1
|
Luo W, Egger M, Cruz-Ochoa N, Tse A, Maloveczky G, Tamás B, Lukacsovich D, Seng C, Amrein I, Lukacsovich T, Wolfer D, Földy C. Activation of feedforward wiring in adult hippocampal neurons by the basic-helix-loop-helix transcription factor Ascl4. PNAS NEXUS 2024; 3:pgae174. [PMID: 38711810 PMCID: PMC11071515 DOI: 10.1093/pnasnexus/pgae174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 04/17/2024] [Indexed: 05/08/2024]
Abstract
Although evidence indicates that the adult brain retains a considerable capacity for circuit formation, adult wiring has not been broadly considered and remains poorly understood. In this study, we investigate wiring activation in adult neurons. We show that the basic-helix-loop-helix transcription factor Ascl4 can induce wiring in different types of hippocampal neurons of adult mice. The new axons are mainly feedforward and reconfigure synaptic weights in the circuit. Mice with the Ascl4-induced circuits do not display signs of pathology and solve spatial problems equally well as controls. Our results demonstrate reprogrammed connectivity by a single transcriptional factor and provide insights into the regulation of brain wiring in adults.
Collapse
Affiliation(s)
- Wenshu Luo
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zürich, Zürich 8057, Switzerland
| | - Matteo Egger
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zürich, Zürich 8057, Switzerland
- Adaptive Brain Circuits in Development and Learning (AdaBD), University Research Priority Program (URPP), University of Zürich, Zürich 8057, Switzerland
| | - Natalia Cruz-Ochoa
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zürich, Zürich 8057, Switzerland
- Adaptive Brain Circuits in Development and Learning (AdaBD), University Research Priority Program (URPP), University of Zürich, Zürich 8057, Switzerland
| | - Alice Tse
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zürich, Zürich 8057, Switzerland
| | - Gyula Maloveczky
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zürich, Zürich 8057, Switzerland
| | - Bálint Tamás
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zürich, Zürich 8057, Switzerland
| | - David Lukacsovich
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zürich, Zürich 8057, Switzerland
| | - Charlotte Seng
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zürich, Zürich 8057, Switzerland
| | - Irmgard Amrein
- Institute of Anatomy, Faculty of Medicine, University of Zürich, Zürich 8057, Switzerland
| | - Tamás Lukacsovich
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zürich, Zürich 8057, Switzerland
| | - David Wolfer
- Institute of Anatomy, Faculty of Medicine, University of Zürich, Zürich 8057, Switzerland
- Institute of Human Movement Sciences and Sport, D-HEST, ETH Zürich, Zürich 8057, Switzerland
| | - Csaba Földy
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zürich, Zürich 8057, Switzerland
- Adaptive Brain Circuits in Development and Learning (AdaBD), University Research Priority Program (URPP), University of Zürich, Zürich 8057, Switzerland
| |
Collapse
|
2
|
Vandael D, Jonas P. Structure, biophysics, and circuit function of a "giant" cortical presynaptic terminal. Science 2024; 383:eadg6757. [PMID: 38452088 DOI: 10.1126/science.adg6757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 01/19/2024] [Indexed: 03/09/2024]
Abstract
The hippocampal mossy fiber synapse, formed between axons of dentate gyrus granule cells and dendrites of CA3 pyramidal neurons, is a key synapse in the trisynaptic circuitry of the hippocampus. Because of its comparatively large size, this synapse is accessible to direct presynaptic recording, allowing a rigorous investigation of the biophysical mechanisms of synaptic transmission and plasticity. Furthermore, because of its placement in the very center of the hippocampal memory circuit, this synapse seems to be critically involved in several higher network functions, such as learning, memory, pattern separation, and pattern completion. Recent work based on new technologies in both nanoanatomy and nanophysiology, including presynaptic patch-clamp recording, paired recording, super-resolution light microscopy, and freeze-fracture and "flash-and-freeze" electron microscopy, has provided new insights into the structure, biophysics, and network function of this intriguing synapse. This brings us one step closer to answering a fundamental question in neuroscience: how basic synaptic properties shape higher network computations.
Collapse
Affiliation(s)
- David Vandael
- Institute of Science and Technology Austria (ISTA), A-3400 Klosterneuburg, Austria
| | - Peter Jonas
- Institute of Science and Technology Austria (ISTA), A-3400 Klosterneuburg, Austria
| |
Collapse
|
3
|
Cline HT, Lau M, Hiramoto M. Activity-dependent Organization of Topographic Neural Circuits. Neuroscience 2023; 508:3-18. [PMID: 36470479 PMCID: PMC9839526 DOI: 10.1016/j.neuroscience.2022.11.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022]
Abstract
Sensory information in the brain is organized into spatial representations, including retinotopic, somatotopic, and tonotopic maps, as well as ocular dominance columns. The spatial representation of sensory inputs is thought to be a fundamental organizational principle that is important for information processing. Topographic maps are plastic throughout an animal's life, reflecting changes in development and aging of brain circuitry, changes in the periphery and sensory input, and changes in circuitry, for instance in response to experience and learning. Here, we review mechanisms underlying the role of activity in the development, stability and plasticity of topographic maps, focusing on recent work suggesting that the spatial information in the visual field, and the resulting spatiotemporal patterns of activity, provide instructive cues that organize visual projections.
