51
|
Hwang S, Ham S, Lee SE, Lee Y, Lee GH. Hypoxia regulates the level of glutamic acid decarboxylase enzymes and interrupts inhibitory synapse stability in primary cultured neurons. Neurotoxicology 2018; 65:221-230. [DOI: 10.1016/j.neuro.2017.10.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 09/19/2017] [Accepted: 10/27/2017] [Indexed: 01/23/2023]
|
52
|
Gu F, Parada I, Yang T, Longo FM, Prince DA. Partial TrkB receptor activation suppresses cortical epileptogenesis through actions on parvalbumin interneurons. Neurobiol Dis 2018; 113:45-58. [PMID: 29408225 DOI: 10.1016/j.nbd.2018.01.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 01/21/2018] [Accepted: 01/24/2018] [Indexed: 01/17/2023] Open
Abstract
Post-traumatic epilepsy is one of the most common and difficult to treat forms of acquired epilepsy worldwide. Currently, there is no effective way to prevent post-traumatic epileptogenesis. It is known that abnormalities of interneurons, particularly parvalbumin-containing interneurons, play a critical role in epileptogenesis following traumatic brain injury. Thus, enhancing the function of existing parvalbumin interneurons might provide a logical therapeutic approach to prevention of post-traumatic epilepsy. The known positive effects of brain-derived neurotrophic factor on interneuronal growth and function through activation of its receptor tropomyosin receptor kinase B, and its decrease after traumatic brain injury, led us to hypothesize that enhancing trophic support might improve parvalbumin interneuronal function and decrease epileptogenesis. To test this hypothesis, we used the partial neocortical isolation ('undercut', UC) model of posttraumatic epileptogenesis in mature rats that were treated for 2 weeks, beginning on the day of injury, with LM22A-4, a newly designed partial agonist at the tropomyosin receptor kinase B. Effects of treatment were assessed with Western blots to measure pAKT/AKT; immunocytochemistry and whole cell patch clamp recordings to examine functional and structural properties of GABAergic interneurons; field potential recordings of epileptiform discharges in vitro; and video-EEG recordings of PTZ-induced seizures in vivo. Results showed that LM22A-4 treatment 1) increased pyramidal cell perisomatic immunoreactivity for VGAT, GAD65 and parvalbumin; 2) increased the density of close appositions of VGAT/gephyrin immunoreactive puncta (putative inhibitory synapses) on pyramidal cell somata; 3) increased the frequency of mIPSCs in pyramidal cells; and 4) decreased the incidence of spontaneous and evoked epileptiform discharges in vitro. 5) Treatment of rats with PTX BD4-3, another partial TrkB receptor agonist, reduced the incidence of bicuculline-induced ictal episodes in vitro and PTZ induced electrographic and behavioral ictal episodes in vivo. 6) Inactivation of TrkB receptors in undercut TrkBF616A mice with 1NMPP1 abolished both LM22A-4-induced effects on mIPSCs and on increased perisomatic VGAT-IR. Results indicate that chronic activation of the tropomyosin receptor kinase B by a partial agonist after cortical injury can enhance structural and functional measures of GABAergic inhibition and suppress posttraumatic epileptogenesis. Although the full agonist effects of brain-derived neurotrophic factor and tropomyosin receptor kinase B activation in epilepsy models have been controversial, the present results indicate that such trophic activation by a partial agonist may potentially serve as an effective therapeutic option for prophylactic treatment of posttraumatic epileptogenesis, and treatment of other neurological and psychiatric disorders whose pathogenesis involves impaired parvalbumin interneuronal function.
Collapse
Affiliation(s)
- Feng Gu
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, United States
| | - Isabel Parada
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, United States
| | - Tao Yang
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, United States
| | - Frank M Longo
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, United States
| | - David A Prince
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, United States.
| |
Collapse
|
53
|
Han JY, Kim JK, Kim JH, Oh BS, Cho WJ, Jung YD, Lee SG. Neurorestorative effects of epigallocatechin-3-Gallate on cognitive function in a chronic cerebral hypoperfusion rat model. Restor Neurol Neurosci 2018; 34:367-77. [PMID: 27080069 DOI: 10.3233/rnn-150586] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
PURPOSE This study investigated whether (-)-epigallocatechin-3-gallate (EGCG) can enhance cognition by a neurorestorative effect in a rat model of bilateral common carotid artery occlusion (BCCAO). METHODS Forty-eight male, 8-week-old Sprague-Dawley rats were randomly allocated to four groups 6 weeks after BCCAO or sham operation: EGCG-single intravenous injection (25 mg/kg/day; SIV group), EGCG-multiple intraperitoneal injection (50 mg/kg/day for 5 days; MIP group), untreated BCCAO group (untreated group), and sham-operated group (sham group). RESULTS Escape latency was significantly shorter in the SIV and MIP groups than in the untreated group. SIV and MIP groups were significantly different from the untreated group in the activity of superoxide dismutase and the content of malondialdehyde (p < 0.05). Protein expression level of brain-derived neurotrophic factor was not significantly different between groups (p > 0.05), while protein expression of vascular endothelial growth factor was significantly lower in the SIV group than in the untreated group (p < 0.05). Protein expression of N-methyl-D-aspartate receptor subunits NR1 and NR2B was significantly higher in the MIP group than in the untreated group (p < 0.05). CONCLUSIONS EGCG administration at 6 weeks after BCCAO is neurorestorative via an anti-oxidant effect and synaptogenesis, except for angiogenesis.
Collapse
Affiliation(s)
- Jae-Young Han
- Department of Physical & Rehabilitation Medicine, Chonnam National University Medical School & Hospital, Gwangju City, Republic of Korea
| | - Jung-Kook Kim
- Department of Rehabilitation Standard & Policy, National Rehabilitation Center Research Institute, Seoul City, Republic of Korea
| | - Jae-Hong Kim
- Department of Acupuncture & Moxibustion Medicine, College of Korean Medicine, Dong-Shin University, Naju City, Republic of Korea
| | - Bong-Seok Oh
- Department of Sports and Leisure Science, Sunchon National University, Sunchon City, Republic of Korea
| | - Wan-Ju Cho
- Department of Physical Education, Chosun University, Gwangju City, Republic of Korea
| | - Young Do Jung
- Department of Biochemistry, Chonnam National University Medical School, Gwangju City, Republic of Korea
| | - Sam-Gyu Lee
- Department of Physical & Rehabilitation Medicine, Chonnam National University Medical School & Hospital, Gwangju City, Republic of Korea
| |
Collapse
|
54
|
Abstract
K+-Cl- co-transporter 2 (KCC2/SLC12A5) is a neuronal specific cation chloride co-transporter which is active under isotonic conditions, and thus a key regulator of intracellular Cl- levels. It also has an ion transporter-independent structural role in modulating the maturation and regulation of excitatory glutamatergic synapses. KCC2 levels are developmentally regulated, and a postnatal upregulation of KCC2 generates a low intracellular chloride concentration that allows the neurotransmitters γ-aminobutyric acid (GABA) and glycine to exert inhibitory neurotransmission through its Cl- permeating channel. Functional expression of KCC2 at the neuronal cell surface is necessary for its activity, and impairment in KCC2 cell surface transport and/or internalization may underlie a range of neuropathological conditions. Although recent advances have shed light on a range of cellular mechanisms regulating KCC2 activity, little is known about its membrane trafficking itinerary and regulatory proteins. In this review, known membrane trafficking signals, pathways and mechanisms pertaining to KCC2's functional surface expression are discussed.
Collapse
Affiliation(s)
- Bor Luen Tang
- a Department of Biochemistry, Yong Loo Lin School of Medicine , National University Health System , Singapore.,b NUS Graduate School for Integrative Sciences and Engineering , National University of Singapore , Singapore
| |
Collapse
|
55
|
Jean-Xavier C, Sharples SA, Mayr KA, Lognon AP, Whelan PJ. Retracing your footsteps: developmental insights to spinal network plasticity following injury. J Neurophysiol 2017; 119:521-536. [PMID: 29070632 DOI: 10.1152/jn.00575.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
During development of the spinal cord, a precise interaction occurs between descending projections and sensory afferents, with spinal networks that lead to expression of coordinated motor output. In the rodent, during the last embryonic week, motor output first occurs as regular bursts of spontaneous activity, progressing to stochastic patterns of episodes that express bouts of coordinated rhythmic activity perinatally. Locomotor activity becomes functionally mature in the 2nd postnatal wk and is heralded by the onset of weight-bearing locomotion on the 8th and 9th postnatal day. Concomitantly, there is a maturation of intrinsic properties and key conductances mediating plateau potentials. In this review, we discuss spinal neuronal excitability, descending modulation, and afferent modulation in the developing rodent spinal cord. In the adult, plastic mechanisms are much more constrained but become more permissive following neurotrauma, such as spinal cord injury. We discuss parallel mechanisms that contribute to maturation of network function during development to mechanisms of pathological plasticity that contribute to aberrant motor patterns, such as spasticity and clonus, which emerge following central injury.
Collapse
Affiliation(s)
- C Jean-Xavier
- Hotchkiss Brain Institute, University of Calgary , Calgary, Alberta , Canada.,Department of Comparative Biology and Experimental Medicine, University of Calgary , Calgary, Alberta , Canada
| | - S A Sharples
- Hotchkiss Brain Institute, University of Calgary , Calgary, Alberta , Canada.,Department of Neuroscience, University of Calgary , Calgary, Alberta , Canada
| | - K A Mayr
- Hotchkiss Brain Institute, University of Calgary , Calgary, Alberta , Canada.,Department of Neuroscience, University of Calgary , Calgary, Alberta , Canada
| | - A P Lognon
- Department of Comparative Biology and Experimental Medicine, University of Calgary , Calgary, Alberta , Canada
| | - P J Whelan
- Hotchkiss Brain Institute, University of Calgary , Calgary, Alberta , Canada.,Department of Comparative Biology and Experimental Medicine, University of Calgary , Calgary, Alberta , Canada
| |
Collapse
|
56
|
Yang J, Ju L, Jia M, Zhang H, Sun X, Ji M, Yang J, Martynyuk AE. Subsequent maternal separation exacerbates neurobehavioral abnormalities in rats neonatally exposed to sevoflurane anesthesia. Neurosci Lett 2017; 661:137-142. [PMID: 28982596 DOI: 10.1016/j.neulet.2017.09.063] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 09/24/2017] [Accepted: 09/29/2017] [Indexed: 12/16/2022]
Abstract
Several recent studies suggest that in the human population, a routine, short anesthetic in otherwise healthy infants is void of neurodevelopmental insult. On the other hand, many human retrospective epidemiological studies report evidence of cognitive abnormalities in children after testing those who had different anesthesia-requiring procedures in early childhood. We tested in a rat model whether post-anesthesia stressful environmental factors can contribute to developmental abnormalities that were initiated by a relatively short exposure to sevoflurane, the most widely used anesthetic in pediatric anesthesia, whose polyvalent actions include enhancement of gamma-aminobutyric acid type A receptor (GABAAR) activity. Postnatal day 6 (P6) male Sprague-Dawley rats were anesthetized with sevoflurane for 60min. To simulate subsequent stress, the animals were subjected to a single maternal separation for 180min at P10. To study the role of GABAAR-mediated depolarization, subgroups of P6 rats received a single injection of the Na+-K+-2Cl- (NKCC1) inhibitor, bumetanide, prior to initiation of anesthesia with sevoflurane. Rats that were exposed to sevoflurane had decreased hypothalamic K+-2Cl- (KCC2) mRNA level (F(2,13)=3.839, P=0.049), increased NKCC1/KCC2 mRNA ratio (F(2,13)=5.043, P=0.024) and increased corticotropin-releasing hormone (CRH) mRNA level (F(2,12)=9.450, P=0.003) at P10, the age at which maternal separation was imposed. Adult rats, neonatally exposed to a combination of sevoflurane and maternal separation, exhibited increases in the escape latencies greater than animals exposed to sevoflurane only (P=0.012), and only rats in the sevoflurane plus maternal separation group spent significantly less time in the target quadrant during the Morris water maze test (F(4,55)=4.856, P=0.002). Bumetanide ameliorated abnormalities induced by sevoflurane and a combination of sevoflurane plus maternal separation. Neonatal exposure to sevoflurane may sensitize to stressors later in life, and post-exposure stress may exacerbate neurodevelopmental abnormalities even after a relatively short exposure to sevoflurane in rodents. The NKCC1 downregulation prior to exposure to the anesthetic may be therapeutic.
