPDR group (p = 0.041). The mean AH levels of BDNF were significantly reduced in both NPDR groups compared to the control group (DME NPDR group, p < 0.001; no-DME NPDR group, p = 0.006). Further, the mean AH level of BDNF was significantly lower in the DME NPDR group compared to the no-DME NPDR group (p = 0.037). CONCLUSION Serum and AH levels of BDNF were reduced in NPDR patients with DME than without DME.
Collapse
Affiliation(s)
- Yong Hyun Jun
- Department of Anatomy, School of Medicine, Chosun University, Gwangju, Republic of Korea
| | - Seong Taeck Kim
- Department of Ophthalmology, School of Medicine, Chosun University, Gwangju, Republic of Korea
| |
Collapse
|
5
|
Gaidin SG, Turovskaya MV, Gavrish MS, Babaev AA, Mal'tseva VN, Blinova EV, Turovsky EA. The selective BDNF overexpression in neurons protects neuroglial networks against OGD and glutamate-induced excitotoxicity. Int J Neurosci 2019; 130:363-383. [PMID: 31694441 DOI: 10.1080/00207454.2019.1691205] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Objective: Cerebral ischemia is accompanied by damage and death of a significant number of neurons due to glutamate excitotoxicity with subsequent a global increase of cytosolic Ca2+ concentration ([Ca2+]i). This study aimed to investigate the neuroprotective action of BDNF overexpression in hippocampal neurons against injury under ischemia-like conditions (oxygen and glucose deprivation) and glutamate-induced excitotoxicity (GluTox).Methods: The overexpression of BDNF was reached by the transduction of cell cultures with the adeno-associated (AAV)-Syn-BDNF-EGFP virus construct. Neuroprotective effects were mediated by Ca2+-dependent BDNF release followed by activation of the neuroprotective signaling cascades and changes of the gene expression. Thus, BDNF overexpression modulates Ca2+ homeostasis in cells, preventing Ca2+ overload and initiation of apoptotic and necrotic processes.Results:Antiapoptotic effect of BDNF overexpression is mediated via activation of phosphoinositide-3-kinase (PI3K) pathway and changing the expression of PI3K, HIF-1, Src and an anti-inflammatory cytokine IL-10. On the contrary, the decrease of expression of proapoptotic proteins such as Jun, Mapk8, caspase-3 and an inflammatory cytokine IL-1β was observed. These changes of expression were accompanied by the decrease of quantity of IL-1β receptors and the level of TNFα in cells in control, as well as 24 h after OGD. Besides, BDNF overexpression changes the expression of GABA(B) receptors. Also, the expression of NMDA and AMPA receptor subunits was altered towards a change in the conductivity of the receptors for Ca2+.Conclusion: Thus, our results demonstrate that neuronal BDNF overexpression reveals complex neuroprotective effects on the neurons and astrocytes under OGD and GluTox via inhibition of Ca2+ responses and regulation of gene expression.
Collapse
Affiliation(s)
- S G Gaidin
- Institute of Cell Biophysics, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Russian Academy of Sciences, Pushchino, Russia
| | - M V Turovskaya
- Institute of Cell Biophysics, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Russian Academy of Sciences, Pushchino, Russia
| | - M S Gavrish
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - A A Babaev
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - V N Mal'tseva
- Institute of Cell Biophysics, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Russian Academy of Sciences, Pushchino, Russia
| | - E V Blinova
- I.M. Sechenov First Moscow State Medical University, Moscow, Russia.,N. P. Ogarev Mordovia State University, Saransk, Russia
| | - E A Turovsky
- Institute of Cell Biophysics, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Russian Academy of Sciences, Pushchino, Russia
| |
Collapse
|
6
|
Kletkiewicz H, Maliszewska J, Jaworski K, Jermacz Ł, Smoliński DJ, Rogalska J. Thermal conditions during neonatal anoxia affect the endogenous level of brain-derived neurotrophic factor. J Neurosci Res 2019; 97:1266-1277. [PMID: 31257630 DOI: 10.1002/jnr.24486] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 06/11/2019] [Accepted: 06/11/2019] [Indexed: 12/19/2022]
Abstract
Anoxia during delivery is a complication that can disturb infant brain development leading to various types of neurological disorders. Our studies have shown that increased body temperature of newborn rats of both sexes intensifies the postanoxic oxidative stress and prevents triggering the endogenous adaptive response such as HIF-1α activation. Currently, brain-derived neurotrophic factor-BDNF is considered to be a modulator of neuronal plasticity. In the developing brain, mature BDNF and its precursor exhibit prosurvival action through the TrkB receptor and proapoptotic functions binding to p75NTR , respectively. The aim of our experiments was to check the effects of body temperature on the postanoxic level of BDNF and on the expression of its receptors as well as on the marker of apoptosis-caspase-3 in the rat brain. Two-day-old Wistar Han rats (male/female ratio, 1:1) were exposed to anoxia in 100% nitrogen atmosphere for 10 min in different thermal conditions, which allowed them to regulate their rectal temperature at the following levels: normothermic-33°C; hyperthermic-37°C; and extremely hyperthermic-39°C. Thermal conditions during neonatal anoxia affected the level of proBDNF, BDNF as well as their receptors and caspase-3 in the forebrain. The increased BDNF protein level followed by decreased caspase-3 protein level was probably dependent on body temperature under anoxic conditions and was observed only in rats maintaining decreased body temperature. The positive effect of BDNF was not observed under hyperthermic conditions. Moreover, BDNF level changes correlated with body temperature probably affected the learning and spatial memory in juvenile rats.
Collapse
Affiliation(s)
- Hanna Kletkiewicz
- Department of Animal Physiology, Faculty of Biology and Environmental Protection, Nicolaus Copernicus University, Toruń, Poland
| | - Justyna Maliszewska
- Department of Animal Physiology, Faculty of Biology and Environmental Protection, Nicolaus Copernicus University, Toruń, Poland
| | - Krzysztof Jaworski
- Chair of Plant Physiology and Biotechnology, Faculty of Biology and Environmental Protection, Nicolaus Copernicus University, Toruń, Poland
| | - Łukasz Jermacz
- Department of Hydrobiology, Faculty of Biology and Environmental Protection, Nicolaus Copernicus University, Toruń, Poland
| | - Dariusz J Smoliński
- Department of Cellular and Molecular Biology, Faculty of Biology and Environmental Protection, Nicolaus Copernicus University, Toruń, Poland.,Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University, Toruń, Poland
| | - Justyna Rogalska
- Department of Animal Physiology, Faculty of Biology and Environmental Protection, Nicolaus Copernicus University, Toruń, Poland
| |
Collapse
|
7
|
González‐Rodríguez P, Ugidos IF, Pérez‐Rodríguez D, Anuncibay‐Soto B, Santos‐Galdiano M, Font‐Belmonte E, Gonzalo‐Orden JM, Fernández‐López A. Brain‐derived neurotrophic factor alleviates the oxidative stress induced by oxygen and glucose deprivation in an ex vivo brain slice model. J Cell Physiol 2018; 234:9592-9604. [DOI: 10.1002/jcp.27646] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 09/02/2018] [Indexed: 01/22/2023]
Affiliation(s)
| | - Irene F. Ugidos
- Área de Biología Celular, Instituto de Biomedicina, University of León León Spain
| | | | - Berta Anuncibay‐Soto
- Área de Biología Celular, Instituto de Biomedicina, University of León León Spain
| | | | | | | | | |
Collapse
|
8
|
Chronic asthma-induced behavioral and hippocampal neuronal morphological changes are concurrent with BDNF, cofilin1 and Cdc42/RhoA alterations in immature mice. Brain Res Bull 2018; 143:194-206. [PMID: 30227235 DOI: 10.1016/j.brainresbull.2018.09.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 09/03/2018] [Accepted: 09/11/2018] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Recent studies have found that persistent hypoxia caused by chronic asthma, especially during childhood, affects the development and function of the brain, but the mechanism is unclear. In the present study, BDNF and its signal pathway was investigated in mediating chronic asthma induced-neuronal changes that lead to behavior alterations. METHODS The chronic asthma model was induced by sensitization with ovalbumin for more than 9 weeks in immature mice. Morris water maze test (MWMT), open field test (OFT) and elevated plus maze test (EPMT) were used to conduct behavioral evaluation. Neuronal morphology in hippocampal CA1, CA3 and DG was assessed using ImageJ's Sholl plugin and RESCONSTRUCT software. BDNF signaling pathway related molecules was determined by Western blotting. RESULTS Chronic asthma does affect the behavioral performances of immature mice evaluated in MWMT, OFT, and EPMT. The analysis by three-dimensional reconstruction software found that following the behavioral alteration of asthmatic mice, dendritic changes also occurred in hippocampal neurons, including shortened dendrite length, significantly reduced number of dendritic branches, decreased density of dendritic spines, and reduced percentage of functional dendritic spine types. At the same time, by immunofluorescence and western blotting, we also found that alterations in dendritic morphology were consistent with activation of cofilin1 and changes in BDNF-Cdc42/RhoA levels. Some of the changes mentioned above can be alleviated by intranasal administration of budesonide. CONCLUSION Our data suggest that response similar to nicotine withdrawal or/and hypoxia induced by childhood chronic asthma enhances the BDNF-Cdc42/RhoA signaling pathway and activates cofilin1, leading to the remodeling of actin, causing the loss of dendritic spines and atrophy of dendrites, eventually resulting in behavioral alterations.