Collapse
Affiliation(s)
- Hollis T Cline
- Department of Neuroscience and the Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA, USA.
| | - Melissa Lau
- Department of Neuroscience and the Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA, USA
| | - Masaki Hiramoto
- Department of Neuroscience and the Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
4
|
Yasuoka A, Nagai T, Lee S, Miyaguchi H, Saito Y, Abe K, Asakura T. Mastication stimuli enhance the learning ability of weaning-stage rats, altering the hippocampal neuron transcriptome and micromorphology. Front Behav Neurosci 2022; 16:1006359. [PMID: 36263297 PMCID: PMC9574334 DOI: 10.3389/fnbeh.2022.1006359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 09/08/2022] [Indexed: 11/13/2022] Open
Abstract
Mastication stimuli are known to relieve senile dementia in human and animal studies. However, few studies have focused on its effect on weaning-stage animals and the underlying molecular processes. In this study, 3-week-old male rats were raised on a powdered (P-group) or chow (C-group) diet for 8 days, and their behavior was examined using the Y-maze and novel object recognition tests. In the Y-maze test, the C-group rats showed a larger alternation ratio than the P-group rats. In the novel object recognition test, the C-group rats exhibited a significantly larger discrimination index for novel objects than for familiar objects, but the P-group rats did not. We then compared the hippocampal neuron morphology and transcriptome between the groups. C-group rats exhibited larger dendrite branch numbers in the apical dendrites of pyramidal cells in the cornu ammonis 1 (CA1) region and a larger spine density in the basal dendrites of CA1 neurons than the P-group rats. Using DNA microarray analysis, we identified 621 (P < C) and 96 (P > C) genes that were differentially expressed between the groups. These genes were enriched in functional terms related to dendrite growth and included the Igf2, RhoA, and Rho GEF genes, most of which were upregulated in the C-group. These results suggest that the mastication stimuli during the weaning period can enhance the learning ability of rats by increasing the dendrite branches of hippocampal CA1 neurons and by regulating genes related to dendrite growth.
Collapse
Affiliation(s)
- Akihito Yasuoka
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
- Department of Human Nutrition, Seitoku University, Chiba, Japan
| | - Toshitada Nagai
- Department of Applied Biological Science, Takasaki University of Health and Welfare, Takasaki, Japan
| | - Seonmi Lee
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Hitonari Miyaguchi
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Yoshikazu Saito
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Keiko Abe
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
- Kanagawa Institute of Industrial Science and Technology, Kawasaki, Japan
| | - Tomiko Asakura
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
- *Correspondence: Tomiko Asakura,
| |
Collapse
|
5
|
Sex Differences in the Spatial Behavior Functions of Adult-Born Neurons in Rats. eNeuro 2022; 9:ENEURO.0054-22.2022. [PMID: 35473765 PMCID: PMC9116935 DOI: 10.1523/eneuro.0054-22.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/13/2022] [Accepted: 04/15/2022] [Indexed: 01/04/2023] Open
Abstract
Adult neurogenesis modifies hippocampal circuits and behavior, but removing newborn neurons does not consistently alter spatial processing, a core function of the hippocampus. Additionally, little is known about sex differences in neurogenesis since few studies have compared males and females. Since adult-born neurons regulate the stress response, we hypothesized that spatial functions may be more prominent under aversive conditions and may differ between males and females given sex differences in stress responding. We therefore trained intact and neurogenesis-deficient rats in the spatial water maze at temperatures that vary in their degree of aversiveness. In the standard water maze, ablating neurogenesis did not alter spatial learning in either sex. However, in cold water, ablating neurogenesis had divergent sex-dependent effects: relative to intact rats, male neurogenesis-deficient rats were slower to escape the maze and female neurogenesis-deficient rats were faster. Neurogenesis promoted temperature-related changes in search strategy in females, but it promoted search strategy stability in males. Females displayed greater recruitment (Fos expression) of the dorsal hippocampus than males, particularly in cold water. However, blocking neurogenesis did not alter Fos expression in either sex. Finally, morphologic analyses revealed greater experience-dependent plasticity in males. Adult-born neurons in males and females had similar morphology at baseline but training increased spine density and reduced presynaptic terminal size, specifically in males. Collectively, these findings indicate that adult-born neurons contribute to spatial learning in stressful conditions and they provide new evidence for sex differences in their behavioral functions.