Collapse
Affiliation(s)
- Jiaojiao Yang
- Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China; Department of Anesthesiology, University of Florida College of Medicine, Gainesville, FL, United States; Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Lingsha Ju
- Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China; Department of Anesthesiology, University of Florida College of Medicine, Gainesville, FL, United States; Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Min Jia
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Hui Zhang
- Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China; Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Xiaoru Sun
- Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China; Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Muhuo Ji
- Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China; Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Jianjun Yang
- Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China; Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China.
| | - Anatoly E Martynyuk
- Department of Anesthesiology, University of Florida College of Medicine, Gainesville, FL, United States; McKnight Brain Institute, University of FL College of Medicine, Gainesville, FL, United States.
| |
Collapse
|
57
|
Kambli L, Bhatt LK, Oza M, Prabhavalkar K. Novel therapeutic targets for epilepsy intervention. Seizure 2017; 51:27-34. [DOI: 10.1016/j.seizure.2017.07.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 07/23/2017] [Accepted: 07/24/2017] [Indexed: 12/11/2022] Open
|
58
|
Sen A, Nelson TJ, Alkon DL. ApoE isoforms differentially regulates cleavage and secretion of BDNF. Mol Brain 2017; 10:19. [PMID: 28569173 PMCID: PMC5452344 DOI: 10.1186/s13041-017-0301-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 05/25/2017] [Indexed: 01/12/2023] Open
Abstract
Apolipoprotein E4 (ApoE4) is a major genetic risk factor for sporadic or late onset Alzheimer’s disease (AD). Brain-derived neurotrophic factor (BDNF) is decreased by 3 to 4-fold in the brains of AD patients at autopsy. ApoE4 mice also have reduced BDNF levels. However, there have been no reports relating the different ApoE isoforms or AD to differential regulation of BDNF. Here we report that in the hippocampal regions of AD patients both prepro-BDNF and pro-BDNF expression showed a 40 and 60% decrease respectively compared to that expression in the hippocampi of age-matched control patients. We further report that ApoE isoforms differentially regulate maturation and secretion of BDNF from primary human astrocytes. After 24 h, ApoE3 treated astrocytes secreted 1.75- fold higher pro-BDNF than ApoE2-treated astrocytes, and ApoE2-treated astrocytes secreted 3-fold more mature-BDNF (m-BDNF) than ApoE3-treated astrocytes. In contrast, ApoE4-treated cells secreted negligible amounts of m-BDNF or pro-BDNF. ApoE2 increased the level of intracellular pre-pro BDNF by 19.04 ± 6.68%, while ApoE4 reduced the pre-pro BDNF by 21.61 ± 5.9% compared to untreated cells. Similar results were also seen in ApoE2, ApoE3 or ApoE4 treated cells at 4 h. Together, these results indicate that an ApoE2 or ApoE3 mediated positive regulation of BDNF may be protective while ApoE4 related defects in BDNF processing could lead to AD pathophysiology. These interactions of the ApoE isoforms with BDNF may help explain the increased risk of AD associated with the ApoE4 isoform.
Collapse
Affiliation(s)
- Abhik Sen
- Blanchette Rockefeller Neurosciences Institute, 8 Medical Center Drive, Morgantown, WV, 26505, USA.
| | - Thomas J Nelson
- Blanchette Rockefeller Neurosciences Institute, 8 Medical Center Drive, Morgantown, WV, 26505, USA
| | - Daniel L Alkon
- Blanchette Rockefeller Neurosciences Institute, 8 Medical Center Drive, Morgantown, WV, 26505, USA
| |
Collapse
|
59
|
Moog NK, Entringer S, Heim C, Wadhwa PD, Kathmann N, Buss C. Influence of maternal thyroid hormones during gestation on fetal brain development. Neuroscience 2017; 342:68-100. [PMID: 26434624 PMCID: PMC4819012 DOI: 10.1016/j.neuroscience.2015.09.070] [Citation(s) in RCA: 238] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 09/22/2015] [Accepted: 09/25/2015] [Indexed: 01/09/2023]
Abstract
Thyroid hormones (THs) play an obligatory role in many fundamental processes underlying brain development and maturation. The developing embryo/fetus is dependent on maternal supply of TH. The fetal thyroid gland does not commence TH synthesis until mid gestation, and the adverse consequences of severe maternal TH deficiency on offspring neurodevelopment are well established. Recent evidence suggests that even more moderate forms of maternal thyroid dysfunction, particularly during early gestation, may have a long-lasting influence on child cognitive development and risk of neurodevelopmental disorders. Moreover, these observed alterations appear to be largely irreversible after birth. It is, therefore, important to gain a better understanding of the role of maternal thyroid dysfunction on offspring neurodevelopment in terms of the nature, magnitude, time-specificity, and context-specificity of its effects. With respect to the issue of context specificity, it is possible that maternal stress and stress-related biological processes during pregnancy may modulate maternal thyroid function. The possibility of an interaction between the thyroid and stress systems in the context of fetal brain development has, however, not been addressed to date. We begin this review with a brief overview of TH biology during pregnancy and a summary of the literature on its effect on the developing brain. Next, we consider and discuss whether and how processes related to maternal stress and stress biology may interact with and modify the effects of maternal thyroid function on offspring brain development. We synthesize several research areas and identify important knowledge gaps that may warrant further study. The scientific and public health relevance of this review relates to achieving a better understanding of the timing, mechanisms and contexts of thyroid programing of brain development, with implications for early identification of risk, primary prevention and intervention.
Collapse
Affiliation(s)
- N K Moog
- Department of Medical Psychology, Charité University Medicine Berlin, Luisenstrasse 57, 10117 Berlin, Germany
| | - S Entringer
- Department of Medical Psychology, Charité University Medicine Berlin, Luisenstrasse 57, 10117 Berlin, Germany; University of California, Irvine, Development, Health, and Disease Research Program, 333 The City Drive West, Suite 1200, Orange, CA 92868, USA; Department of Pediatrics, University of California, Irvine, School of Medicine, 505 South Main Street, Suite 525, Orange, CA 92868, USA
| | - C Heim
- Department of Medical Psychology, Charité University Medicine Berlin, Luisenstrasse 57, 10117 Berlin, Germany; Department of Biobehavioral Health, Pennsylvania State University, College of Health and Human Development, 219 Biobehavioral Health Building, University Park, PA 16802, USA
| | - P D Wadhwa
- University of California, Irvine, Development, Health, and Disease Research Program, 333 The City Drive West, Suite 1200, Orange, CA 92868, USA; Department of Pediatrics, University of California, Irvine, School of Medicine, 505 South Main Street, Suite 525, Orange, CA 92868, USA; Department of Psychiatry and Human Behavior, University of California, Irvine, School of Medicine, 3117 Gillespie Neuroscience Research Facility, 837 Health Sciences Drive, Irvine, CA 92697, USA; Department of Obstetrics and Gynecology, University of California, Irvine, School of Medicine, 3117 Gillespie Neuroscience Research Facility, 837 Health Sciences Drive, Irvine, CA 92697, USA; Department of Epidemiology, University of California, Irvine, School of Medicine, 3117 Gillespie Neuroscience Research Facility, 837 Health Sciences Drive, Irvine, CA 92697, USA
| | - N Kathmann
- Department of Clinical Psychology, Humboldt-Universität zu Berlin, Rudower Chaussee 18, 12489 Berlin, Germany
| | - C Buss
- Department of Medical Psychology, Charité University Medicine Berlin, Luisenstrasse 57, 10117 Berlin, Germany; University of California, Irvine, Development, Health, and Disease Research Program, 333 The City Drive West, Suite 1200, Orange, CA 92868, USA; Department of Pediatrics, University of California, Irvine, School of Medicine, 505 South Main Street, Suite 525, Orange, CA 92868, USA.
| |
Collapse
|
60
|
Ruan H, Gao J, Qi X, Tao Y, Guo X, Guo Z, Zheng L, Song Y, Liao Y, Shen W. Visual experience dependent regulation of neuronal structure and function by histone deacetylase 1 in developing Xenopus tectum in vivo. Dev Neurobiol 2017; 77:947-962. [PMID: 28033671 DOI: 10.1002/dneu.22480] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 12/13/2016] [Accepted: 12/16/2016] [Indexed: 01/28/2023]
Abstract
Histone deacetylase 1 (HDAC1) is thought to play pivotal roles in neurogenesis and neurodegeneration. However, the role of HDAC1 in neuronal growth and structural plasticity in the developing brain in vivo remains unclear. Here, we show that in the optic tectum of Xenopus laevis, HDAC1 knockdown dramatically decreased the frequency of AMPAR-mediated synaptic currents and increased the frequency of GABAAR-mediated currents, whereas HDAC1 overexpression significantly decreased the frequency of GABAAR-mediated synaptic currents. Both HDAC1 knockdown and overexpression adversely affected dendritic arbor growth and visual experience-dependent structural plasticity. Furthermore, HDAC1 knockdown decreased BDNF expression via a mechanism that involves acetylation of specific histone H4 residues at lysine K5. In particular, the deficits in dendritic growth and visually guided avoidance behavior in HDAC1-knockdown tadpoles could be rescued by acute tectal infusion of BDNF. These results establish a relationship between HDAC1 expression, histone H4 modification and BDNF signaling in the visual-experience dependent regulation of dendritic growth, structural plasticity and function in intact animals in vivo. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 77: 947-962, 2017.
Collapse
Affiliation(s)
- Hangze Ruan
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, 310036, China
| | - Juanmei Gao
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, 310036, China
| | - Xianjie Qi
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, 310036, China
| | - Yi Tao
- Department of Neurosurgery, Nanjing Medical University Affiliated Jiangsu Cancer Hospital, Nanjing, Jiangsu, 210029, China
| | - Xia Guo
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, 310036, China
| | - Zhaoyi Guo
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, 310036, China
| | - Lijun Zheng
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, 310036, China
| | - Yaling Song
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, 310036, China
| | - Yuan Liao
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, 310036, China
| | - Wanhua Shen
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, 310036, China
| |
Collapse
|
61
|
Barnes AK, Koul-Tiwari R, Garner JM, Geist PA, Datta S. Activation of brain-derived neurotrophic factor-tropomyosin receptor kinase B signaling in the pedunculopontine tegmental nucleus: a novel mechanism for the homeostatic regulation of rapid eye movement sleep. J Neurochem 2017; 141:111-123. [PMID: 28027399 PMCID: PMC5364057 DOI: 10.1111/jnc.13938] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 11/09/2016] [Accepted: 12/09/2016] [Indexed: 02/04/2023]
Abstract
Rapid eye movement (REM) sleep dysregulation is a symptom of many neuropsychiatric disorders, yet the mechanisms of REM sleep homeostatic regulation are not fully understood. We have shown that, after REM sleep deprivation, the pedunculopontine tegmental nucleus (PPT) plays a critical role in the generation of recovery REM sleep. In this study, we used multidisciplinary techniques to show a causal relationship between brain-derived neurotrophic factor (BDNF)-tropomyosin receptor kinase B (TrkB) signaling in the PPT and the development of REM sleep homeostatic drive. Rats were randomly assigned to conditions of unrestricted sleep or selective REM sleep deprivation (RSD) with PPT microinjections of vehicle control or a dose of a TrkB receptor inhibitor (2, 3, or 4 nmol K252a or 4 nmol ANA-12). On experimental days, rats received PPT microinjections and their sleep-wake physiological signals were recorded for 3 or 6 h, during which selective RSD was performed in the first 3 h. At the end of all 3 h recordings, rats were killed and the PPT was dissected out for BDNF quantification. Our results show that K252a and ANA-12 dose-dependently reduced the homeostatic responses to selective RSD. Specifically, TrkB receptor inhibition reduced REM sleep homeostatic drive and limited REM sleep rebound. There was also a dose-dependent suppression of PPT BDNF up-regulation, and regression analysis revealed a significant positive relationship between REM sleep homeostatic drive and the level of PPT BDNF expression. These data provide the first direct evidence that activation of BDNF-TrkB signaling in the PPT is a critical step for the development of REM sleep homeostatic drive.
Collapse
Affiliation(s)
- Abigail K Barnes
- Department of Anesthesiology, Graduate School of Medicine, The University of Tennessee, Knoxville, Tennessee, USA.,Department of Psychology, College of Arts and Sciences, The University of Tennessee, Knoxville, Tennessee, USA
| | - Richa Koul-Tiwari
- Department of Anesthesiology, Graduate School of Medicine, The University of Tennessee, Knoxville, Tennessee, USA.,Department of Psychology, College of Arts and Sciences, The University of Tennessee, Knoxville, Tennessee, USA
| | - Jennifer M Garner
- Department of Anesthesiology, Graduate School of Medicine, The University of Tennessee, Knoxville, Tennessee, USA.,Department of Psychology, College of Arts and Sciences, The University of Tennessee, Knoxville, Tennessee, USA
| | - Phillip A Geist
- Department of Anesthesiology, Graduate School of Medicine, The University of Tennessee, Knoxville, Tennessee, USA.,Department of Psychology, College of Arts and Sciences, The University of Tennessee, Knoxville, Tennessee, USA
| | - Subimal Datta
- Department of Anesthesiology, Graduate School of Medicine, The University of Tennessee, Knoxville, Tennessee, USA.,Department of Psychology, College of Arts and Sciences, The University of Tennessee, Knoxville, Tennessee, USA.,Program in Comparative and Experimental Medicine, The University of Tennessee, Knoxville, Tennessee, USA
| |
Collapse
|
62
|
Cissé M, Duplan E, Checler F. The transcription factor XBP1 in memory and cognition: Implications in Alzheimer disease. Mol Med 2017; 22:905-917. [PMID: 28079229 DOI: 10.2119/molmed.2016.00229] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 12/23/2016] [Indexed: 12/21/2022] Open
Abstract
X-box binding protein 1 (XBP1) is a unique basic region leucine zipper transcription factor isolated two decades ago in a search for regulators of major histocompatibility complex class II gene expression. XBP1 is a very complex protein regulating many physiological functions, including immune system, inflammatory responses, and lipid metabolism. Evidence over the past few years suggests that XBP1 also plays important roles in pathological settings since its activity as transcription factor has profound effects on the prognosis and progression of diseases such as cancer, neurodegeneration, and diabetes. Here we provide an overview on recent advances in our understanding of this multifaceted molecule, particularly in regulating synaptic plasticity and memory function, and the implications in neurodegenerative diseases with emphasis on Alzheimer disease.