Collapse
|
9
|
Hung PL, Hsu MH, Yu HR, Wu KLH, Wang FS. Thyroxin Protects White Matter from Hypoxic-Ischemic Insult in the Immature Sprague⁻Dawley Rat Brain by Regulating Periventricular White Matter and Cortex BDNF and CREB Pathways. Int J Mol Sci 2018; 19:ijms19092573. [PMID: 30158497 PMCID: PMC6164053 DOI: 10.3390/ijms19092573] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 08/20/2018] [Accepted: 08/21/2018] [Indexed: 11/16/2022] Open
Abstract
Background: Periventricular white-matter (WM) injury is a prominent feature of brain injury in preterm infants. Thyroxin (T4) treatment reduces the severity of hypoxic-ischemic (HI)-mediated WM injury in the immature brain. This study aimed to delineate molecular events underlying T4 protection following periventricular WM injury in HI rats. Methods: Right common-carotid-artery ligation, followed by hypoxia, was performed on seven-day-old rat pups. The HI pups were injected with saline, or 0.2 or 1 mg/kg of T4 at 48–96 h postoperatively. Cortex and periventricular WM were dissected for real-time (RT)-quantitative polymerase chain reactions (PCRs), immunoblotting, and for immunofluorescence analysis of neurotrophins, myelin, oligodendrocyte precursors, and neointimal. Results: T4 significantly mitigated hypomyelination and oligodendrocyte death in HI pups, whereas angiogenesis of periventricular WM, observed using antiendothelium cell antibody (RECA-1) immunofluorescence and vascular endothelium growth factor (VEGF) immunoblotting, was not affected. T4 also increased the brain-derived neurotrophic factors (BDNFs), but not the nerve growth factor (NGF) expression of injured periventricular WM. However, phosphorylated extracellular signal regulated kinase (p-ERK) and phosphorylated cyclic adenosine monophosphate response element-binding protein (p-CREB) concentrations, but not the BDNF downstream pathway kinases, p38, c-Jun amino-terminal kinase (c-JNK), or Akt, were reduced in periventricular WM with T4 treatment. Notably, T4 administration significantly increased BDNF and phosphorylated CREB in the overlying cortex of the HI-induced injured cortex. Conclusion: Our findings reveal that T4 reversed BNDF signaling to attenuate HI-induced WM injury by activating ERK and CREB pathways in the cortex, but not directly in periventricular WM. This study offers molecular insight into the neuroprotective actions of T4 in HI-mediated WM injury in the immature brain.
Collapse
Affiliation(s)
- Pi-Lien Hung
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 33303, Taiwan.
| | - Mei-Hsin Hsu
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 33303, Taiwan.
| | - Hong-Ren Yu
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 33303, Taiwan.
| | - Kay L H Wu
- Center for Translational Research in Biomedical Sciences, College of Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, Kaohsiung 33303, Taiwan.
| | - Feng-Sheng Wang
- Core facility for Phenomics & Diagnostics, Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, No123, Rd Ta-Pei, Niao-Song District, Kaohsiung 33303, Taiwan.
| |
Collapse
|
10
|
AAV-Syn-BDNF-EGFP Virus Construct Exerts Neuroprotective Action on the Hippocampal Neural Network during Hypoxia In Vitro. Int J Mol Sci 2018; 19:ijms19082295. [PMID: 30081596 PMCID: PMC6121472 DOI: 10.3390/ijms19082295] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/01/2018] [Accepted: 08/03/2018] [Indexed: 02/07/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is one of the key signaling molecules that supports the viability of neural cells in various brain pathologies, and can be considered a potential therapeutic agent. However, several methodological difficulties, such as overcoming the blood–brain barrier and the short half-life period, challenge the potential use of BDNF in clinical practice. Gene therapy could overcome these limitations. Investigating the influence of viral vectors on the neural network level is of particular interest because viral overexpression affects different aspects of cell metabolism and interactions between neurons. The present work aimed to investigate the influence of the adeno-associated virus (AAV)-Syn-BDNF-EGFP virus construct on neural network activity parameters in an acute hypobaric hypoxia model in vitro. Materials and methods. An adeno-associated virus vector carrying the BDNF gene was constructed using the following plasmids: AAV-Syn-EGFP, pDP5, DJvector, and pHelper. The developed virus vector was then tested on primary hippocampal cultures obtained from C57BL/6 mouse embryos (E18). Acute hypobaric hypoxia was induced on day 21 in vitro. Spontaneous bioelectrical and calcium activity of neural networks in primary cultures and viability tests were analysed during normoxia and during the posthypoxic period. Results. BDNF overexpression by AAV-Syn-BDNF-EGFP does not affect cell viability or the main parameters of spontaneous bioelectrical activity in normoxia. Application of the developed virus construct partially eliminates the negative hypoxic consequences by preserving cell viability and maintaining spontaneous bioelectrical activity in the cultures. Moreover, the internal functional structure, including the activation pattern of network bursts, the number of hubs, and the number of connections within network elements, is also partially preserved. BDNF overexpression prevents a decrease in the number of cells exhibiting calcium activity and maintains the frequency of calcium oscillations. Conclusion. This study revealed the pronounced antihypoxic and neuroprotective effects of AAV-Syn-BDNF-EGFP virus transduction in an acute normobaric hypoxia model.
Collapse
|
11
|
Tecuatl C, Herrrera-López G, Martín-Ávila A, Yin B, Weber S, Barrionuevo G, Galván EJ. TrkB-mediated activation of the phosphatidylinositol-3-kinase/Akt cascade reduces the damage inflicted by oxygen-glucose deprivation in area CA3 of the rat hippocampus. Eur J Neurosci 2018; 47:1096-1109. [PMID: 29480936 PMCID: PMC5938095 DOI: 10.1111/ejn.13880] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 02/17/2018] [Accepted: 02/20/2018] [Indexed: 12/31/2022]
Abstract
The selective vulnerability of hippocampal area CA1 to ischemia-induced injury is a well-known phenomenon. However, the cellular mechanisms that confer resistance to area CA3 against ischemic damage remain elusive. Here, we show that oxygen-glucose deprivation-reperfusion (OGD-RP), an in vitro model that mimic the pathological conditions of the ischemic stroke, increases the phosphorylation level of tropomyosin receptor kinase B (TrkB) in area CA3. Slices preincubated with brain-derived neurotrophic factor (BDNF) or 7,8-dihydroxyflavone (7,8-DHF) exhibited reduced depression of the electrical activity triggered by OGD-RP. Consistently, blockade of TrkB suppressed the resistance of area CA3 to OGD-RP. The protective effect of TrkB activation was limited to area CA3, as OGD-RP caused permanent suppression of CA1 responses. At the cellular level, TrkB activation leads to phosphorylation of the accessory proteins SHC and Gab as well as the serine/threonine kinase Akt, members of the phosphoinositide 3-kinase/Akt (PI-3-K/Akt) pathway, a cascade involved in cell survival. Hence, acute slices pretreated with the Akt antagonist MK2206 in combination with BDNF lost the capability to resist the damage inflicted with OGD-RP. Consistently, with these results, CA3 pyramidal cells exhibited reduced propidium iodide uptake and caspase-3 activity in slices pretreated with BDNF and exposed to OGD-RP. We propose that PI-3-K/Akt downstream activation mediated by TrkB represents an endogenous mechanism responsible for the resistance of area CA3 to ischemic damage.