Collapse
Affiliation(s)
- Moustapha Cissé
- Université Côte d'Azur, INSERM, CNRS, IPMC, team labeled "Fondation pour la Recherche Médicale" and "Laboratory of Excellence (LABEX) Distalz", 660 route des Lucioles, 06560, Sophia-Antipolis, Valbonne, France
| | - Eric Duplan
- Université Côte d'Azur, INSERM, CNRS, IPMC, team labeled "Fondation pour la Recherche Médicale" and "Laboratory of Excellence (LABEX) Distalz", 660 route des Lucioles, 06560, Sophia-Antipolis, Valbonne, France
| | - Frédéric Checler
- Université Côte d'Azur, INSERM, CNRS, IPMC, team labeled "Fondation pour la Recherche Médicale" and "Laboratory of Excellence (LABEX) Distalz", 660 route des Lucioles, 06560, Sophia-Antipolis, Valbonne, France
| |
Collapse
|
63
|
Bode C, Richter F, Spröte C, Brigadski T, Bauer A, Fietz S, Fritschy JM, Richter A. Altered postnatal maturation of striatal GABAergic interneurons in a phenotypic animal model of dystonia. Exp Neurol 2017; 287:44-53. [PMID: 27780732 DOI: 10.1016/j.expneurol.2016.10.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 09/30/2016] [Accepted: 10/21/2016] [Indexed: 01/05/2023]
Abstract
GABAergic disinhibition has been suggested to play a critical role in the pathophysiology of several basal ganglia disorders, including dystonia, a common movement disorder. Previous studies have shown a deficit of striatal GABAergic interneurons (IN) in the dtsz mutant hamster, one of the few phenotypic animal models of dystonia. However, mechanisms underlying this deficit are largely unknown. In the present study, we investigated the migration and maturation of striatal IN during postnatal development (18days of age) and at age of highest severity of dystonia (33days of age) in this hamster model. In line with previous findings, the density of GAD67-positive IN and the level of parvalbumin mRNA, a marker for fast spiking GABAergic IN, were lower in the dtsz mutant than in control hamsters. However, an unaltered density of Nkx2.1 labeled cells and Nkx2.1 mRNA level suggested that the migration of GABAergic IN into the striatum was not retarded. Therefore, different factors that indicate maturation of GABAergic IN were determined. While mRNA of the KCC2 cation/chloride transporters and the cytosolic carboanhydrase VII, used as markers for the so called GABA switch, as well as BDNF were unaltered, we found a reduced number of IN expressing the alpha1 subunit of the GABAA-receptor (37.5%) in dtsz hamsters at an age of 33days, but not after spontaneous remission of dystonia at an age of 90days. Since IN shift expression from alpha2 to alpha1 subunits during postnatal maturation, this result together with a decreased parvalbumin mRNA expression suggest a delayed maturation of striatal GABAergic IN in this animal model, which might underlie abnormal neuronal activity and striatal plasticity.
Collapse
Affiliation(s)
- Christoph Bode
- Institute of Pharmacology, Pharmacy and Toxicology, Faculty of Veterinary Medicine, University of Leipzig, 04103 Leipzig, Germany
| | - Franziska Richter
- Institute of Pharmacology, Pharmacy and Toxicology, Faculty of Veterinary Medicine, University of Leipzig, 04103 Leipzig, Germany.
| | - Christine Spröte
- Institute of Pharmacology, Pharmacy and Toxicology, Faculty of Veterinary Medicine, University of Leipzig, 04103 Leipzig, Germany
| | - Tanja Brigadski
- Institute for Physiology, Medical Faculty, Otto-von-Guericke University, 39120 Magdeburg, Germany; Center of Behavioral Brain Sciences (CBBS), 39120 Magdeburg, Germany
| | - Anne Bauer
- Institute of Pharmacology, Pharmacy and Toxicology, Faculty of Veterinary Medicine, University of Leipzig, 04103 Leipzig, Germany
| | - Simone Fietz
- Institute of Veterinary Anatomy, Histology and Embryology, Faculty of Veterinary Medicine, University of Leipzig, 04103 Leipzig, Germany
| | - Jean-Marc Fritschy
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich 8057, Switzerland
| | - Angelika Richter
- Institute of Pharmacology, Pharmacy and Toxicology, Faculty of Veterinary Medicine, University of Leipzig, 04103 Leipzig, Germany.
| |
Collapse
|
64
|
Veerasakul S, Watiktinkorn P, Thanoi S, Reynolds GP, Nudmamud-Thanoi S. Association of polymorphisms in GAD1 and GAD2 genes with methamphetamine dependence. Pharmacogenomics 2016; 18:17-22. [PMID: 27967329 DOI: 10.2217/pgs-2016-0101] [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] [Indexed: 12/13/2022] Open
Abstract
AIM Association between polymorphisms in GAD genes and methamphetamine (METH) dependence was investigated in the Thai population. MATERIALS & METHODS Genotypes of rs769404 and rs701492 in GAD1 and rs2236418 in GAD2 polymorphisms were determined in 100 METH-dependent male subjects and 102 matched controls. RESULTS The genotype and allele frequencies of rs2236418 (GAD2) were associated with METH dependence and METH with psychosis, in which the G allele was related to increased risk. The presence of the rs769404-rs701492 (GAD1) C-C haplotype was associated with METH psychosis. CONCLUSION This study indicates that genetic variability in GAD1 and GAD2 contributes to risk of METH dependence and METH psychosis in the Thai population and indicates the role of the GABAergic system in these disorders.
Collapse
Affiliation(s)
- Siriluk Veerasakul
- Department of Anatomy, Faculty of Medical Science, Naresuan University, Phitsanulok, 65000, Thailand.,Centre of Excellence in Medical Biotechnology, Faculty of Medical Science, Naresuan University, Phitsanulok, 65000, Thailand
| | | | - Samur Thanoi
- Department of Anatomy, Faculty of Medical Science, Naresuan University, Phitsanulok, 65000, Thailand.,Centre of Excellence in Medical Biotechnology, Faculty of Medical Science, Naresuan University, Phitsanulok, 65000, Thailand
| | - Gavin P Reynolds
- Centre of Excellence in Medical Biotechnology, Faculty of Medical Science, Naresuan University, Phitsanulok, 65000, Thailand.,Biomedical Research Centre, Sheffield Hallam University, Sheffield, S1 1WB, UK
| | - Sutisa Nudmamud-Thanoi
- Department of Anatomy, Faculty of Medical Science, Naresuan University, Phitsanulok, 65000, Thailand.,Centre of Excellence in Medical Biotechnology, Faculty of Medical Science, Naresuan University, Phitsanulok, 65000, Thailand
| |
Collapse
|
65
|
Sedmak G, Jovanov-Milošević N, Puskarjov M, Ulamec M, Krušlin B, Kaila K, Judaš M. Developmental Expression Patterns of KCC2 and Functionally Associated Molecules in the Human Brain. Cereb Cortex 2016; 26:4574-4589. [PMID: 26428952 DOI: 10.1093/cercor/bhv218] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Work on rodents demonstrated that steep upregulation of KCC2, a neuron-specific Cl- extruder of cation-chloride cotransporter (CCC) family, commences in supraspinal structures at around birth, leading to establishment of hyperpolarizing GABAergic responses. We describe spatiotemporal expression profiles of the entire CCC family in human brain. KCC2 mRNA was observed already at 10th postconceptional week (PCW) in amygdala, cerebellum, and thalamus. KCC2-immunoreactive (KCC2-ir) neurons were abundant in subplate at 18 PCW. By 25 PCW, numerous subplate and cortical plate neurons became KCC2-ir. The mRNA expression profiles of α- and β-isoforms of Na-K ATPase, which fuels cation-chloride cotransport, as well of tropomyosin receptor kinase B (TrkB), which promotes developmental upregulation of KCC2, were consistent with data from studies on rodents about their interactions with KCC2. Thus, in human brain, expression of KCC2 and its functionally associated proteins begins in early fetal period. Our work facilitates translation of results on CCC functions from animal studies to human and refutes the view that poor efficacy of anticonvulsants in the term human neonate is attributable to the lack of KCC2. We propose that perinatally low threshold for activation of Ca2+-dependent protease calpain renders neonates susceptible to downregulation of KCC2 by traumatic events, such as perinatal hypoxia ischemia.
Collapse
Affiliation(s)
| | | | - Martin Puskarjov
- Department of Biosciences and Neuroscience Center, University of Helsinki, Viikinkaari 1, Helsinki FI-00014, Finland
| | - Monika Ulamec
- Department of Pathology, Clinical Hospital Center Sisters of Mercy, University of Zagreb School of Medicine, Zagreb 10 000, Croatia
| | - Božo Krušlin
- Department of Pathology, Clinical Hospital Center Sisters of Mercy, University of Zagreb School of Medicine, Zagreb 10 000, Croatia
| | - Kai Kaila
- Department of Biosciences and Neuroscience Center, University of Helsinki, Viikinkaari 1, Helsinki FI-00014, Finland
| | | |
Collapse
|
66
|
Wu H, Shao A, Zhao M, Chen S, Yu J, Zhou J, Liang F, Shi L, Dixon BJ, Wang Z, Ling C, Hong Y, Zhang J. Melatonin attenuates neuronal apoptosis through up-regulation of K(+) -Cl(-) cotransporter KCC2 expression following traumatic brain injury in rats. J Pineal Res 2016; 61:241-50. [PMID: 27159133 DOI: 10.1111/jpi.12344] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 05/05/2016] [Indexed: 01/01/2023]
Abstract
Traumatic brain injury (TBI) initiates a complex cascade of neurochemical and signaling changes that leads to neuronal apoptosis, which contributes to poor outcomes for patients with TBI. The neuron-specific K(+) -Cl(-) cotransporter-2 (KCC2), the principal Cl(-) extruder in adult neurons, plays an important role in Cl(-) homeostasis and neuronal function. This present study was designed to investigate the expression pattern of KCC2 following TBI and to evaluate whether or not melatonin is able to prevent neuronal apoptosis by modulating KCC2 expression in a Sprague Dawley rat controlled cortical impact model of TBI. The time course study showed decreased mRNA and protein expression of KCC2 in the ipsilateral peri-core parietal cortex after TBI. Double immunofluorescence staining demonstrated that KCC2 is located in the plasma membrane of neurons. In addition, melatonin (10 mg/kg) was injected intraperitoneally at 5 minutes and repeated at 1, 2, 3, and 4 hours after brain trauma, and brain samples were extracted 24 hours after TBI. Compared to the vehicle group, melatonin treatment altered the down-regulation of KCC2 expression in both mRNA and protein levels after TBI. Also, melatonin treatment increased the protein levels of brain-derived neurotrophic factor (BDNF) and phosphorylated extracellular signal-regulated kinase (p-ERK). Simultaneously, melatonin administration ameliorated cortical neuronal apoptosis, reduced brain edema, and attenuated neurological deficits after TBI. In conclusion, our findings suggested that melatonin restores KCC2 expression, inhibits neuronal apoptosis and attenuates secondary brain injury after TBI, partially through activation of BDNF/ERK pathway.
Collapse
Affiliation(s)
- Haijian Wu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Department of Neurosurgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Anwen Shao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Mingfei Zhao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Sheng Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jun Yu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jingyi Zhou
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Feng Liang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Ligen Shi
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Brandon J Dixon
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Zhen Wang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Chenhan Ling
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yuan Hong
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Brain Research Institute, Zhejiang University, Hangzhou, Zhejiang, China
- Collaborative Innovation Center for Brain Science, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
67
|
Yuan Q, Yang F, Xiao Y, Tan S, Husain N, Ren M, Hu Z, Martinowich K, Ng JS, Kim PJ, Han W, Nagata KI, Weinberger DR, Je HS. Regulation of Brain-Derived Neurotrophic Factor Exocytosis and Gamma-Aminobutyric Acidergic Interneuron Synapse by the Schizophrenia Susceptibility Gene Dysbindin-1. Biol Psychiatry 2016; 80:312-322. [PMID: 26386481 DOI: 10.1016/j.biopsych.2015.08.019] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 08/12/2015] [Accepted: 08/12/2015] [Indexed: 10/23/2022]
Abstract
BACKGROUND Genetic variations in dystrobrevin binding protein 1 (DTNBP1 or dysbindin-1) have been implicated as risk factors in the pathogenesis of schizophrenia. The encoded protein dysbindin-1 functions in the regulation of synaptic activity and synapse development. Intriguingly, a loss of function mutation in Dtnbp1 in mice disrupted both glutamatergic and gamma-aminobutyric acidergic transmission in the cerebral cortex; pyramidal neurons displayed enhanced excitability due to reductions in inhibitory synaptic inputs. However, the mechanism by which reduced dysbindin-1 activity causes inhibitory synaptic deficits remains unknown. METHODS We investigated the role of dysbindin-1 in the exocytosis of brain-derived neurotrophic factor (BDNF) from cortical excitatory neurons, organotypic brain slices, and acute slices from dysbindin-1 mutant mice and determined how this change in BDNF exocytosis transsynaptically affected the number of inhibitory synapses formed on excitatory neurons via whole-cell recordings, immunohistochemistry, and live-cell imaging using total internal reflection fluorescence microscopy. RESULTS A decrease in dysbindin-1 reduces the exocytosis of BDNF from cortical excitatory neurons, and this reduction in BDNF exocytosis transsynaptically resulted in reduced inhibitory synapse numbers formed on excitatory neurons. Furthermore, application of exogenous BDNF rescued the inhibitory synaptic deficits caused by the reduced dysbindin-1 level in both cultured cortical neurons and slice cultures. CONCLUSIONS Taken together, our results demonstrate that these two genes linked to risk for schizophrenia (BDNF and dysbindin-1) function together to regulate interneuron development and cortical network activity. This evidence supports the investigation of the association between dysbindin-1 and BDNF in humans with schizophrenia.
Collapse
Affiliation(s)
- Qiang Yuan
- Molecular Neurophysiology Laboratory, Signature Program in Neuroscience and Behavioral Disorders, Duke-National University of Singapore Graduate Medical School, Singapore, Singapore
| | - Feng Yang
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
| | - Yixin Xiao
- Molecular Neurophysiology Laboratory, Signature Program in Neuroscience and Behavioral Disorders, Duke-National University of Singapore Graduate Medical School, Singapore, Singapore
| | - Shawn Tan
- Molecular Neurophysiology Laboratory, Signature Program in Neuroscience and Behavioral Disorders, Duke-National University of Singapore Graduate Medical School, Singapore, Singapore
| | - Nilofer Husain
- Molecular Neurophysiology Laboratory, Signature Program in Neuroscience and Behavioral Disorders, Duke-National University of Singapore Graduate Medical School, Singapore, Singapore
| | - Ming Ren
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
| | - Zhonghua Hu
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
| | - Keri Martinowich
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
| | - Julia S Ng
- Molecular Neurophysiology Laboratory, Signature Program in Neuroscience and Behavioral Disorders, Duke-National University of Singapore Graduate Medical School, Singapore, Singapore
| | - Paul J Kim
- Molecular Neurophysiology Laboratory, Signature Program in Neuroscience and Behavioral Disorders, Duke-National University of Singapore Graduate Medical School, Singapore, Singapore
| | - Weiping Han
- Singapore Bioimaging Consortium, Singapore, Singapore
| | - Koh-Ichi Nagata
- Institute for Developmental Research, Aichi Human Service Center, Kasugai, Japan
| | - Daniel R Weinberger
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
| | - H Shawn Je
- Molecular Neurophysiology Laboratory, Signature Program in Neuroscience and Behavioral Disorders, Duke-National University of Singapore Graduate Medical School, Singapore, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
68
|
Jaggi AS, Kaur A, Bali A, Singh N. Expanding Spectrum of Sodium Potassium Chloride Co-transporters in the Pathophysiology of Diseases. Curr Neuropharmacol 2016; 13:369-88. [PMID: 26411965 PMCID: PMC4812803 DOI: 10.2174/1570159x13666150205130359] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Sodium potassium chloride co-transporter (NKCC) belongs to cation-dependent chloride co-transporter family, whose activation allows the entry of Na(+), K(+) and 2Cl(-) inside the cell. It acts in concert with K(+) Cl(-) co-transporter (KCC), which extrudes K(+) and Cl(-) ions from cell. NKCC1 is widely distributed throughout the body, while NKCC2 is exclusively present in kidney. Protein kinase A, protein kinase C, Ste20-related proline-alanine-rich kinase, oxidative stress responsive kinases, With No K=lysine kinase and protein phosphatase type 1 control the phosphorylation/dephosphorylation of key threonine residues of in regulatory domain of NKCC1. The selective inhibitors of NKCC1 including bumetanide and furosemide are conventionally employed as diuretics. However, recent studies have indicated that NKCC1 may be involved in the pathophysiology of anxiety, cerebral ischemia, epilepsy, neuropathic pain, fragile X syndrome, autism and schizophrenia. The inhibitors of NKCC1 are shown to produce anxiolytic effects; attenuate cerebral ischemia-induced neuronal injury; produce antiepileptic effects and attenuate neuropathic pain. In the early developing brain, GABAA activation primarily produces excitatory actions due to high NKCC1/KCC2 ratio. However, as the development progresses, the ratio of NKCC1/KCC2 ratio reverses and there is switch in the polarity of GABAA actions and latter acquires the inhibitory actions. The recapitulation of developmental-like state during pathological state may be associated with increase in the expression and functioning of NKCC1, which decreases the strength of inhibitory GABAergic neurotransmission. The present review describes the expanding role and mechanism of NKCC1 in the pathophysiology of different diseases.
Collapse
Affiliation(s)
- Amteshwar Singh Jaggi
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University Patiala, Patiala- 147002.
| | | | | | | |
Collapse
|
69
|
Allain AE, Cazenave W, Delpy A, Exertier P, Barthe C, Meyrand P, Cattaert D, Branchereau P. Nonsynaptic glycine release is involved in the early KCC2 expression. Dev Neurobiol 2016; 76:764-79. [PMID: 26506510 DOI: 10.1002/dneu.22358] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 09/09/2015] [Accepted: 10/26/2015] [Indexed: 12/20/2022]
Abstract
The cation-chloride co-transporters are important regulators of the cellular Cl(-) homeostasis. Among them the Na(+) -K(+) -2Cl(-) co-transporter (NKCC1) is responsible for intracellular chloride accumulation in most immature brain structures, whereas the K(+) -Cl(-) co-transporter (KCC2) extrudes chloride from mature neurons, ensuring chloride-mediated inhibitory effects of GABA/glycine. We have shown that both KCC2 and NKCC1 are expressed at early embryonic stages (E11.5) in the ventral spinal cord (SC). The mechanisms by which KCC2 is prematurely expressed are unknown. In this study, we found that chronically blocking glycine receptors (GlyR) by strychnine led to a loss of KCC2 expression, without affecting NKCC1 level. This effect was not dependent on the firing of Na(+) action potentials but was mimicked by a Ca(2+) -dependent PKC blocker. Blocking the vesicular release of neurotransmitters did not impinge on strychnine effect whereas blocking volume-sensitive outwardly rectifying (VSOR) chloride channels reproduced the GlyR blockade, suggesting that KCC2 is controlled by a glycine release from progenitor radial cells in immature ventral spinal networks. Finally, we showed that the strychnine treatment prevented the maturation of rhythmic spontaneous activity. Thereby, the GlyR-activation is a necessary developmental process for the expression of functional spinal motor networks. © 2015 Wiley Periodicals, Inc. Develop Neurobiol 76: 764-779, 2016.
Collapse
Affiliation(s)
- Anne-Emilie Allain
- Univ. Bordeaux, INCIA, UMR 5287, Site Talence, F33615 Pessac cedex, France. CNRS, INCIA, UMR 5287, Site Talence, F33615 Pessac cedex, France
| | - William Cazenave
- Univ. Bordeaux, INCIA, UMR 5287, Site Talence, F33615 Pessac cedex, France. CNRS, INCIA, UMR 5287, Site Talence, F33615 Pessac cedex, France
| | - Alain Delpy
- Univ. Bordeaux, INCIA, UMR 5287, Site Talence, F33615 Pessac cedex, France. CNRS, INCIA, UMR 5287, Site Talence, F33615 Pessac cedex, France
| | - Prisca Exertier
- Univ. Bordeaux, INCIA, UMR 5287, Site Talence, F33615 Pessac cedex, France. CNRS, INCIA, UMR 5287, Site Talence, F33615 Pessac cedex, France
| | - Christophe Barthe
- Univ. Bordeaux, Laboratoire De Biotechnologie Des Protéines Recombinantes À Visée Santé, EA 4135, Bordeaux, F-33076, Bordeaux
| | - Pierre Meyrand
- Univ. Bordeaux, IMN, UMR 5293, Site Talence, F33615 Pessac cedex, France. CNRS, IMN, UMR 5293, Site Talence, F33615, Pessac cedex, France
| | - Daniel Cattaert
- Univ. Bordeaux, INCIA, UMR 5287, Site Talence, F33615 Pessac cedex, France. CNRS, INCIA, UMR 5287, Site Talence, F33615 Pessac cedex, France
| | - Pascal Branchereau
- Univ. Bordeaux, INCIA, UMR 5287, Site Talence, F33615 Pessac cedex, France. CNRS, INCIA, UMR 5287, Site Talence, F33615 Pessac cedex, France
| |
Collapse
|
70
|
Eftekhari S, Mehrabi S, Karimzadeh F, Joghataei MT, Khaksarian M, Hadjighassem MR, Katebi M, Soleimani M. Brain Derived Neurotrophic Factor Modification of Epileptiform Burst Discharges in a Temporal Lobe Epilepsy Model. Basic Clin Neurosci 2016; 7:115-20. [PMID: 27303606 PMCID: PMC4892316 DOI: 10.15412/j.bcn.03070205] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
INTRODUCTION Transforming Growth Factor-Beta 1 (TGF-β1) is a pleiotropic cytokine with potent anti-inflammatory property, which has been considered as an essential risk factor in the inflammatory process of Ischemic Stroke (IS), by involving in the pathophysiological progression of hypertension, atherosclerosis, and lipid metabolisms. -509C/T TGF-β1 gene polymorphism has been found to be associated with the risk of IS. The aim of this meta-analysis was to provide a relatively comprehensive account of the relation between -509C/T gene polymorphisms of TGF-β1 and susceptibility to IS. METHODS Male Wistar rats were divided into sham (receiving phosphate buffered saline within dorsal hippocampus), pilocarpine (epileptic model of TLE), single injection BDNF (epileptic rats which received single high dose of BDBF within dorsal hippocampus), and multiple injections BDNF (epileptic rats which received BDNF in days 10, 11, 12, and 13 after induction of TLE) groups. Their electrocorticogram was recorded and amplitude, frequency, and duration of spikes were evaluated. RESULTS Amplitude and frequency of epileptiform burst discharges were significantly decreased in animals treated with BDNF compared to pilocarpine group. CONCLUSION Our findings suggested that BDNF may modulate the epileptic activity in the animal model of TLE. In addition, it may have therapeutic effect for epilepsy. More studies are necessary to clarify the exact mechanisms of BDNF effects.
Collapse
Affiliation(s)
- Sanaz Eftekhari
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.; Department of Neuroscience, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Soraya Mehrabi
- Shefa Neuroscience Center, Khatam-Alanbia Hospital, Tehran, Iran
| | - Fariba Karimzadeh
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad-Taghi Joghataei
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.; Department of Neuroscience, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.; Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mojtaba Khaksarian
- Department of Physiology, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Mahmoud Reza Hadjighassem
- Department of Neuroscience, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.; Brain and Spinal Cord Injury Research Centre, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Majid Katebi
- Department of Anatomy, School of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Mansooreh Soleimani
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.; Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
71
|
Wu H, Che X, Tang J, Ma F, Pan K, Zhao M, Shao A, Wu Q, Zhang J, Hong Y. The K(+)-Cl(-) Cotransporter KCC2 and Chloride Homeostasis: Potential Therapeutic Target in Acute Central Nervous System Injury. Mol Neurobiol 2016; 53:2141-51. [PMID: 25941074 DOI: 10.1007/s12035-015-9162-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 03/26/2015] [Indexed: 12/11/2022]
Abstract
The K(+)-Cl(-) cotransporter-2 (KCC2) is a well-known member of the electroneutral cation-chloride cotransporters with a restricted expression pattern to neurons. This transmembrane protein mediates the efflux of Cl(-) out of neurons and exerts a critical role in inhibitory γ-aminobutyric acidergic (GABAergic) and glycinergic neurotransmission. Moreover, KCC2 participates in the regulation of various physiological processes of neurons, including cell migration, dendritic outgrowth, spine morphology, and dendritic synaptogenesis. It is important to note that down-regulation of KCC2 is associated with the pathogenesis of multiple neurological diseases, which is of particular relevance to acute central nervous system (CNS) injury. In this review, we aim to survey the pathogenic significance of KCC2 down-regulation under the condition of acute CNS injuries. We propose that further elucidation of the molecular mechanisms regarding KCC2 down-regulation after acute CNS injuries is necessary because of potential promising avenues for prevention and treatment of acute CNS injury.
Collapse
Affiliation(s)
- Haijian Wu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Xiaoru Che
- Department of Cardiology, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Junjia Tang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Feiqiang Ma
- Department of Emergency Medicine, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Kun Pan
- Department of Neurosurgery, New York-Presbyterian Hospital, New York, NY, USA
| | - Mingfei Zhao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Anwen Shao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Qun Wu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Yuan Hong
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.
| |
Collapse
|
72
|
Rauti R, Lozano N, León V, Scaini D, Musto M, Rago I, Ulloa Severino FP, Fabbro A, Casalis L, Vázquez E, Kostarelos K, Prato M, Ballerini L. Graphene Oxide Nanosheets Reshape Synaptic Function in Cultured Brain Networks. ACS NANO 2016; 10:4459-71. [PMID: 27030936 DOI: 10.1021/acsnano.6b00130] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Graphene offers promising advantages for biomedical applications. However, adoption of graphene technology in biomedicine also poses important challenges in terms of understanding cell responses, cellular uptake, or the intracellular fate of soluble graphene derivatives. In the biological microenvironment, graphene nanosheets might interact with exposed cellular and subcellular structures, resulting in unexpected regulation of sophisticated biological signaling. More broadly, biomedical devices based on the design of these 2D planar nanostructures for interventions in the central nervous system require an accurate understanding of their interactions with the neuronal milieu. Here, we describe the ability of graphene oxide nanosheets to down-regulate neuronal signaling without affecting cell viability.