Collapse
Affiliation(s)
- Carolina Tecuatl
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Calzada de los Tenorios No. 235, México City, 14330, México
| | - Gabriel Herrrera-López
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Calzada de los Tenorios No. 235, México City, 14330, México
| | - Alejandro Martín-Ávila
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Calzada de los Tenorios No. 235, México City, 14330, México
| | - Bocheng Yin
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Stephen Weber
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Germán Barrionuevo
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Emilio J. Galván
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Calzada de los Tenorios No. 235, México City, 14330, México
| |
Collapse
|
12
|
Berberine Ameliorates MCAO Induced Cerebral Ischemia/Reperfusion Injury via Activation of the BDNF–TrkB–PI3K/Akt Signaling Pathway. Neurochem Res 2018; 43:702-710. [DOI: 10.1007/s11064-018-2472-4] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 12/25/2017] [Accepted: 01/09/2018] [Indexed: 11/25/2022]
|
13
|
Kovács V, Tóth-Szűki V, Németh J, Varga V, Remzső G, Domoki F. Active forms of Akt and ERK are dominant in the cerebral cortex of newborn pigs that are unaffected by asphyxia. Life Sci 2017; 192:1-8. [PMID: 29138115 DOI: 10.1016/j.lfs.2017.11.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 11/03/2017] [Accepted: 11/10/2017] [Indexed: 01/13/2023]
Abstract
AIMS Perinatal asphyxia (PA) often results in hypoxic-ischemic encephalopathy (HIE) in term neonates. Introduction of therapeutic hypothermia improved HIE outcome, but further neuroprotective therapies are still warranted. The present study sought to determine the feasibility of the activation of the cytoprotective PI-3-K/Akt and the MAPK/ERK signaling pathways in the subacute phase of HIE development in a translational newborn pig PA/HIE model. MAIN METHODS Phosphorylated and total levels of Akt and ERK were determined by Western blotting in brain samples obtained from untreated naive, time control, and PA/HIE animals at 24-48h survival (n=3-3-6,respectively). PA (20min) was induced in anesthetized piglets by ventilation with a hypoxic/hypercapnic (6%O220%CO2) gas mixture. Furthermore, we studied the effect of topically administered specific Akt1/2 and MAPK/ERK kinase inhibitors on Akt and ERK phosphorylation (n=4-4) in the cerebral cortex under normoxic conditions. KEY FINDINGS PA resulted in significant neuronal injury shown by neuropathology assessment of haematoxylin/eosin stained sections. However, there were no significant differences among the groups in the high phosphorylation levels of both ERK and Akt in the cerebral cortex, hippocampus and subcortical structures. However, the Akt1/2 and MAPK/ERK kinase inhibitors significantly reduced cerebrocortical Akt and ERK phosphorylation within 30min. SIGNIFICANCE The major finding of the present study is that the PI-3-K/Akt and the MAPK/ERK signaling pathways appear to be constitutively active in the piglet brain, and this activation remains unaltered during HIE development. Thus, neuroprotective strategies aiming to activate these pathways to limit apoptotic neuronal death may offer limited efficacy in this translational model.
Collapse
Affiliation(s)
- Viktória Kovács
- Department of Physiology, University of Szeged, School of Medicine, Szeged, Hungary.
| | - Valéria Tóth-Szűki
- Department of Physiology, University of Szeged, School of Medicine, Szeged, Hungary
| | - János Németh
- Department of Physiology, University of Szeged, School of Medicine, Szeged, Hungary
| | - Viktória Varga
- Department of Physiology, University of Szeged, School of Medicine, Szeged, Hungary
| | - Gábor Remzső
- Department of Physiology, University of Szeged, School of Medicine, Szeged, Hungary
| | - Ferenc Domoki
- Department of Physiology, University of Szeged, School of Medicine, Szeged, Hungary
| |
Collapse
|
14
|
Terraneo L, Samaja M. Comparative Response of Brain to Chronic Hypoxia and Hyperoxia. Int J Mol Sci 2017; 18:ijms18091914. [PMID: 28880206 PMCID: PMC5618563 DOI: 10.3390/ijms18091914] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 09/01/2017] [Accepted: 09/03/2017] [Indexed: 12/25/2022] Open
Abstract
Two antithetic terms, hypoxia and hyperoxia, i.e., insufficient and excess oxygen availability with respect to needs, are thought to trigger opposite responses in cells and tissues. This review aims at summarizing the molecular and cellular mechanisms underlying hypoxia and hyperoxia in brain and cerebral tissue, a context that may prove to be useful for characterizing not only several clinically relevant aspects, but also aspects related to the evolution of oxygen transport and use by the tissues. While the response to acute hypoxia/hyperoxia presumably recruits only a minor portion of the potentially involved cell machinery, focusing into chronic conditions, instead, enables to take into consideration a wider range of potential responses to oxygen-linked stress, spanning from metabolic to genic. We will examine how various brain subsystems, including energetic metabolism, oxygen sensing, recruitment of pro-survival pathways as protein kinase B (Akt), mitogen-activated protein kinases (MAPK), neurotrophins (BDNF), erythropoietin (Epo) and its receptors (EpoR), neuroglobin (Ngb), nitric oxide (NO), carbon monoxide (CO), deal with chronic hypoxia and hyperoxia to end-up with the final outcomes, oxidative stress and brain damage. A more complex than expected pattern results, which emphasizes the delicate balance between the severity of the stress imposed by hypoxia and hyperoxia and the recruitment of molecular and cellular defense patterns. While for certain functions the expectation that hypoxia and hyperoxia should cause opposite responses is actually met, for others it is not, and both emerge as dangerous treatments.
Collapse
Affiliation(s)
- Laura Terraneo
- Department of Health Science, University of Milan, I-20142 Milano, Italy.
| | - Michele Samaja
- Department of Health Science, University of Milan, I-20142 Milano, Italy.
| |
Collapse
|
15
|
Zhao X, Chen R, Liu M, Feng J, Chen J, Hu K. Remodeling the blood-brain barrier microenvironment by natural products for brain tumor therapy. Acta Pharm Sin B 2017; 7:541-553. [PMID: 28924548 PMCID: PMC5595291 DOI: 10.1016/j.apsb.2017.07.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 06/08/2017] [Accepted: 07/12/2017] [Indexed: 12/23/2022] Open
Abstract
Brain tumor incidence shows an upward trend in recent years; brain tumors account for 5% of adult tumors, while in children, this figure has increased to 70%. Moreover, 20%-30% of malignant tumors will eventually metastasize into the brain. Both benign and malignant tumors can cause an increase in intracranial pressure and brain tissue compression, leading to central nervous system (CNS) damage which endangers the patients' lives. Despite the many approaches to treating brain tumors and the progress that has been made, only modest gains in survival time of brain tumor patients have been achieved. At present, chemotherapy is the treatment of choice for many cancers, but the special structure of the blood-brain barrier (BBB) limits most chemotherapeutic agents from passing through the BBB and penetrating into tumors in the brain. The BBB microenvironment contains numerous cell types, including endothelial cells, astrocytes, peripheral cells and microglia, and extracellular matrix (ECM). Many chemical components of natural products are reported to regulate the BBB microenvironment near brain tumors and assist in their treatment. This review focuses on the composition and function of the BBB microenvironment under both physiological and pathological conditions, and the current research progress in regulating the BBB microenvironment by natural products to promote the treatment of brain tumors.