Collapse
Affiliation(s)
- Rossana Rauti
- Life Science Department, University of Trieste , 34127 Trieste, Italy
| | - Neus Lozano
- Nanomedicine Lab, School of Medicine and National Graphene Institute, Faculty of Medical & Human Sciences, University of Manchester , M13 9PL Manchester, United Kingdom
| | - Veronica León
- Departamento de Química Orgánica, Facultad de Ciencias y Tecnologías Químicas-IRICA, Universidad de Castilla La Mancha , 13071 Ciudad Real, Spain
| | - Denis Scaini
- Life Science Department, University of Trieste , 34127 Trieste, Italy
- ELETTRA Synchrotron Light Source , 34149 Trieste, Italy
| | - Mattia Musto
- International School for Advanced Studies (SISSA) , 34136 Trieste, Italy
| | - Ilaria Rago
- ELETTRA Synchrotron Light Source , 34149 Trieste, Italy
| | | | - Alessandra Fabbro
- Department of Chemical and Pharmaceutical Sciences, University of Trieste , 34127 Trieste, Italy
| | | | - Ester Vázquez
- Departamento de Química Orgánica, Facultad de Ciencias y Tecnologías Químicas-IRICA, Universidad de Castilla La Mancha , 13071 Ciudad Real, Spain
| | - Kostas Kostarelos
- Nanomedicine Lab, School of Medicine and National Graphene Institute, Faculty of Medical & Human Sciences, University of Manchester , M13 9PL Manchester, United Kingdom
| | - Maurizio Prato
- Department of Chemical and Pharmaceutical Sciences, University of Trieste , 34127 Trieste, Italy
- CIC BiomaGUNE, Parque Tecnológico de San Sebastián, Paseo Miramón, 182, 20009 San Sebastián, Guipúzcoa, Spain
- Basque Foundation for Science , Ikerbasque, Bilbao 48013, Spain
| | - Laura Ballerini
- Life Science Department, University of Trieste , 34127 Trieste, Italy
- International School for Advanced Studies (SISSA) , 34136 Trieste, Italy
| |
Collapse
|
73
|
Kahle KT, Delpire E. Kinase-KCC2 coupling: Cl- rheostasis, disease susceptibility, therapeutic target. J Neurophysiol 2016; 115:8-18. [PMID: 26510764 PMCID: PMC4760510 DOI: 10.1152/jn.00865.2015] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 10/25/2015] [Indexed: 01/06/2023] Open
Abstract
The intracellular concentration of Cl(-) ([Cl(-)]i) in neurons is a highly regulated variable that is established and modulated by the finely tuned activity of the KCC2 cotransporter. Despite the importance of KCC2 for neurophysiology and its role in multiple neuropsychiatric diseases, our knowledge of the transporter's regulatory mechanisms is incomplete. Recent studies suggest that the phosphorylation state of KCC2 at specific residues in its cytoplasmic COOH terminus, such as Ser940 and Thr906/Thr1007, encodes discrete levels of transporter activity that elicit graded changes in neuronal Cl(-) extrusion to modulate the strength of synaptic inhibition via Cl(-)-permeable GABAA receptors. In this review, we propose that the functional and physical coupling of KCC2 to Cl(-)-sensitive kinase(s), such as the WNK1-SPAK kinase complex, constitutes a molecular "rheostat" that regulates [Cl(-)]i and thereby influences the functional plasticity of GABA. The rapid reversibility of (de)phosphorylation facilitates regulatory precision, and multisite phosphorylation allows for the control of KCC2 activity by different inputs via distinct or partially overlapping upstream signaling cascades that may become more or less important depending on the physiological context. While this adaptation mechanism is highly suited to maintaining homeostasis, its adjustable set points may render it vulnerable to perturbation and dysregulation. Finally, we suggest that pharmacological modulation of this kinase-KCC2 rheostat might be a particularly efficacious strategy to enhance Cl(-) extrusion and therapeutically restore GABA inhibition.
Collapse
Affiliation(s)
- Kristopher T Kahle
- Departments of Neurosurgery and Pediatrics, Yale School of Medicine, New Haven, Connecticut; Yale Neurogenetics Program, Yale School of Medicine, New Haven, Connecticut; and
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, Tennessee
| |
Collapse
|
74
|
Taste Bud-Derived BDNF Is Required to Maintain Normal Amounts of Innervation to Adult Taste Buds. eNeuro 2015; 2:eN-NWR-0097-15. [PMID: 26730405 PMCID: PMC4697083 DOI: 10.1523/eneuro.0097-15.2015] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 11/29/2015] [Accepted: 11/30/2015] [Indexed: 11/26/2022] Open
Abstract
Gustatory neurons transmit chemical information from taste receptor cells, which reside in taste buds in the oral cavity, to the brain. As adult taste receptor cells are renewed at a constant rate, nerve fibers must reconnect with new taste receptor cells as they arise. Therefore, the maintenance of gustatory innervation to the taste bud is an active process. Understanding how this process is regulated is a fundamental concern of gustatory system biology. We speculated that because brain-derived neurotrophic factor (BDNF) is required for taste bud innervation during development, it might function to maintain innervation during adulthood. If so, taste buds should lose innervation when Bdnf is deleted in adult mice. To test this idea, we first removed Bdnf from all cells in adulthood using transgenic mice with inducible CreERT2 under the control of the Ubiquitin promoter. When Bdnf was removed, approximately one-half of the innervation to taste buds was lost, and taste buds became smaller because of the loss of taste bud cells. Individual taste buds varied in the amount of innervation each lost, and those that lost the most innervation also lost the most taste bud cells. We then tested the idea that that the taste bud was the source of this BDNF by reducing Bdnf levels specifically in the lingual epithelium and taste buds. Taste buds were confirmed as the source of BDNF regulating innervation. We conclude that BDNF expressed in taste receptor cells is required to maintain normal levels of innervation in adulthood.
Collapse
|
75
|
Veerawatananan B, Surakul P, Chutabhakdikul N. Maternal restraint stress delays maturation of cation-chloride cotransporters and GABAA receptor subunits in the hippocampus of rat pups at puberty. Neurobiol Stress 2015; 3:1-7. [PMID: 26844244 PMCID: PMC4730793 DOI: 10.1016/j.ynstr.2015.12.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 12/09/2015] [Indexed: 12/13/2022] Open
Abstract
The GABAergic synapse undergoes structural and functional maturation during early brain development. Maternal stress alters GABAergic synapses in the pup's brain that are associated with the pathophysiology of neuropsychiatric disorders in adults; however, the mechanism for this is still unclear. In this study, we examined the effects of maternal restraint stress on the development of Cation-Chloride Cotransporters (CCCs) and the GABAA receptor α1 and α5 subunits in the hippocampus of rat pups at different postnatal ages. Our results demonstrate that maternal restraint stress induces a transient but significant increase in the level of NKCC1 (Sodium–Potassium Chloride Cotransporter 1) only at P14, followed by a brief, yet significant increase in the level of KCC2 (Potassium-Chloride Cotransporter 2) at P21, which then decreases from P28 until P40. Thus, maternal stress alters NKCC1 and KCC2 ratio in the hippocampus of rat pups, especially during P14 to P28. Maternal restraint stress also caused biphasic changes in the level of GABAA receptor subunits in the pup's hippocampus. GABAA receptor α1 subunit gradually increased at P14 then decreased thereafter. On the contrary, GABAA receptor α5 subunit showed a transient decrease followed by a long-term increase from P21 until P40. Altogether, our study suggested that the maternal restraint stress might delay maturation of the GABAergic system by altering the expression of NKCC1, KCC2 and GABAA receptor α1 and α5 subunits in the hippocampus of rat pups. These changes demonstrate the dysregulation of inhibitory neurotransmission during early life, which may underlie the pathogenesis of psychiatric diseases at adolescence.
Collapse
Affiliation(s)
- Bovorn Veerawatananan
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Nakornpathom 73170, Thailand
| | - Pornprom Surakul
- Faculty of Allied Health Sciences, Burapha University, Chonburi 20131, Thailand
| | - Nuanchan Chutabhakdikul
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Nakornpathom 73170, Thailand
| |
Collapse
|
76
|
Watanabe M, Fukuda A. Development and regulation of chloride homeostasis in the central nervous system. Front Cell Neurosci 2015; 9:371. [PMID: 26441542 PMCID: PMC4585146 DOI: 10.3389/fncel.2015.00371] [Citation(s) in RCA: 141] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 09/04/2015] [Indexed: 12/22/2022] Open
Abstract
γ-Aminobutyric acid (GABA) is the main inhibitory neurotransmitter of the mature central nervous system (CNS). The developmental switch of GABAergic transmission from excitation to inhibition is induced by changes in Cl− gradients, which are generated by cation-Cl− co-transporters. An accumulation of Cl− by the Na+-K+-2Cl− co-transporter (NKCC1) increases the intracellular Cl− concentration ([Cl−]i) such that GABA depolarizes neuronal precursors and immature neurons. The subsequent ontogenetic switch, i.e., upregulation of the Cl−-extruder KCC2, which is a neuron-specific K+-Cl− co-transporter, with or without downregulation of NKCC1, results in low [Cl−]i levels and the hyperpolarizing action of GABA in mature neurons. Development of Cl− homeostasis depends on developmental changes in NKCC1 and KCC2 expression. Generally, developmental shifts (decreases) in [Cl−]i parallel the maturation of the nervous system, e.g., early in the spinal cord, hypothalamus and thalamus, followed by the limbic system, and last in the neocortex. There are several regulators of KCC2 and/or NKCC1 expression, including brain-derived neurotrophic factor (BDNF), insulin-like growth factor (IGF), and cystic fibrosis transmembrane conductance regulator (CFTR). Therefore, regionally different expression of these regulators may also contribute to the regional developmental shifts of Cl− homeostasis. KCC2 and NKCC1 functions are also regulated by phosphorylation by enzymes such as PKC, Src-family tyrosine kinases, and WNK1–4 and their downstream effectors STE20/SPS1-related proline/alanine-rich kinase (SPAK)-oxidative stress responsive kinase-1 (OSR1). In addition, activation of these kinases is modulated by humoral factors such as estrogen and taurine. Because these transporters use the electrochemical driving force of Na+ and K+ ions, topographical interaction with the Na+-K+ ATPase and its modulators such as creatine kinase (CK) should modulate functions of Cl− transporters. Therefore, regional developmental regulation of these regulators and modulators of Cl− transporters may also play a pivotal role in the development of Cl− homeostasis.
Collapse
Affiliation(s)
- Miho Watanabe
- Department of Neurophysiology, Hamamatsu University School of Medicine Hamamatsu, Japan
| | - Atsuo Fukuda
- Department of Neurophysiology, Hamamatsu University School of Medicine Hamamatsu, Japan
| |
Collapse
|
77
|
Zhou L, Zhou W, Zhang S, Liu B, Liang P, Zhou Y, Zhou T, Zhang K, Leng Y, Kong W. BDNF signaling in the rat cerebello-vestibular pathway during vestibular compensation: BDNF signaling in vestibular compensation. FEBS J 2015; 282:3579-91. [PMID: 26111610 DOI: 10.1111/febs.13360] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2014] [Revised: 05/18/2015] [Accepted: 06/23/2015] [Indexed: 02/04/2023]
Abstract
Vestibular compensation, which is the behavioral recovery from lesions to the peripheral vestibular system, is attributed to plasticity of the central vestibular system. It has been reported that brain-derived neurotrophic factor (BDNF) is expressed and released in an activity-dependent manner. Upon binding to the tyrosine receptor kinase B (TrkB), BDNF can acutely modulate synaptic transmission and plasticity in the central nervous system. To assess the possible contribution of BDNF to this recovery process, we studied the expression of BDNF, TrkB.FL, TrkB.T1 and KCC2 (K(+) -Cl(-) cotransporter isoform 2) in the bilateral medial vestibular nucleus (MVN) and the flocculus of rats at 4 h, 8 h, 1, 3 and 7 days following unilateral labyrinthectomy (UL) using immunohistochemistry, quantitative real-time PCR and western blotting. Our results have shown that, compared with the sham controls and the contra-lesional side, (a) the expression of BDNF and TrkB.FL increased at 4 h in the ipsi-lesional flocculus after UL; (b) the expression of TrkB.T1 decreased at 4 h and KCC2 decreased at 8 h and 1 day in the ipsi-lesional flocculus after UL; and (c) BDNF and TrkB.FL expression was enhanced and KCC2 expression was reduced in the ipsi-lesional MVN at 8 h after UL. Our data supported the hypothesis that BDNF upregulation may reduce the inhibitory effects of the flocculus and commissural inhibition system by regulating inhibitory GABAergic synaptic transmission in floccular Purkinje cells and Purkinje cell terminals in the MVN. Additionally, KCC2 may be a switch in this process.
Collapse
Affiliation(s)
- Liuqing Zhou
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wen Zhou
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sulin Zhang
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Liu
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pei Liang
- Department of Neurobiology, Bielefeld University, Germany
| | - Yan Zhou
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Zhou
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kun Zhang
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yangming Leng
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weijia Kong
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
78
|
Saenen ND, Plusquin M, Bijnens E, Janssen BG, Gyselaers W, Cox B, Fierens F, Molenberghs G, Penders J, Vrijens K, De Boever P, Nawrot TS. In Utero Fine Particle Air Pollution and Placental Expression of Genes in the Brain-Derived Neurotrophic Factor Signaling Pathway: An ENVIRONAGE Birth Cohort Study. ENVIRONMENTAL HEALTH PERSPECTIVES 2015; 123:834-40. [PMID: 25816123 PMCID: PMC4529006 DOI: 10.1289/ehp.1408549] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 03/24/2015] [Indexed: 05/18/2023]
Abstract
BACKGROUND Developmental processes in the placenta and the fetal brain are shaped by the same biological signals. Recent evidence suggests that adaptive responses of the placenta to the maternal environment may influence central nervous system development. OBJECTIVES We studied the association between in utero exposure to fine particle air pollution with a diameter ≤ 2.5 μm (PM2.5) and placental expression of genes implicated in neural development. METHODS Expression of 10 target genes in the brain-derived neurotrophic factor (BDNF) signaling pathway were quantified in placental tissue of 90 mother-infant pairs from the ENVIRONAGE birth cohort using quantitative real-time polymerase chain reaction. Trimester-specific PM2.5 exposure levels were estimated for each mother's home address using a spatiotemporal model. Mixed-effects models were used to evaluate the association between the target genes and PM2.5 exposure measured in different time windows of pregnancy. RESULTS A 5-μg/m3 increase in residential PM2.5 exposure during the first trimester of pregnancy was associated with a 15.9% decrease [95% confidence interval (CI): -28.7, -3.2%, p = 0.015] in expression of placental BDNF at birth. The corresponding estimate for synapsin 1 (SYN1) was a 24.3% decrease (95% CI: -42.8, -5.8%, p = 0.011). CONCLUSIONS Placental expression of BDNF and SYN1, two genes implicated in normal neurodevelopmental trajectories, decreased with increasing in utero exposure to PM2.5. Future studies are needed to confirm our findings and evaluate the potential relevance of associations between PM2.5 and placental expression of BDNF and SYN1 on neurodevelopment. We provide the first molecular epidemiological evidence concerning associations between in utero fine particle air pollution exposure and the expression of genes that may influence neurodevelopmental processes.
Collapse
Affiliation(s)
- Nelly D Saenen
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Limburg, Belgium
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
79
|
Tashiro S, Shinozaki M, Mukaino M, Renault-Mihara F, Toyama Y, Liu M, Nakamura M, Okano H. BDNF Induced by Treadmill Training Contributes to the Suppression of Spasticity and Allodynia After Spinal Cord Injury via Upregulation of KCC2. Neurorehabil Neural Repair 2015; 29:677-89. [PMID: 25527489 DOI: 10.1177/1545968314562110] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Spasticity and allodynia are major sequelae that affect the quality of life and daily activities of spinal cord injury (SCI) patients. Although rehabilitation ameliorates spasticity and allodynia, the molecular mechanisms involved in these processes remain elusive. OBJECTIVE To investigate the molecular mechanisms by which rehabilitation ameliorates spasticity and allodynia after SCI in rats. METHODS The expression levels of brain-derived neurotrophic factor (BDNF) and potassium-chloride cotransporter-2 (KCC2), as well as the localization of KCC2, were examined in the lumbar enlargements of untrained and treadmill-trained thoracic SCI model rats. Spasticity and allodynia were determined via behavioral and electrophysiological analyses. The effects of BDNF on spasticity, allodynia, and KCC2 activation were determined by inhibition of BDNF signaling via intrathecal administration of TrkB-IgG. The effects of SCI and training on the expression levels of functional phospholipase C-γ in the lumbar enlargement were also examined. RESULTS Treadmill training after SCI upregulated endogenous BDNF expression and posttranslational modification of KCC2 in the lumbar enlargement significantly. There were also significant correlations between increased KCC2 expression and ameliorated spasticity and allodynia. Administration of TrkB-IgG abrogated the training-induced upregulation of KCC2 and beneficial effects on spasticity and allodynia. The expression level of functional phospholipase C-γ was reduced significantly after SCI, which may have contributed to the change in the function of BDNF, whereby it did not trigger short-term downregulation or induce long-term upregulation of KCC2 expression secondary to training. CONCLUSIONS BDNF-mediated restoration of KCC2 expression underlies the suppression of spasticity and allodynia caused by rehabilitation.
Collapse
Affiliation(s)
- Syoichi Tashiro
- Department of Rehabilitation Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Munehisa Shinozaki
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | | | | | - Yoshiaki Toyama
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Meigen Liu
- Department of Rehabilitation Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Masaya Nakamura
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
80
|
Abstract
Axonal guidance and synaptic specification depends on specific signaling mechanisms that occur in growth cones. While several signaling pathways implicated in cone navigation have been identified, membrane dynamics in growth cones remains largely unknown. We took advantage of SynaptopHluorin and high-speed optical recordings to monitor the patterns of membrane dynamics in rat hippocampal growth cones. We show that exocytosis occurs both at the peripheral and central domains, including filopodia, and that SynaptopHluorin signals occur as spontaneous patterned peaks. Such transients average approximately two per minute and last ∼30 s. We also demonstrate that the chemoattractant Netrin-1 elicits increases in the frequency and slopes of these transients, with peaks averaging up to six per minute in the peripheral domain. Netrin-1-dependent regulation of exocytotic events requires the activation of the Erk1/2 and SFK pathways. Furthermore, we show that domains with high SynaptopHluorin signals correlate with high local calcium concentrations and that local, spontaneous calcium increases are associated with higher SynaptopHluorin signals. These findings demonstrate highly stereotyped, spontaneous transients of local exocytosis in growth cones and that these transients are positively regulated by chemoattractant molecules such as Netrin-1.
Collapse
|
81
|
From 2D to 3D: novel nanostructured scaffolds to investigate signalling in reconstructed neuronal networks. Sci Rep 2015; 5:9562. [PMID: 25910072 PMCID: PMC5407555 DOI: 10.1038/srep09562] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 03/10/2015] [Indexed: 12/21/2022] Open
Abstract
To recreate in vitro 3D neuronal circuits will ultimately increase the relevance of results from cultured to whole-brain networks and will promote enabling technologies for neuro-engineering applications. Here we fabricate novel elastomeric scaffolds able to instruct 3D growth of living primary neurons. Such systems allow investigating the emerging activity, in terms of calcium signals, of small clusters of neurons as a function of the interplay between the 2D or 3D architectures and network dynamics. We report the ability of 3D geometry to improve functional organization and synchronization in small neuronal assemblies. We propose a mathematical modelling of network dynamics that supports such a result. Entrapping carbon nanotubes in the scaffolds remarkably boosted synaptic activity, thus allowing for the first time to exploit nanomaterial/cell interfacing in 3D growth support. Our 3D system represents a simple and reliable construct, able to improve the complexity of current tissue culture models.
Collapse
|
82
|
Das SK, Barhwal K, Hota SK, Thakur MK, Srivastava RB. Disrupting monotony during social isolation stress prevents early development of anxiety and depression like traits in male rats. BMC Neurosci 2015; 16:2. [PMID: 25880744 PMCID: PMC4336522 DOI: 10.1186/s12868-015-0141-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 01/21/2015] [Indexed: 01/11/2023] Open
Abstract
Background Although there have been several reports on social isolation induced mood alterations, the independent contribution of monotonous environment in mediating mood alterations has been less studied. In view of the above, the present study is aimed at investigating the relative contribution of monotony towards mood alterations during isolation stress. Monotony was induced in a specially designed isolation chamber in male Sprague-Dawley rats in the presence or absence of isolation by housing animals singly (SH) or in pairs (PH). Novel objects were introduced to disrupt monotony in singly housed animals (SHNO) or paired housed animals (PHNO). Behavioural alterations were assessed using Open field test (OFT), Elevated Plus Maze (EPM) and Forced Swim Test (FST). Neuro-morphological changes in the CA3 region of hippocampus were studied by cresyl violet and golgi-cox staining. Hippocampal serotonin and 5-hydroxy indole acetic acid (5-HIAA) levels were estimated along with the expression of phospho-insulin like growth factor-1 receptor (pIGF-1R) and phospho cyclic AMP response-element binding protein (pCREB). Serotonin was depleted by administering Para-chlorophenylalanine (PCPA) to a separate PH group (PHPCPA), PHNO group (PHNOPCPA) and SHNO group (SHNOPCPA) to determine the role of serotonin in mediating monotony induced emotional mal-adaptations. Results The results showed anxiety and depression like traits in both PH and SH groups during behavioural test such as OFT, EPM and FST. Pyknosis along with decrease in apical dendritic arborization was observed in the CA3 region of SH group along with decrease in serotonin and reduced expression of pIGF-1R and pCREB. Disrupting monotony through intervention of novel objects in PHNO and SHNO groups ameliorated anxiety and depression like traits and augmented pIGF-1R along with increase in serotonin level. Depletion of hippocampal serotonin level by PCPA administration in PHNOPCPA and SHNOPCPA groups on the other hand resulted in altered mood state despite disruption of monotony by novel objects intervention. Conclusion The findings of our study suggest that monotonous environment independently contributes to impairment in mood state and disrupting monotony by intervention of novel objects during social isolation prevents mood disorders and emotional maladaptation through up regulation of hippocampal pIGF-1R and increase in serotonin.
Collapse
Affiliation(s)
- Saroj Kumar Das
- Experimental Biology Division, Defence Institute of High Altitude Research, Defence Research Development Organisation, Leh-Ladakh, C/O- 56 APO, Jammu and Kashmir, 901205, India.
| | - Kalpana Barhwal
- Experimental Biology Division, Defence Institute of High Altitude Research, Defence Research Development Organisation, Leh-Ladakh, C/O- 56 APO, Jammu and Kashmir, 901205, India.
| | - Sunil Kumar Hota
- Experimental Biology Division, Defence Institute of High Altitude Research, Defence Research Development Organisation, Leh-Ladakh, C/O- 56 APO, Jammu and Kashmir, 901205, India.
| | | | - Ravi Bihari Srivastava
- Experimental Biology Division, Defence Institute of High Altitude Research, Defence Research Development Organisation, Leh-Ladakh, C/O- 56 APO, Jammu and Kashmir, 901205, India.
| |
Collapse
|
83
|
Ansell BRE, Dwyer DB, Wood SJ, Bora E, Brewer WJ, Proffitt TM, Velakoulis D, McGorry PD, Pantelis C. Divergent effects of first-generation and second-generation antipsychotics on cortical thickness in first-episode psychosis. Psychol Med 2015; 45:515-527. [PMID: 25077698 PMCID: PMC4413868 DOI: 10.1017/s0033291714001652] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2013] [Revised: 03/19/2014] [Accepted: 06/16/2014] [Indexed: 01/31/2023]
Abstract
BACKGROUND Whether there are differential effects of first-generation antipsychotics (FGAs) and second-generation antipsychotics (SGAs) on the brain is currently debated. Although some studies report that FGAs reduce grey matter more than SGAs, others do not, and research to date is limited by a focus on schizophrenia spectrum disorders. To address this limitation, this study investigated the effects of medication in patients being treated for first-episode schizophrenia or affective psychoses. METHOD Cortical thickness was compared between 52 first-episode psychosis patients separated into diagnostic (i.e. schizophrenia or affective psychosis) and medication (i.e. FGA and SGA) subgroups. Patients in each group were also compared to age- and sex-matched healthy controls (n = 28). A whole-brain cortical thickness interaction analysis of medication and diagnosis was then performed. Correlations between cortical thickness with antipsychotic dose and psychotic symptoms were examined. RESULTS The effects of medication and diagnosis did not interact, suggesting independent effects. Compared with controls, diagnostic differences were found in frontal, parietal and temporal regions. Decreased thickness in FGA-treated versus SGA-treated groups was found in a large frontoparietal region (p < 0.001, corrected). Comparisons with healthy controls revealed decreased cortical thickness in the FGA group whereas the SGA group showed increases in addition to decreases. In FGA-treated patients cortical thinning was associated with higher negative symptoms whereas increased cortical thickness in the SGA-treated group was associated with lower positive symptoms. CONCLUSIONS Our results suggest that FGA and SGA treatments have divergent effects on cortical thickness during the first episode of psychosis that are independent from changes due to illness.
Collapse
Affiliation(s)
- B. R. E. Ansell
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, University of Melbourne and Melbourne Health, Carlton South, Victoria, Australia
| | - D. B. Dwyer
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, University of Melbourne and Melbourne Health, Carlton South, Victoria, Australia
| | - S. J. Wood
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, University of Melbourne and Melbourne Health, Carlton South, Victoria, Australia
- School of Psychology, University of Birmingham, UK
| | - E. Bora
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, University of Melbourne and Melbourne Health, Carlton South, Victoria, Australia
| | - W. J. Brewer
- Orygen Youth Health Research Centre, University of Melbourne, Parkville, Victoria, Australia
| | - T. M. Proffitt
- Orygen Youth Health Research Centre, University of Melbourne, Parkville, Victoria, Australia
| | - D. Velakoulis
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, University of Melbourne and Melbourne Health, Carlton South, Victoria, Australia
| | - P. D. McGorry
- Orygen Youth Health Research Centre, University of Melbourne, Parkville, Victoria, Australia
| | - C. Pantelis
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, University of Melbourne and Melbourne Health, Carlton South, Victoria, Australia
| |
Collapse
|
84
|
Luscher B, Fuchs T. GABAergic control of depression-related brain states. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2015; 73:97-144. [PMID: 25637439 DOI: 10.1016/bs.apha.2014.11.003] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The GABAergic deficit hypothesis of major depressive disorders (MDDs) posits that reduced γ-aminobutyric acid (GABA) concentration in brain, impaired function of GABAergic interneurons, altered expression and function of GABA(A) receptors, and changes in GABAergic transmission dictated by altered chloride homeostasis can contribute to the etiology of MDD. Conversely, the hypothesis posits that the efficacy of currently used antidepressants is determined by their ability to enhance GABAergic neurotransmission. We here provide an update for corresponding evidence from studies of patients and preclinical animal models of depression. In addition, we propose an explanation for the continued lack of genetic evidence that explains the considerable heritability of MDD. Lastly, we discuss how alterations in GABAergic transmission are integral to other hypotheses of MDD that emphasize (i) the role of monoaminergic deficits, (ii) stress-based etiologies, (iii) neurotrophic deficits, and (iv) the neurotoxic and neural circuit-impairing consequences of chronic excesses of glutamate. We propose that altered GABAergic transmission serves as a common denominator of MDD that can account for all these other hypotheses and that plays a causal and common role in diverse mechanistic etiologies of depressive brain states and in the mechanism of action of current antidepressant drug therapies.