Collapse
Affiliation(s)
- Xiao Zhao
- Murad Research Center for Modernized Chinese Medicine, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Rujing Chen
- Murad Research Center for Modernized Chinese Medicine, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Mei Liu
- Murad Research Center for Modernized Chinese Medicine, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jianfang Feng
- Murad Research Center for Modernized Chinese Medicine, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jun Chen
- Key Laboratory of Smart Drug Delivery, Fudan University, Ministry of Education, Shanghai 201203, China
| | - Kaili Hu
- Murad Research Center for Modernized Chinese Medicine, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
16
|
Yuan JH, Pan F, Chen J, Chen CE, Xie DP, Jiang XZ, Guo SJ, Zhou J. Neuroprotection by plumbagin involves BDNF-TrkB-PI3K/Akt and ERK1/2/JNK pathways in isoflurane-induced neonatal rats. ACTA ACUST UNITED AC 2017; 69:896-906. [PMID: 28464236 DOI: 10.1111/jphp.12681] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 11/12/2016] [Indexed: 12/19/2022]
Abstract
OBJECTIVES This study was designed to assess the effects of plumbagin on isoflurane-induced neurotoxicity. METHODS Neonatal Sprague Dawley rat pups were treated with plumbagin (50, 100 or 150 mg/kg body weight, orally) from postnatal day 2. The pups on postnatal day 7 were subjected to isoflurane (0.75%) exposure for 6 h. Neuronal apoptosis in the hippocampal tissues was detected by TUNEL assay and FluroJade B staining following isoflurane exposure. Protein expressions were analysed by immunoblotting. RT-PCR was performed to assess mRNA levels of brain-derived neurotrophic factor (BDNF) and TrkB. KEY FINDINGS We observed reduced apoptosis in hippocampal CA1, CA3 and dentate gyrus regions along with severely reduced pro-apoptotic factors (Bad, Bax and cleaved caspase-3) expression and raised levels of Bcl-2, Bcl-xL, survivin, xIAP and cIAPs (cell survival proteins) in plumbagin supplemented rats. Decrease in the levels of JNK, phospho-JNK, c-Jun and phospho-c-Jun with enhanced ERK1/2 levels was observed on plumbagin pretreatment. Down-regulated PI3K/Akt signalling following isoflurane was activated by plumbagin as evidenced by raised PI3K/Akt pathway proteins - mTORc1, Akt, phospho-Akt, GSK-3β, phospho-GSK-3β, PTEN and NF-κBp65 in the hippocampal tissues as detected by Western blotting. The mRNA levels were enhanced on plumbagin supplementation. CONCLUSIONS Plumbagin exerted its neuroprotective effects by effectively suppressing isoflurane-induced neuronal apoptosis via regulating BDNF-TrkB-PI3/Akt and ERK/JNK signalling.
Collapse
Affiliation(s)
- Jun-Hui Yuan
- Department of Neonatology, Wenling Maternal and Child Health Hospital, Wenling, Zhejiang, China
| | - Feng Pan
- Department of Neonatology, Wenling Maternal and Child Health Hospital, Wenling, Zhejiang, China
| | - Jie Chen
- Taizhou University Medical School, Taizhou, Zhejiang, China
| | - Cai-Er Chen
- Department of Neonatology, Wenling Maternal and Child Health Hospital, Wenling, Zhejiang, China
| | - Deng-Pan Xie
- Department of Neonatology, Wenling Maternal and Child Health Hospital, Wenling, Zhejiang, China
| | - Xing-Zhu Jiang
- Department of Neonatology, Wenling Maternal and Child Health Hospital, Wenling, Zhejiang, China
| | - Su-Juan Guo
- Department of Neonatology, Wenling Maternal and Child Health Hospital, Wenling, Zhejiang, China
| | - Jun Zhou
- Taizhou University Medical School, Taizhou, Zhejiang, China
| |
Collapse
|
17
|
Lim W, Bae H, Bazer FW, Song G. Brain-derived neurotrophic factor improves proliferation of endometrial epithelial cells by inhibition of endoplasmic reticulum stress during early pregnancy. J Cell Physiol 2017; 232:3641-3651. [PMID: 28150859 DOI: 10.1002/jcp.25834] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 01/31/2017] [Indexed: 12/23/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is a member of the neurotrophin family binds to two transmembrane receptors; neurotrophic receptor tyrosine kinase 2 (NTRK2) with high affinity and p75 with low affinity. Although BDNF-NTRK2 signaling in the central nervous system is known, signaling in the female reproductive system is unknown. Therefore, we determined effects of BDNF on porcine endometrial luminal epithelial (pLE) cells isolated from Day 12 of pregnancy, as well as expression of BDNF and NTRK2 in endometria of cyclic and pregnant pigs. BDNF-NTRK2 genes were expressed in uterine glandular (GE) and luminal (LE) epithelia during early pregnancy. In addition, their expression in uterine GE and LE decreased with increasing parity of sows. Recombinant BDNF increased proliferation in pLE cells in a dose-dependent, as well as expression of PCNA and Cyclin D1 in nuclei of pLE cells. BDNF also activated phosphorylation of AKT, P70S6K, S6, ERK1/2, JNK, P38 proteins in pLE cells. In addition, cell death resulting from tunicamycin-induced ER stress was prevented when pLE cells were treated with the combination of tunicamycin and BDNF which also decreased cells in the Sub-G1 phase of the cell cycle. Furthermore, tunicamycin-induced unfolded protein response genes were mostly down-regulated to the basal levels as compared to non-treated pLE cells. Our finding suggests that BDNF acts via NTRK2 to induce development of pLE cells for maintenance of implantation and pregnancy by activating cell signaling via the PI3K and MAPK pathways and by inhibiting ER stress.
Collapse
Affiliation(s)
- Whasun Lim
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seongbok-gu, Seoul, Republic of Korea.,Department of Biomedical Sciences, Catholic Kwandong University, Gangneung, Republic of Korea
| | - Hyocheol Bae
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seongbok-gu, Seoul, Republic of Korea
| | - Fuller W Bazer
- Center for Animal Biotechnology and Genomics and Department of Animal Science, Texas A&M University, College Station, Texas
| | - Gwonhwa Song
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seongbok-gu, Seoul, Republic of Korea
| |
Collapse
|
18
|
Giacoppo S, Pollastro F, Grassi G, Bramanti P, Mazzon E. Target regulation of PI3K/Akt/mTOR pathway by cannabidiol in treatment of experimental multiple sclerosis. Fitoterapia 2016; 116:77-84. [PMID: 27890794 DOI: 10.1016/j.fitote.2016.11.010] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 11/11/2016] [Accepted: 11/19/2016] [Indexed: 02/07/2023]
Abstract
This study was aimed to investigate whether treatment with purified cannabidiol (CBD) may counteract the development of experimental multiple sclerosis (MS), by targeting the PI3K/Akt/mTOR pathway. Although the PI3K/Akt/mTOR pathway was found to be activated by cannabinoids in several immune and non-immune cells, currently, there is no data about the effects of CBD in the PI3K/Akt/mTOR activity in MS. Experimental Autoimmune Encephalomyelitis (EAE), the most common model of MS, was induced in C57BL/6 mice by immunization with myelin oligodendroglial glycoprotein peptide (MOG)35-55. After EAE onset, which occurs approximately 14days after disease induction, mice were daily intraperitoneally treated with CBD (10mg/kg mouse) and observed for clinical signs of EAE. At 28days from EAE-induction, mice were euthanized and spinal cord tissues were sampled to perform immunohistochemical evaluations and western blot analysis. Our results showed a clear downregulation of the PI3K/Akt/mTOR pathway following EAE induction. CBD treatment was able to restore it, increasing significantly the phosphorylation of PI3K, Akt and mTOR. Also, an increased level of BNDF in CBD-treated mice seems to be involved in the activation of PI3K/Akt/mTOR pathway. In addition, our data demonstrated that therapeutic efficacy of CBD treatment is due to reduction of pro-inflammatory cytokines, like IFN-γ and IL-17 together with an up-regulation of PPARγ. Finally, CBD was found to promote neuronal survival by inhibiting JNK and p38 MAP kinases. These results provide an interesting discovery about the regulation of the PI3K/Akt/mTOR pathway by cannabidiol administration, that could be a new potential therapeutic target for MS management.