Collapse
Affiliation(s)
- Bernhard Luscher
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania, USA; Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, USA; Center for Molecular Investigation of Neurological Disorders, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, USA.
| | - Thomas Fuchs
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania, USA; Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, USA; Center for Molecular Investigation of Neurological Disorders, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, USA
| |
Collapse
|
85
|
Puskarjov M, Ahmad F, Khirug S, Sivakumaran S, Kaila K, Blaesse P. BDNF is required for seizure-induced but not developmental up-regulation of KCC2 in the neonatal hippocampus. Neuropharmacology 2015; 88:103-9. [PMID: 25229715 DOI: 10.1016/j.neuropharm.2014.09.005] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 09/01/2014] [Accepted: 09/04/2014] [Indexed: 01/21/2023]
Abstract
A robust increase in the functional expression of the neuronal K-Cl cotransporter KCC2 during CNS development is necessary for the emergence of hyperpolarizing ionotropic GABAergic transmission. BDNF-TrkB signaling has been implicated in the developmental up-regulation of KCC2 and, in mature animals, in fast activity-dependent down-regulation of KCC2 function following seizures and trauma. In contrast to the decrease in KCC2 expression observed in the adult hippocampus following trauma, seizures in the neonate trigger a TrkB-dependent up-regulation of neuronal Cl(-) extrusion capacity associated with enhanced surface expression of KCC2. Here, we show that this effect is transient, and impaired in the hippocampus of Bdnf(-/-) mice. Notably, however, a complete absence of BDNF does not compromise the increase in KCC2 protein or K-Cl transport functionality during neuronal development. Furthermore, we present data indicating that the functional up-regulation of KCC2 by neonatal seizures is temporally limited by calpain activity.
Collapse
Affiliation(s)
- Martin Puskarjov
- Department of Biosciences and Neuroscience Center, University of Helsinki, FI-00014 Helsinki, Finland
| | - Faraz Ahmad
- Department of Biosciences and Neuroscience Center, University of Helsinki, FI-00014 Helsinki, Finland
| | - Stanislav Khirug
- Department of Biosciences and Neuroscience Center, University of Helsinki, FI-00014 Helsinki, Finland
| | - Sudhir Sivakumaran
- Department of Biosciences and Neuroscience Center, University of Helsinki, FI-00014 Helsinki, Finland
| | - Kai Kaila
- Department of Biosciences and Neuroscience Center, University of Helsinki, FI-00014 Helsinki, Finland
| | - Peter Blaesse
- Department of Biosciences and Neuroscience Center, University of Helsinki, FI-00014 Helsinki, Finland.
| |
Collapse
|
86
|
Kaila K, Price TJ, Payne JA, Puskarjov M, Voipio J. Cation-chloride cotransporters in neuronal development, plasticity and disease. Nat Rev Neurosci 2014; 15:637-54. [PMID: 25234263 DOI: 10.1038/nrn3819] [Citation(s) in RCA: 505] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Electrical activity in neurons requires a seamless functional coupling between plasmalemmal ion channels and ion transporters. Although ion channels have been studied intensively for several decades, research on ion transporters is in its infancy. In recent years, it has become evident that one family of ion transporters, cation-chloride cotransporters (CCCs), and in particular K(+)-Cl(-) cotransporter 2 (KCC2), have seminal roles in shaping GABAergic signalling and neuronal connectivity. Studying the functions of these transporters may lead to major paradigm shifts in our understanding of the mechanisms underlying brain development and plasticity in health and disease.
Collapse
Affiliation(s)
- Kai Kaila
- 1] Department of Biosciences, University of Helsinki, 00014 Helsinki, Finland. [2] Neuroscience Center, University of Helsinki, 00014 Helsinki, Finland
| | - Theodore J Price
- University of Texas at Dallas, School of Behavior and Brain Sciences, Dallas, Texas 75093, USA
| | - John A Payne
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, California 95616, USA
| | - Martin Puskarjov
- 1] Department of Biosciences, University of Helsinki, 00014 Helsinki, Finland. [2] Neuroscience Center, University of Helsinki, 00014 Helsinki, Finland
| | - Juha Voipio
- Department of Biosciences, University of Helsinki, 00014 Helsinki, Finland
| |
Collapse
|
87
|
Liu DY, Shen XM, Yuan FF, Guo OY, Zhong Y, Chen JG, Zhu LQ, Wu J. The Physiology of BDNF and Its Relationship with ADHD. Mol Neurobiol 2014; 52:1467-1476. [PMID: 25354496 DOI: 10.1007/s12035-014-8956-6] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 10/20/2014] [Indexed: 12/16/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is a major neurotrophin in the central nervous system that plays a critical role in the physiological brain functions via its two independent receptors: tropomyosin-related kinase B (TrkB) and p75, especially in the neurodevelopment. Disrupting of BDNF and its downstream signals has been found in many neuropsychological diseases, including attention-deficit hyperactivity disorder (ADHD), a common mental disorder which is prevalent in childhood. Understanding the physiological functions of BDNF during neural development and its potential relationship with ADHD will help us to elucidate the possible mechanisms of ADHD and to develop therapeutic approaches for this disease. In this review, we summarized the important literatures for the physiological functions of BDNF in the neurodevelopment. We also performed an association study on the functional genetic variation of BDNF and ADHD by a case-control study in the Chinese mainland population and revealed the potential correlation between BDNF and ADHD which needs further research to confirm.
Collapse
Affiliation(s)
- De-Yi Liu
- Department of Pathophysiology, School of Basic Medicine, Institute of Brain Research, Sino-Canada Collaborative Platform on Molecular Biology of Neurological Disease, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Xue-Mei Shen
- Key Laboratory of Environment and Health, Ministry of Education & Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Fang-Fen Yuan
- Key Laboratory of Environment and Health, Ministry of Education & Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Ou-Yang Guo
- Department of Pathophysiology, School of Basic Medicine, Institute of Brain Research, Sino-Canada Collaborative Platform on Molecular Biology of Neurological Disease, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Yan Zhong
- Department of Child Health Care, Hunan Children's Hospital, Changsha, 410007, People's Republic of China
| | - Jian-Guo Chen
- Department of Pharmacology, School of Basic Medicine, Institute of Brain Research, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Ling-Qiang Zhu
- Department of Pathophysiology, School of Basic Medicine, Institute of Brain Research, Sino-Canada Collaborative Platform on Molecular Biology of Neurological Disease, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
| | - Jing Wu
- Key Laboratory of Environment and Health, Ministry of Education & Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
| |
Collapse
|
88
|
Ishrat T, Pillai B, Soliman S, Fouda AY, Kozak A, Johnson MH, Ergul A, Fagan SC. Low-dose candesartan enhances molecular mediators of neuroplasticity and subsequent functional recovery after ischemic stroke in rats. Mol Neurobiol 2014; 51:1542-53. [PMID: 25084762 DOI: 10.1007/s12035-014-8830-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 07/22/2014] [Indexed: 01/19/2023]
Abstract
We have previously reported that angiotensin type 1 receptor (AT1R) blockade with candesartan exerts neurovascular protection after experimental cerebral ischemia. Here, we tested the hypothesis that a low, subhypotensive dose of candesartan enhances neuroplasticity and subsequent functional recovery through enhanced neurotrophic factor expression in rats subjected to ischemia reperfusion injury. Male Wistar rats (290-300 g) underwent 90 min of middle cerebral artery occlusion (MCAO) and received candesartan (0.3 mg/kg) or saline at reperfusion and then once every 24 h for 7 days. Functional deficits were assessed in a blinded manner at 1, 3, 7, and 14 days after MCAO. Animals were sacrificed 14-day post-stroke and the brains perfused for infarct size by cresyl violet. Western blot and immunohistochemistry were used to assess the expression of growth factors and synaptic proteins. Candesartan-treated animals showed a significant reduction in the infarct size [t (13) = -5.5, P = 0.0001] accompanied by functional recovery in Bederson [F (1, 13) = 7.9, P = 0.015], beam walk [F (1, 13) = 6.7, P = 0.023], grip strength [F (1, 13) = 15.2, P = 0.0031], and rotarod performance [F (1, 14) = 29.8, P < 0.0001]. In addition, candesartan-treated animals showed significantly higher expression of active metalloproteinase-3 (MMP-3), laminin, and angiopoietin-1 (Ang-1). The expression of vascular endothelial growth factor (VEGF) and brain-derived neurotrophic factor (BDNF) and its receptor was significantly increased in the animals treated with candesartan. Also, we observed significant increases in neuroplasticity markers, synaptophysin, and PSD-95. These results indicate that low-dose candesartan had a large and enduring effect on measures of plasticity, and this accompanied the functional recovery after ischemic stroke.
Collapse
|
89
|
A time course analysis of the electrophysiological properties of neurons differentiated from human induced pluripotent stem cells (iPSCs). PLoS One 2014; 9:e103418. [PMID: 25072157 PMCID: PMC4114788 DOI: 10.1371/journal.pone.0103418] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2013] [Accepted: 07/02/2014] [Indexed: 11/19/2022] Open
Abstract
Many protocols have been designed to differentiate human embryonic stem cells (ESCs) and human induced pluripotent stem cells (iPSCs) into neurons. Despite the relevance of electrophysiological properties for proper neuronal function, little is known about the evolution over time of important neuronal electrophysiological parameters in iPSC-derived neurons. Yet, understanding the development of basic electrophysiological characteristics of iPSC-derived neurons is critical for evaluating their usefulness in basic and translational research. Therefore, we analyzed the basic electrophysiological parameters of forebrain neurons differentiated from human iPSCs, from day 31 to day 55 after the initiation of neuronal differentiation. We assayed the developmental progression of various properties, including resting membrane potential, action potential, sodium and potassium channel currents, somatic calcium transients and synaptic activity. During the maturation of iPSC-derived neurons, the resting membrane potential became more negative, the expression of voltage-gated sodium channels increased, the membrane became capable of generating action potentials following adequate depolarization and, at day 48–55, 50% of the cells were capable of firing action potentials in response to a prolonged depolarizing current step, of which 30% produced multiple action potentials. The percentage of cells exhibiting miniature excitatory post-synaptic currents increased over time with a significant increase in their frequency and amplitude. These changes were associated with an increase of Ca2+ transient frequency. Co-culturing iPSC-derived neurons with mouse glial cells enhanced the development of electrophysiological parameters as compared to pure iPSC-derived neuronal cultures. This study demonstrates the importance of properly evaluating the electrophysiological status of the newly generated neurons when using stem cell technology, as electrophysiological properties of iPSC-derived neurons mature over time.
Collapse
|
90
|
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: 11.5] [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
|
91
|
Akman O, Moshé SL, Galanopoulou AS. Sex-specific consequences of early life seizures. Neurobiol Dis 2014; 72 Pt B:153-66. [PMID: 24874547 DOI: 10.1016/j.nbd.2014.05.021] [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: 03/25/2014] [Revised: 05/07/2014] [Accepted: 05/17/2014] [Indexed: 12/29/2022] Open
Abstract
Seizures are very common in the early periods of life and are often associated with poor neurologic outcome in humans. Animal studies have provided evidence that early life seizures may disrupt neuronal differentiation and connectivity, signaling pathways, and the function of various neuronal networks. There is growing experimental evidence that many signaling pathways, like GABAA receptor signaling, the cellular physiology and differentiation, or the functional maturation of certain brain regions, including those involved in seizure control, mature differently in males and females. However, most experimental studies of early life seizures have not directly investigated the importance of sex on the consequences of early life seizures. The sexual dimorphism of the developing brain raises the question that early seizures could have distinct effects in immature females and males that are subjected to seizures. We will first discuss the evidence for sex-specific features of the developing brain that could be involved in modifying the susceptibility and consequences of early life seizures. We will then review how sex-related biological factors could modify the age-specific consequences of induced seizures in the immature animals. These include signaling pathways (e.g., GABAA receptors), steroid hormones, growth factors. Overall, there are very few studies that have specifically addressed seizure outcomes in developing animals as a function of sex. The available literature indicates that a variety of outcomes (histopathological, behavioral, molecular, epileptogenesis) may be affected in a sex-, age-, region-specific manner after seizures during development. Obtaining a better understanding for the gender-related mechanisms underlying epileptogenesis and seizure comorbidities will be necessary to develop better gender and age appropriate therapies.