Collapse
Affiliation(s)
- Sabrina Giacoppo
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Federica Pollastro
- Dipartimento di Scienze del Farmaco, Università del Piemonte Orientale, Largo Donegani 2, 28100 Novara, Italy
| | - Gianpaolo Grassi
- Council for Research and Experimentation in Agriculture - Research Centre for Industrial Crops (CRA-CIN), Rovigo, Italy
| | - Placido Bramanti
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy.
| |
Collapse
|
19
|
Jalsrai A, Numakawa T, Kunugi H, Dieterich D, Becker A. The neuroprotective effects and possible mechanism of action of a methanol extract from Asparagus cochinchinensis: In vitro and in vivo studies. Neuroscience 2016; 322:452-63. [DOI: 10.1016/j.neuroscience.2016.02.065] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 02/25/2016] [Accepted: 02/25/2016] [Indexed: 12/14/2022]
|
20
|
Bak DH, Zhang E, Yi MH, Kim DK, Lim K, Kim JJ, Kim DW. High ω3-polyunsaturated fatty acids in fat-1 mice prevent streptozotocin-induced Purkinje cell degeneration through BDNF-mediated autophagy. Sci Rep 2015; 5:15465. [PMID: 26503303 PMCID: PMC4621527 DOI: 10.1038/srep15465] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Accepted: 09/17/2015] [Indexed: 12/14/2022] Open
Abstract
Loss of Purkinje cells has been implicated in the development of diabetic neuropathy, and this degeneration is characterized by impairment of autophagic processes. We evaluated whether fat-1 transgenic mice, a well-established animal model that endogenously synthesizes ω3 polyunsaturated fatty acids (ω3-PUFA), are protected from Purkinje cell degeneration in streptozotocin (STZ)-treated model with fat-1 mice. STZ-treated fat-1 mice did not develop hyperglycemia, motor deficits, or Purkinje cell loss. The expression of LC3 I, II, Beclin-1 and p62 were increased in the cerebellum of STZ-treated wild-type mice, and these expressions were more increased in STZ-treated fat-1 mice, but not of p62. Moreover, cerebellar Rab7, Cathepsin D, and ATP6E were increased in STZ-treated fat-1 mice. There was also increased BDNF expression in Purkinje cells without any changes in TrkB, and phosphorylation of Akt and CREB in the cerebellums of fat-1 mice. Collectively, these findings indicate that STZ-treated fat-1 mice were protected from Purkinje cell loss and exhibited increased BDNF signaling, enhancing autophagic flux activity in cerebellar Purkinje neurons. These processes may underlie Purkinje cell survival and may be potential therapeutic targets for treatment of motor deficits related to diabetic neuropathy.
Collapse
Affiliation(s)
- Dong Ho Bak
- Department of Anatomy, College of Medicine, Konyang University of Korea, Daejeon, South Korea
| | - Enji Zhang
- Department of Anatomy, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, South Korea.,Department of Anesthesiology, Yanbian University Hospital, Yanbian, 133000, China
| | - Min-Hee Yi
- Department of Anatomy, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Do-Kyung Kim
- Department of Pharmacology, College of Medicine, Konyang University of Korea, Daejeon, South Korea
| | - Kyu Lim
- Department of Biochemistry, Infection Signaling Network Research Center, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Jwa-Jin Kim
- Department of Anatomy, College of Medicine, Konyang University of Korea, Daejeon, South Korea
| | - Dong Woon Kim
- Department of Anatomy, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, South Korea
| |
Collapse
|
21
|
TrkB-Mediated Neuroprotective and Antihypoxic Properties of Brain-Derived Neurotrophic Factor. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:453901. [PMID: 26075035 PMCID: PMC4444591 DOI: 10.1155/2015/453901] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 04/15/2015] [Accepted: 04/15/2015] [Indexed: 11/18/2022]
Abstract
The neuroprotective and antihypoxic effects of brain-derived neurotrophic factor (BDNF) on dissociated hippocampal cultures in a hypoxia model were investigated. These experiments demonstrate that 10 minutes of normobaric hypoxia increased the number of dead cells in primary culture, whereas a preventive application of BDNF increased the number of viable cells. Spontaneous bioelectrical and calcium activity in neural networks was analyzed using multielectrode arrays and functional intravital calcium imaging. The results indicate that BDNF affects the functional parameters of neuronal networks in dissociated hippocampal cultures over the 7-day posthypoxic period. In addition, the effects of k252a, an antagonist of tropomyosin-related kinase B (TrkB), on functional bioelectrical activity during and after acute hypoxia were investigated. It was shown that the protective effects of BDNF are associated with binding to the TrkB receptor. Finally, intravital fluorescent mRNA probes were used to study the role of NF-κB1 in the protective effects of BDNF. Our experiments revealed that BDNF application stimulates NF-κB1 mRNA synthesis in primary dissociated hippocampal cells under normal conditions but not in hypoxic state.
Collapse
|
22
|
Li J, Wang B, Wu H, Yu Y, Xue G, Hou Y. 17β-estradiol attenuates ketamine-induced neuroapoptosis and persistent cognitive deficits in the developing brain. Brain Res 2014; 1593:30-9. [PMID: 25234726 DOI: 10.1016/j.brainres.2014.09.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 09/03/2014] [Accepted: 09/04/2014] [Indexed: 02/07/2023]
Abstract
Previous studies have demonstrated that the commonly used anesthetic ketamine can induce widespread neuroapoptosis in the neonatal brain and can cause persistent cognitive impairments as the animal matures. Therefore, searching for adjunctive neuroprotective strategies that inhibit ketamine-induced neuroapoptosis and persistent cognitive impairments is highly warranted. The primary goal of this study was to investigate the protective effect of 17β-estradiol against ketamine-induced neuroapoptosis and persistent cognitive impairments in adult rats. Starting from postnatal day 7, Sprague-Dawley male rat pups were given a daily administration of ketamine (75mg/kg, i.p.) or 17β-estradiol (600μg/kg, s.c.) in combination with ketamine (75mg/kg, i.p.). The animals were treated for three consecutive days. 24h after the last injection, the rats were decapitated, and the prefrontal cortex (PFC) was isolated to detect neuroapoptosis by cleaved caspase-3 immunohistochemistry and by using the TUNEL assay. The neuroactive steroid 17β-estradiol was quantified using high-performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS). The protein levels of BDNF and pAkt were measured by western blot analysis. At two months of age (60 days), the learning and memory abilities were tested using the Morris water maze. The results showed that ketamine triggered significant neuroapoptosis in the neonatal PFC accompanied by the downregulation of 17β-estradiol, BDNF and pAkt. The co-administration of 17β-estradiol with ketamine attenuated these changes. Moreover, 17β-estradiol significantly reversed the learning and memory deficits observed at 60 days of age. In brief, our present data demonstrate that 17β-estradiol attenuates ketamine-induced neuroapoptosis and reverses long-term cognitive deficits in developing rats and thus may be a potential therapeutic and neuroprotective method for the treatment of neurodevelopmental disorders. This article is part of a Special Issue entitled SI: Brain and Memory.
Collapse
Affiliation(s)
- Jianli Li
- Department of Anesthesiology, Hebei General Hospital, Shijiazhuang, Hebei province 050051, China
| | - Bei Wang
- Department of Gynecology, Hebei General Hospital, Shijiazhuang, Hebei province 050051, China
| | - Honghai Wu
- Department of Pharmacy, Bethune International Peace Hospital of Chinese PLA, Shijiazhuang, Hebei province 050082, China
| | - Yang Yu
- Department of Pharmacy, Bethune International Peace Hospital of Chinese PLA, Shijiazhuang, Hebei province 050082, China
| | - Gai Xue
- Department of Pharmacy, Bethune International Peace Hospital of Chinese PLA, Shijiazhuang, Hebei province 050082, China
| | - Yanning Hou
- Department of Pharmacy, Bethune International Peace Hospital of Chinese PLA, Shijiazhuang, Hebei province 050082, China.
| |
Collapse
|
23
|
Qi D, Ouyang C, Wang Y, Zhang S, Ma X, Song Y, Yu H, Tang J, Fu W, Sheng L, Yang L, Wang M, Zhang W, Miao L, Li T, Huang X, Dong H. HO-1 attenuates hippocampal neurons injury via the activation of BDNF–TrkB–PI3K/Akt signaling pathway in stroke. Brain Res 2014; 1577:69-76. [DOI: 10.1016/j.brainres.2014.06.031] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 06/24/2014] [Accepted: 06/25/2014] [Indexed: 01/07/2023]
|
24
|
Cheng CY, Lin JG, Su SY, Tang NY, Kao ST, Hsieh CL. Electroacupuncture-like stimulation at Baihui and Dazhui acupoints exerts neuroprotective effects through activation of the brain-derived neurotrophic factor-mediated MEK1/2/ERK1/2/p90RSK/bad signaling pathway in mild transient focal cerebral ischemia in rats. Altern Ther Health Med 2014; 14:92. [PMID: 24606810 PMCID: PMC3975570 DOI: 10.1186/1472-6882-14-92] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2013] [Accepted: 03/03/2014] [Indexed: 01/12/2023]
Abstract
Background This study was designed to evaluate the effects of electroacupuncture-like stimulation at Baihui (GV20) and Dazhui (GV14) acupoints (EA at acupoints) following mild cerebral ischemia-reperfusion (I/R) injury. Furthermore, we investigated whether brain-derived neurotrophic factor (BDNF)-mediated activation of extracellular signal-regulated kinase (ERK)1/2 signaling pathway is involved in the neuroprotection induced by EA at acupoints. Methods Rats were subjected to middle cerebral artery occlusion (MCAo) for 15 min followed by reperfusion for 3 d. EA at acupoints was applied 1 d postreperfusion then once daily for 2 consecutive days. Results Following the application of EA at acupoints, initiated 1 d postreperfusion, we observed significant reductions in the cerebral infarct area, neurological deficit scores, active caspase-3 protein expression, and apoptosis in the ischemic cortex after 3 d of reperfusion. We also observed markedly upregulated BDNF, phospho-Raf-1 (pRaf-1), phospho-MEK1/2 (pMEK1/2), phospho-ERK1/2 (pERK1/2), phospho-90 kDa ribosomal S6 kinase (pp90RSK), and phospho-Bad (pBad) expression, and restored neuronal nuclear antigen (NeuN) expression. Pretreatment with the MEK1/2 inhibitor U0126 abrogated the effects of EA at acupoints on cerebral infarct size, neurological deficits, active caspase-3 protein, and apoptosis in the ischemic cortex after 3 d of reperfusion. Pretreatment with U0126 also abrogated the effects of EA at acupoints on pMEK1/2, pERK1/2, pp90RSK, pBad, and NeuN expression, but did not influence BDNF and pRaf-1 expression. Conclusion Overall, our study results indicated that EA at acupoints, initiated 1 d postreperfusion, upregulates BDNF expression to provide BDNF-mediated neuroprotection against caspase-3-dependent neuronal apoptosis through activation of the Raf-1/MEK1/2/ERK1/2/p90RSK/Bad signaling cascade after 3 d of reperfusion in mild MCAo.
Collapse
|
25
|
Environmental enrichment decreases asphyxia-induced neurobehavioral developmental delay in neonatal rats. Int J Mol Sci 2013; 14:22258-73. [PMID: 24232451 PMCID: PMC3856064 DOI: 10.3390/ijms141122258] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Revised: 10/18/2013] [Accepted: 10/28/2013] [Indexed: 01/01/2023] Open
Abstract
Perinatal asphyxia during delivery produces long-term disability and represents a major problem in neonatal and pediatric care. Numerous neuroprotective approaches have been described to decrease the effects of perinatal asphyxia. Enriched environment is a popular strategy to counteract nervous system injuries. The aim of the present study was to investigate whether enriched environment is able to decrease the asphyxia-induced neurobehavioral developmental delay in neonatal rats. Asphyxia was induced in ready-to-deliver mothers by removing the pups by caesarian section after 15 min of asphyxia. Somatic and neurobehavioral development was tested daily and motor coordination weekly. Our results show that rats undergoing perinatal asphyxia had a marked developmental delay and worse performance in motor coordination tests. However, pups kept in enriched environment showed a decrease in the developmental delay observed in control asphyctic pups. Rats growing up in enriched environment did not show decrease in weight gain after the first week and the delay in reflex appearance was not as marked as in control rats. In addition, the development of motor coordination was not as strikingly delayed as in the control group. Short-term neurofunctional outcome are known to correlate with long-term deficits. Our results thus show that enriched environment could be a powerful strategy to decrease the deleterious developmental effects of perinatal asphyxia.
Collapse
|
26
|
Chen A, Xiong LJ, Tong Y, Mao M. Neuroprotective effect of brain-derived neurotrophic factor mediated by autophagy through the PI3K/Akt/mTOR pathway. Mol Med Rep 2013; 8:1011-6. [PMID: 23942837 DOI: 10.3892/mmr.2013.1628] [Citation(s) in RCA: 138] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2013] [Accepted: 07/03/2013] [Indexed: 11/05/2022] Open
Abstract
Brain‑derived neurotrophic factor (BDNF) has been demonstrated to be a potent growth factor that is beneficial in neuronal functions following hypoxia‑ischemia (HI). Mature BDNF triggers three enzymes, mitogen‑activated protein kinase (MAPK), phosphatidylinositol 3‑kinase (PI3K) and phosphoinositide phospholipase C-γ (PLCγ), which are its predominant downstream regulators. The PI3K‑Akt signaling pathway is upstream of the mammalian target of rapamycin (mTOR), which is important in the induction of autophagy. However, whether the neuroprotective effect of BDNF is mediated by autophagy through the PI3K/Akt/mTOR pathway remains to be elucidated. Cortical neurons were cultured following isolation from pregnant rats (gestational days 16‑18). The induction of autophagy following BDNF treatment was analyzed by microtubule‑associated protein light chain 3 (LC3) conversion and autophagosome formation. The phosphorylation of Akt, mTOR and ribosomal protein S6 kinase (p70S6K) was analyzed in cultured cells with or without BDNF treatment. Cell viability was determined by a Cell Counting Kit‑8 for estimating the protective effect of BDNF. Results demonstrated that autophagy was induced in cells with oxygen deprivation. BDNF promoted cell viability via the upregulation of autophagy. Moreover, LC3 upregulation was related to Akt/mTOR/p70S6K inhibition by BDNF. In conclusion, the results suggested that the neuroprotective effect of BDNF was mediated by autophagy through the PI3K/Akt/mTOR pathway.