Collapse
Affiliation(s)
- Ozlem Akman
- Department of Physiology, Faculty of Medicine, Istanbul Bilim University, 34394 Istanbul, Turkey.
| | - Solomon L Moshé
- Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Montefiore Epilepsy Management Center, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, 10461, USA; Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, 10461, USA; Department of Pediatrics, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, 10461, USA
| | - Aristea S Galanopoulou
- Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Montefiore Epilepsy Management Center, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, 10461, USA; Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, 10461, USA
| |
Collapse
|
92
|
Gao XP, Liu Q, Nair B, Wong-Riley MTT. Reduced levels of brain-derived neurotrophic factor contribute to synaptic imbalance during the critical period of respiratory development in rats. Eur J Neurosci 2014; 40:2183-95. [PMID: 24666389 DOI: 10.1111/ejn.12568] [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: 08/27/2013] [Revised: 02/15/2014] [Accepted: 02/18/2014] [Indexed: 02/06/2023]
Abstract
Previously, our electrophysiological studies revealed a transient imbalance between suppressed excitation and enhanced inhibition in hypoglossal motoneurons of rats on postnatal days (P) 12-13, a critical period when abrupt neurochemical, metabolic, ventilatory and physiological changes occur in the respiratory system. The mechanism underlying the imbalance is poorly understood. We hypothesised that the imbalance was contributed by a reduced expression of brain-derived neurotrophic factor (BDNF), which normally enhances excitation and suppresses inhibition. We also hypothesised that exogenous BDNF would partially reverse this synaptic imbalance. Immunohistochemistry/single-neuron optical densitometry, real-time quantitative PCR (RT-qPCR) and whole-cell patch-clamp recordings were done on hypoglossal motoneurons in brainstem slices of rats during the first three postnatal weeks. Our results indicated that: (1) the levels of BDNF and its high-affinity tyrosine receptor kinase B (TrkB) receptor mRNAs and proteins were relatively high during the first 1-1.5 postnatal weeks, but dropped precipitously at P12-13 before rising again afterwards; (2) exogenous BDNF significantly increased the normally lowered frequency of spontaneous excitatory postsynaptic currents but decreased the normally heightened amplitude and frequency of spontaneous inhibitory postsynaptic currents (sIPSCs) during the critical period; (3) exogenous BDNF also decreased the normally heightened frequency of miniature IPSCs at P12-13; and (4) the effect of exogenous BDNF was partially blocked by K252a, a TrkB receptor antagonist. Thus, our results are consistent with our hypothesis that BDNF and TrkB play an important role in the synaptic imbalance during the critical period. This may have significant implications for the mechanism underlying sudden infant death syndrome.
Collapse
Affiliation(s)
- Xiu-Ping Gao
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI, 53226, USA
| | | | | | | |
Collapse
|
93
|
Chaudhury S, Nag TC, Jain S, Wadhwa S. Role of sound stimulation in reprogramming brain connectivity. J Biosci 2014; 38:605-14. [PMID: 23938392 DOI: 10.1007/s12038-013-9341-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Sensory stimulation has a critical role to play in the development of an individual. Environmental factors tend to modify the inputs received by the sensory pathway. The developing brain is most vulnerable to these alterations and interacts with the environment to modify its neural circuitry. In addition to other sensory stimuli, auditory stimulation can also act as external stimuli to provide enrichment during the perinatal period. There is evidence that suggests that enriched environment in the form of auditory stimulation can play a substantial role in modulating plasticity during the prenatal period. This review focuses on the emerging role of prenatal auditory stimulation in the development of higher brain functions such as learning and memory in birds and mammals. The molecular mechanisms of various changes in the hippocampus following sound stimulation to effect neurogenesis, learning and memory are described. Sound stimulation can also modify neural connectivity in the early postnatal life to enhance higher cognitive function or even repair the secondary damages in various neurological and psychiatric disorders. Thus, it becomes imperative to examine in detail the possible ameliorating effects of prenatal sound stimulation in existing animal models of various psychiatric disorders, such as autism.
Collapse
Affiliation(s)
- Sraboni Chaudhury
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India.
| | | | | | | |
Collapse
|
94
|
Hypothalamic dysfunction of the thrombospondin receptor α2δ-1 underlies the overeating and obesity triggered by brain-derived neurotrophic factor deficiency. J Neurosci 2014; 34:554-65. [PMID: 24403154 DOI: 10.1523/jneurosci.1572-13.2014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) and its receptor, TrkB, are critical components of the neural circuitry controlling appetite and body weight. Diminished BDNF signaling in mice results in severe hyperphagia and obesity. In humans, BDNF haploinsufficiency and the functional Bdnf Val66Met polymorphism have been linked to elevated food intake and body weight. The mechanisms underlying this dysfunction are poorly defined. We demonstrate a chief role of α2δ-1, a calcium channel subunit and thrombospondin receptor, in triggering overeating in mice with central BDNF depletion. We show reduced α2δ-1 cell-surface expression in the BDNF mutant ventromedial hypothalamus (VMH), an energy balance-regulating center. This deficit contributes to the hyperphagia exhibited by BDNF mutant mice because selective inhibition of α2δ-1 by gabapentin infusion into wild-type VMH significantly increases feeding and body weight gain. Importantly, viral-mediated α2δ-1 rescue in BDNF mutant VMH significantly mitigates their hyperphagia, obesity, and liver steatosis and normalizes deficits in glucose homeostasis. Whole-cell recordings in BDNF mutant VMH neurons revealed normal calcium currents but reduced frequency of EPSCs. These results suggest calcium channel-independent effects of α2δ-1 on feeding and implicate α2δ-1-thrombospondin interactions known to facilitate excitatory synapse assembly. Our findings identify a central mechanism mediating the inhibitory effects of BDNF on feeding. They also demonstrate a novel and critical role for α2δ-1 in appetite control and suggest a mechanism underlying weight gain in humans treated with gabapentinoid drugs.
Collapse
|
95
|
Witte M, Reinert T, Dietz B, Nerlich J, Rübsamen R, Milenkovic I. Depolarizing chloride gradient in developing cochlear nucleus neurons: Underlying mechanism and implication for calcium signaling. Neuroscience 2014; 261:207-22. [DOI: 10.1016/j.neuroscience.2013.12.050] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Revised: 12/16/2013] [Accepted: 12/23/2013] [Indexed: 11/24/2022]
|
96
|
Medina I, Friedel P, Rivera C, Kahle KT, Kourdougli N, Uvarov P, Pellegrino C. Current view on the functional regulation of the neuronal K(+)-Cl(-) cotransporter KCC2. Front Cell Neurosci 2014; 8:27. [PMID: 24567703 PMCID: PMC3915100 DOI: 10.3389/fncel.2014.00027] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 01/18/2014] [Indexed: 12/22/2022] Open
Abstract
In the mammalian central nervous system (CNS), the inhibitory strength of chloride (Cl(-))-permeable GABAA and glycine receptors (GABAAR and GlyR) depends on the intracellular Cl(-) concentration ([Cl(-)]i). Lowering [Cl(-)]i enhances inhibition, whereas raising [Cl(-)]i facilitates neuronal activity. A neuron's basal level of [Cl(-)]i, as well as its Cl(-) extrusion capacity, is critically dependent on the activity of the electroneutral K(+)-Cl(-) cotransporter KCC2, a member of the SLC12 cation-Cl(-) cotransporter (CCC) family. KCC2 deficiency compromises neuronal migration, formation and the maturation of GABAergic and glutamatergic synaptic connections, and results in network hyperexcitability and seizure activity. Several neurological disorders including multiple epilepsy subtypes, neuropathic pain, and schizophrenia, as well as various insults such as trauma and ischemia, are associated with significant decreases in the Cl(-) extrusion capacity of KCC2 that result in increases of [Cl(-)]i and the subsequent hyperexcitability of neuronal networks. Accordingly, identifying the key upstream molecular mediators governing the functional regulation of KCC2, and modifying these signaling pathways with small molecules, might constitute a novel neurotherapeutic strategy for multiple diseases. Here, we discuss recent advances in the understanding of the mechanisms regulating KCC2 activity, and of the role these mechanisms play in neuronal Cl(-) homeostasis and GABAergic neurotransmission. As KCC2 mediates electroneutral transport, the experimental recording of its activity constitutes an important research challenge; we therefore also, provide an overview of the different methodological approaches utilized to monitor function of KCC2 in both physiological and pathological conditions.
Collapse
Affiliation(s)
- Igor Medina
- INSERM, Institut de Neurobiologie de la Méditerranée (INMED)Marseille, France
- Aix-Marseille Université, UMR901Marseille, France
| | - Perrine Friedel
- INSERM, Institut de Neurobiologie de la Méditerranée (INMED)Marseille, France
- Aix-Marseille Université, UMR901Marseille, France
| | - Claudio Rivera
- INSERM, Institut de Neurobiologie de la Méditerranée (INMED)Marseille, France
- Aix-Marseille Université, UMR901Marseille, France
- Neuroscience Center, University of HelsinkiHelsinki, Finland
| | - Kristopher T. Kahle
- Department of Cardiology, Manton Center for Orphan Disease Research, Howard Hughes Medical Institute, Boston Children's HospitalBoston, MA, USA
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical SchoolBoston, MA, USA
| | - Nazim Kourdougli
- INSERM, Institut de Neurobiologie de la Méditerranée (INMED)Marseille, France
- Aix-Marseille Université, UMR901Marseille, France
| | - Pavel Uvarov
- Institute of Biomedicine, Anatomy, University of HelsinkiHelsinki, Finland
| | - Christophe Pellegrino
- INSERM, Institut de Neurobiologie de la Méditerranée (INMED)Marseille, France
- Aix-Marseille Université, UMR901Marseille, France
| |
Collapse
|
97
|
|
98
|
Sawano E, Takahashi M, Negishi T, Tashiro T. Thyroid hormone-dependent development of the GABAergic pre- and post-synaptic components in the rat hippocampus. Int J Dev Neurosci 2013; 31:751-61. [PMID: 24076339 DOI: 10.1016/j.ijdevneu.2013.09.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 08/30/2013] [Accepted: 09/17/2013] [Indexed: 10/26/2022] Open
Abstract
Thyroid hormone (TH) plays essential roles in normal brain development mainly by regulating gene expression through binding to specific nuclear receptors which serve as transcription factors. Previous studies showed that perinatal deficiency of TH or impairment of its signaling severely affect brain development, especially the development of the γ-aminobutyric acid (GABA) system, but cellular and molecular targets of the hormone are only partly uncovered. In the present study, we focused on the developing rat hippocampus which was confirmed to be one of the regions highly sensitive to TH status, and found two new targets of the hormone among the pre- and post-synaptic components of the GABAergic system. One was glutamic acid decarboxylase 65 (GAD65), the protein level of which was reduced to less than 50% of control in the hippocampus of hypothyroid rats (obtained by administering 0.025% methimazole in drinking water to pregnant dams from gestational day 15 until 4 weeks postpartum) and recovered to control levels by daily thyroxine-replacement after birth. Reduction in GAD65 protein was correlated immunohistochemically with a 37% reduction in the number of GAD65-positive cells as well as a reduction in GAD65-positive processes. In contrast, the other GAD isotype, GAD67, was not affected by TH status. A subpopulation of GABAergic neurons containing parvalbumin was also confirmed to be highly dependent on TH status. The second target of thyroid hormone was neuron-specific K(+)/Cl(-) co-transporter, KCC2, which is responsible for switching of GABA action from excitatory to inhibitory. In the euthyroid hippocampus, a sharp rise of kcc2 expression was observed at postnatal day (PND)10 which was followed by a large increase in KCC2 protein at PND15. This transient rise in kcc2 expression was completely suppressed by hypothyroidism, resulting in nearly 80% reduction in KCC2 protein at PND15. These results indicate that the development of GABAergic terminals and the excitatory to inhibitory maturation of GABA signaling are strongly dependent on TH.
Collapse
Affiliation(s)
- Erika Sawano
- Department of Chemistry and Biological Science, School of Science and Engineering, Aoyama Gakuin University, 5-10-1 Fuchinobe, Chou-ku, Sagamihara, Kanagawa 252-5258, Japan.
| | | | | | | |
Collapse
|
99
|
Possible contribution of IGF-1 to depressive disorder. Pharmacol Rep 2013; 65:1622-31. [DOI: 10.1016/s1734-1140(13)71523-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 09/13/2013] [Indexed: 11/20/2022]
|
100
|
Ambrogini P, Lattanzi D, Ciuffoli S, Betti M, Fanelli M, Cuppini R. Physical exercise and environment exploration affect synaptogenesis in adult-generated neurons in the rat dentate gyrus: possible role of BDNF. Brain Res 2013; 1534:1-12. [PMID: 23973748 DOI: 10.1016/j.brainres.2013.08.023] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 08/06/2013] [Accepted: 08/12/2013] [Indexed: 01/02/2023]
Abstract
A brief training in a pool maze, with or without cognitive tasks, modifies the synaptogenesis and maturation of newborn neurons in adult rat dentate gyrus. These types of trainings have many aspects, including physical activity and exploration. Therefore, to evaluate whether physical exercise and environment exploration are able to affect synapse formation and the maturation of adult-generated neurons, GFP-retrovirus infusion was performed on rats which, on the fourth day after injection, were housed under running conditions or allowed to explore an enriched environment briefly in the absence of exercise for the following three days. Afterward, at the end of the trainings, electrophysiological and morphological studies were conducted. Considering that neurotrophic factors increase after exercise or environment exploration, hippocampal BDNF levels and TrkB receptor activation were evaluated. In this study, we show that both spontaneous physical activity and enriched environment exploration induced synaptogenesis and T-type voltage-dependent Ca(2+) currents in very immature neurons. Hippocampal BDNF levels and TrkB receptor activation were determined to be increasing following physical activity and exploration. A possible contribution of BDNF signaling in mediating the observed effects was supported by the use of 7-8-dihydroxyflavone, a selective TrkB agonist, and of ANA-12, an inhibitor of TrkB receptors.
Collapse
Affiliation(s)
- P Ambrogini
- Department of Earth, Life and Environment Sciences, Section of Physiology, University of Urbino "Carlo Bo", Urbino 61029, Italy.
| | | | | | | | | | | |
Collapse
|