Collapse
Affiliation(s)
- Ai Chen
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | | | | | | |
Collapse
|
27
|
Tian X, Hua F, Sandhu HK, Chao D, Balboni G, Salvadori S, He X, Xia Y. Effect of δ-opioid receptor activation on BDNF-TrkB vs. TNF-α in the mouse cortex exposed to prolonged hypoxia. Int J Mol Sci 2013; 14:15959-76. [PMID: 23912236 PMCID: PMC3759895 DOI: 10.3390/ijms140815959] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 07/25/2013] [Accepted: 07/25/2013] [Indexed: 12/15/2022] Open
Abstract
We investigated whether δ-opioid receptor (DOR)-induced neuroprotection involves the brain-derived neurotrophic factor (BDNF) pathway. We studied the effect of DOR activation on the expression of BDNF and other proteins in the cortex of C57BL/6 mice exposed to hypoxia (10% of oxygen) for 1–10 days. The results showed that: (1) 1-day hypoxia had no appreciable effect on BDNF expression, while 3- and 10-day hypoxia progressively decreased BDNF expression, resulting in 37.3% reduction (p < 0.05) after 10-day exposure; (2) DOR activation with UFP-512 (1 mg/kg, i.p., daily) partially reversed the hypoxia-induced reduction of BDNF expression in the 3- or 10-day exposed cortex; (3) DOR activation partially reversed the hypoxia-induced reduction in functional TrkB (140-kDa) and attenuated hypoxia-induced increase in truncated TrkB (90-kDa) in the 3- or 10-day hypoxic cortex; and (4) prolonged hypoxia (10 days) significantly increased TNF-α level and decreased CD11b expression in the cortex, which was completely reversed following DOR activation; and (5) there was no significant change in pCREB and pATF-1 levels in the hypoxic cortex. We conclude that prolonged hypoxia down-regulates BDNF-TrkB signaling leading to an increase in TNF-α in the cortex, while DOR activation up-regulates BDNF-TrkB signaling thereby decreasing TNF-α levels in the hypoxic cortex.
Collapse
Affiliation(s)
- Xuesong Tian
- The Vivan L. Smith Department of Neurosurgery, The University of Texas Medical School at Houston, Houston, TX 77030, USA; E-Mails: (X.T.); (H.K.S.); (D.C.)
| | - Fei Hua
- Research Institute of Modern Medicine, The Third Medical College of Soochow University, Changzhou, Jiangsu 213002, China; E-Mail:
| | - Harleen K Sandhu
- The Vivan L. Smith Department of Neurosurgery, The University of Texas Medical School at Houston, Houston, TX 77030, USA; E-Mails: (X.T.); (H.K.S.); (D.C.)
| | - Dongman Chao
- The Vivan L. Smith Department of Neurosurgery, The University of Texas Medical School at Houston, Houston, TX 77030, USA; E-Mails: (X.T.); (H.K.S.); (D.C.)
| | - Gianfranco Balboni
- Department of Life and Environment Sciences, University of Cagliari, Cagliari I-09124, Italy; E-Mail:
| | - Severo Salvadori
- Department of Pharmaceutical Sciences, University of Ferrara, Ferrara I-44100, Italy; E-Mail:
| | - Xiaozhou He
- Research Institute of Modern Medicine, The Third Medical College of Soochow University, Changzhou, Jiangsu 213002, China; E-Mail:
- Authors to whom correspondence should be addressed; E-Mails: (X.H.); (Y.X.); Tel./Fax: +1-713-500-6288 (Y.X.)
| | - Ying Xia
- The Vivan L. Smith Department of Neurosurgery, The University of Texas Medical School at Houston, Houston, TX 77030, USA; E-Mails: (X.T.); (H.K.S.); (D.C.)
- Authors to whom correspondence should be addressed; E-Mails: (X.H.); (Y.X.); Tel./Fax: +1-713-500-6288 (Y.X.)
| |
Collapse
|
28
|
Guo S, Som AT, Waeber C, Lo EH. Vascular neuroprotection via TrkB- and Akt-dependent cell survival signaling. J Neurochem 2012; 123 Suppl 2:58-64. [PMID: 23050643 DOI: 10.1111/j.1471-4159.2012.07944.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The cerebral endothelium can be a vital source of signaling factors such as brain-derived neurotrophic factor that defends the neuronal parenchyma against stress and injury. But the underlying mechanisms remain to be fully defined. Here, we use cell models to ask how vascular neuroprotection is sustained. Human brain endothelial cells were grown in culture, and conditioned media were transferred to primary rat cortical neurons. Brain endothelial cell-conditioned media activated neuronal Akt signaling and protected neurons against hypoxia and oxygen-glucose deprivation. Blockade of Akt phosphorylation with the PI3-kinase inhibitor LY294002 negated this vascular neuroprotective effect. Upstream of Akt signaling, the brain-derived neurotrophic factor receptor TrkB (neurotrophic tyrosine kinase receptor, type 2) was involved because depletion with TrkB/Fc eliminated the ability of endothelial-conditioned media to protect neurons against hypoxia. Downstream of Akt signaling, activation of GSK-3β (glycogen synthase kinase 3 beta), caspase 9, caspase 3 and Bad pathways were detected. Taken together, these findings suggest that the molecular basis for vascular neuroprotection involves TrkB-Akt signaling that ameliorates neuronal apoptosis. Further investigation of these mechanisms may reveal new approaches for augmenting endogenous vascular neuroprotection in stroke, brain injury, and neurodegeneration.
Collapse
Affiliation(s)
- Shuzhen Guo
- Neuroprotection Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA.
| | | | | | | |
Collapse
|
29
|
Chen A, Xiong LJ, Tong Y, Mao M. The neuroprotective roles of BDNF in hypoxic ischemic brain injury. Biomed Rep 2012; 1:167-176. [PMID: 24648914 DOI: 10.3892/br.2012.48] [Citation(s) in RCA: 195] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2012] [Accepted: 10/16/2012] [Indexed: 12/19/2022] Open
Abstract
Hypoxia-ischemia (H/I) brain injury results in various degrees of damage to the body, and the immature brain is particularly fragile to oxygen deprivation. Hypothermia and erythropoietin (EPO) have long been known to be neuroprotective in ischemic brain injury. Brain-derived neurotrophic factor (BDNF) has recently been recognized as a potent modulator capable of regulating a wide repertoire of neuronal functions. This review was based on studies concerning the involvement of BDNF in the protection of H/I brain injury following a search in PubMed between 1995 and December, 2011. We initially examined the background of BDNF, and then focused on its neuroprotective mechanisms against ischemic brain injury, including its involvement in promoting neural regeneration/cognition/memory rehabilitation, angiogenesis within ischemic penumbra and the inhibition of the inflammatory process, neurotoxicity, epilepsy and apoptosis. We also provided a literature overview of experimental studies, discussing the safety and the potential clinical application of BDNF as a neuroprotective agent in the ischemic brain injury.
Collapse
Affiliation(s)
- Ai Chen
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Li-Jing Xiong
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yu Tong
- Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Chengdu, Sichuan 610041, P.R. China ; ; Laboratory of Early Developmental and Injuries, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Meng Mao
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
30
|
Ke X, McKnight RA, Caprau D, O'Grady S, Fu Q, Yu X, Callaway CW, Albertine KH, Lane RH. Intrauterine growth restriction affects hippocampal dual specificity phosphatase 5 gene expression and epigenetic characteristics. Physiol Genomics 2011; 43:1160-9. [PMID: 21828247 DOI: 10.1152/physiolgenomics.00242.2010] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Intrauterine growth retardation (IUGR) predisposes humans toward hippocampal morbidities, such as impaired learning and memory. Hippocampal dual specificity phosphatase 5 (DUSP5) may be involved in these morbidities because DUSP5 regulates extracellular signal-regulated kinase phosphorylation (Erk). In the rat, IUGR causes postnatal changes in hippocampal gene expression and epigenetic characteristics. However, the impact of IUGR upon hippocampal DUSP5 expression and epigenetic characteristics is not known. We therefore hypothesized that IUGR affects hippocampal 1) DUSP5 expression, DNA CpG methylation, and histone code, and 2) erk1/2 phosphorylation in a well-characterized rat model of IUGR. We found that IUGR significantly decreased DUSP5 expression in the day of life (DOL) 0 and 21 male rat, while decreasing only DUSP5 protein levels in the DOL21 female rat. Fluorescent in situ hybridization and immunohistochemistry analyses localized the changes in DUSP5 mRNA and protein, many of which occurred in the dentate gyrus. IUGR also caused sex-specific differences in DNA CpG methylation and histone code in two sites of the hippocampal DUSP5 gene, a 5'-flanking specificity protein-1 (SP1) site and exon 2. Finally, when IUGR decreased DUSP5 protein levels, Erk phosphorylation increased. We conclude that IUGR affects hippocampal DUSP5 expression and epigenetic characteristics in a sex-specific manner.
Collapse
Affiliation(s)
- Xingrao Ke
- University of Utah School of Medicine, Department of Pediatrics, Division of Neonatology, Salt Lake City, Utah 84132-2202, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Neuroprotective phenolics in medicinal plants. Arch Pharm Res 2010; 33:1611-32. [DOI: 10.1007/s12272-010-1011-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2010] [Revised: 09/02/2010] [Accepted: 09/02/2010] [Indexed: 12/26/2022]
|
32
|
Kemp K, Hares K, Mallam E, Heesom KJ, Scolding N, Wilkins A. Mesenchymal stem cell-secreted superoxide dismutase promotes cerebellar neuronal survival. J Neurochem 2010; 114:1569-80. [PMID: 20028455 DOI: 10.1111/j.1471-4159.2009.06553.x] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
It has been postulated that bone marrow-derived mesenchymal stem cells (MSCs) might be effective treatments for neurodegenerative disorders either by replacement of lost cells by differentiation into functional neural tissue; modulation of the immune system to prevent further neurodegeneration; and/or provision of trophic support for the diseased nervous system. Here we have performed a series of experiments showing that human bone marrow-derived MSCs are able to protect cultured rodent cerebellar neurons, and specifically cells expressing Purkinje cell markers, against either nitric oxide exposure or withdrawal of trophic support via cell-cell contact and/or secretion of soluble factors, or through secretion of soluble factors alone. We have demonstrated that MSCs protect cerebellar neurons against toxic insults via modulation of both the phosphatidylinositol 3-kinase/Akt and MAPK pathways and defined superoxide dismutase 3 as a secreted active antioxidant biomolecule by which MSCs modulate, at least in part, their neuroprotective effect on cerebellar cells in vitro. Together, the results demonstrate new and specific mechanisms by which MSCs promote cerebellar neuronal survival and add further evidence to the concept that MSCs may be potential therapeutic agents for neurological disorders involving the cerebellum.
Collapse
Affiliation(s)
- Kevin Kemp
- Multiple Sclerosis and Stem Cell Group, Institute of Clinical Neurosciences, Clinical Sciences North Bristol, University of Bristol, Bristol, UK.
| | | | | | | | | | | |
Collapse
|
33
|
Alcalá-Barraza SR, Lee MS, Hanson LR, McDonald AA, Frey WH, McLoon LK. Intranasal delivery of neurotrophic factors BDNF, CNTF, EPO, and NT-4 to the CNS. J Drug Target 2010; 18:179-90. [PMID: 19807216 DOI: 10.3109/10611860903318134] [Citation(s) in RCA: 130] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Injury to the central nervous system (CNS) generally results in significant neuronal death and functional loss. In vitro experiments have demonstrated that neurotrophic factors such as brain-derived neurotrophic factor (BDNF), ciliary neurotrophic factor (CNTF), and neurotrophin-4/5 (NT-4/5) can promote neuronal survival. However, delivery to the injured CNS is difficult as these large protein molecules do not efficiently cross the blood-brain barrier. Intranasal delivery of 70 microg [(125)I]-radiolabeled BDNF, CNTF, NT-4, or erythropoietin (EPO) resulted in 0.1-1.0 nM neurotrophin concentrations within 25 min in brain parenchyma. In addition, not only did these neurotrophic factors reach the CNS, they were present in sufficient concentrations to activate the prosurvival PI3Kinase/Akt pathway, even where lower levels of neurotrophic factors were measured. Currently traumatic, ischemic and compressive injuries to the CNS have no effective treatment. There is potential clinical relevancy of this method for rescuing injured CNS tissues in order to maintain CNS function in affected patients. The intranasal delivery method has great clinical potential due to (1) simplicity of administration, (2) noninvasive drug administration, (3) relatively rapid CNS delivery, (4) ability to repeat dosing easily, (5) no requirement for drug modification, and (6) minimal systemic exposure.
Collapse
|
34
|
Duman RS. Neuronal damage and protection in the pathophysiology and treatment of psychiatric illness: stress and depression. DIALOGUES IN CLINICAL NEUROSCIENCE 2009. [PMID: 19877493 PMCID: PMC3181922 DOI: 10.31887/dcns.2009.11.3/rsduman] [Citation(s) in RCA: 143] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The discovery that stress and depression, as well as other psychiatric illnesses, are characterized by structural alterations, and that these changes result from atrophy and loss of neurons and glia in specific limbic regions and circuits, has contributed to a fundamental change in our understanding of these illnesses. These structural changes are accompanied by dysregulation of neuroprotective and neurotrophic signaling mechanisms that are required for the maturation, growth, and survival of neurons and glia. Conversely, behavioral and therapeutic interventions can reverse these structural alterations by stimulating neuroprotective and neurotrophic pathways and by blocking the damaging, excitotoxic, and inflammatory effects of stress. Lifetime exposure to cellular and environmental stressors and interactions with genetic factors contribute to individual susceptibility or resilience. This exciting area of research holds promise and potential for further elucidating the pathophysiology of psychiatric illness and for development of novel therapeutic interventions.
Collapse
Affiliation(s)
- Ronald S Duman
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06508, USA.
| |
Collapse
|
35
|
Xia Y, Wang CZ, Liu J, Anastasio NC, Johnson KM. Brain-derived neurotrophic factor prevents phencyclidine-induced apoptosis in developing brain by parallel activation of both the ERK and PI-3K/Akt pathways. Neuropharmacology 2009; 58:330-6. [PMID: 19887077 DOI: 10.1016/j.neuropharm.2009.10.009] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2009] [Revised: 09/25/2009] [Accepted: 10/27/2009] [Indexed: 12/01/2022]
Abstract
Phencyclidine is an N-methyl d-aspartate receptor (NMDAR) blocker that has been reported to induce neuronal apoptosis during development and schizophrenia-like behaviors in rats later in life. Brain-derived neurotrophic factor (BDNF) has been shown to prevent neuronal death caused by NMDAR blockade, but the precise mechanism is unknown. This study examined the role of the phosphatidylinositol-3 kinase (PI3K)/Akt and extracellular signal-regulated kinase (ERK) pathways in BDNF protection of PCP-induced apoptosis in corticostriatal organotypic cultures. It was observed that BDNF inhibited PCP-induced apoptosis in a concentration-dependent fashion. BDNF effectively prevented PCP-induced inhibition of the ERK and PI-3K/Akt pathways and suppressed GSK-3beta activation. Blockade of either PI-3K/Akt or ERK activation abolished BDNF protection. Western blot analysis revealed that the PI-3K inhibitor LY294002 prevented the stimulating effect of BDNF on the PI-3K/Akt pathway, but had no effect on the ERK pathway. Similarly, the ERK inhibitor PD98059 prevented the stimulating effect of BDNF on the ERK pathway, but not the PI-3K/Akt pathway. Co-application of LY294002 and PD98059 had no additional effect on BDNF-evoked activation of Akt or ERK. However, concurrent exposure to PD98059 and LY294002 caused much greater inhibition of BDNF-evoked phosphorylation of GSK-3beta at serine 9 than did LY294002 alone. Finally, either BDNF or GSK-3beta inhibition prevented PCP-induced suppression of cyclic-AMP response element binding protein (CREB) phosphorylation. These data demonstrate that the protective effect of BDNF against PCP-induced apoptosis is mediated by parallel activation of the PI-3K/Akt and ERK pathways, most likely involves inhibition of GSK-3beta and activation of CREB.
Collapse
Affiliation(s)
- Yan Xia
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555-1031, USA
| | | | | | | | | |
Collapse
|