Collapse
|
6
|
Liu D, Chen J, Hu X, Hu G, Liu Y, Yang K, Xiao C, Zou Y, Liu H. Contralesional homotopic functional plasticity in patients with temporal glioma. J Neurosurg 2021; 134:417-425. [PMID: 31923896 DOI: 10.3171/2019.11.jns191982] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 11/05/2019] [Indexed: 11/06/2022]
Abstract
OBJECTIVE This study aimed to explore the contralesional homotopic functional plasticity in the brain of patients with unilateral temporal glioma. METHODS Demographic, neurocognitive, and resting-state functional MRI data were collected from 17 patients with temporal glioma (10 in the right lobe and 7 in the left lobe), along with 14 age- and sex-matched healthy controls. The amplitude of low-frequency fluctuation (ALFF) of the contralesional homotopic region and 2 control regions was examined. The region-of-interest-based analysis was used to determine the altered functional connectivity (FC) of the contralesional homotopic region, showing significantly different intrinsic regional brain activity between patients and controls. Partial correlation analysis was conducted to determine the association between the altered neural activity and behavioral characteristics. RESULTS Compared with controls, patients with right temporal glioma exhibited significantly increased ALFF in the contralesional homotopic hippocampus and parahippocampal region. In addition, the intrinsic regional activity in these regions was negatively correlated with the visuospatial score (r = -0.718, p = 0.045). Whole-brain FC analysis revealed significantly increased FC between the left hippocampus and parahippocampal regions and the left inferior temporal gyrus, and decreased FC between the left hippocampus and parahippocampal regions and the left inferior frontal gyrus. No significant changes were found in the 2 control regions. CONCLUSIONS Contralesional homotopic regions are instrumental in the process of neural plasticity and functional compensation observed in patients with unilateral temporal glioma. The observed findings might be used to help preoperative evaluation or rehabilitation of postsurgical patients.
Collapse
Affiliation(s)
- Dongming Liu
- 1Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu
| | - Jiu Chen
- 2Institute of Neuropsychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Fourth Clinical College of Nanjing Medical University, Nanjing, Jiangsu
- 3Institute of Brain Functional Imaging, Nanjing Medical University, Nanjing, Jiangsu; and
| | - Xinhua Hu
- 1Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu
- 3Institute of Brain Functional Imaging, Nanjing Medical University, Nanjing, Jiangsu; and
| | - Guanjie Hu
- 1Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu
| | - Yong Liu
- 1Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu
| | - Kun Yang
- 1Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu
| | - Chaoyong Xiao
- 3Institute of Brain Functional Imaging, Nanjing Medical University, Nanjing, Jiangsu; and
- 4Department of Radiology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuanjie Zou
- 1Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu
- 3Institute of Brain Functional Imaging, Nanjing Medical University, Nanjing, Jiangsu; and
| | - Hongyi Liu
- 1Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu
- 3Institute of Brain Functional Imaging, Nanjing Medical University, Nanjing, Jiangsu; and
| |
Collapse
|
7
|
Synapse type-specific proteomic dissection identifies IgSF8 as a hippocampal CA3 microcircuit organizer. Nat Commun 2020; 11:5171. [PMID: 33057002 PMCID: PMC7560607 DOI: 10.1038/s41467-020-18956-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 09/23/2020] [Indexed: 12/20/2022] Open
Abstract
Excitatory and inhibitory neurons are connected into microcircuits that generate circuit output. Central in the hippocampal CA3 microcircuit is the mossy fiber (MF) synapse, which provides powerful direct excitatory input and indirect feedforward inhibition to CA3 pyramidal neurons. Here, we dissect its cell-surface protein (CSP) composition to discover novel regulators of MF synaptic connectivity. Proteomic profiling of isolated MF synaptosomes uncovers a rich CSP composition, including many CSPs without synaptic function and several that are uncharacterized. Cell-surface interactome screening identifies IgSF8 as a neuronal receptor enriched in the MF pathway. Presynaptic Igsf8 deletion impairs MF synaptic architecture and robustly decreases the density of bouton filopodia that provide feedforward inhibition. Consequently, IgSF8 loss impairs excitation/inhibition balance and increases excitability of CA3 pyramidal neurons. Our results provide insight into the CSP landscape and interactome of a specific excitatory synapse and reveal IgSF8 as a critical regulator of CA3 microcircuit connectivity and function. Mossy fiber synapses are key in CA3 microcircuit function. Here, the authors profile the mossy fiber synapse proteome and cell-surface interactome. They uncover a diverse repertoire of cell-surface proteins and identify the receptor IgSF8 as a regulator of CA3 microcircuit connectivity and function.
Collapse
|
8
|
Besnard A, Sahay A. Enhancing adult neurogenesis promotes contextual fear memory discrimination and activation of hippocampal-dorsolateral septal circuits. Behav Brain Res 2020; 399:112917. [PMID: 32949641 DOI: 10.1016/j.bbr.2020.112917] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/01/2020] [Accepted: 09/09/2020] [Indexed: 12/21/2022]
Abstract
Hippocampal circuitry is continuously modified by integration of adult-born dentate granule cells (DGCs). Prior work has shown that enhancing adult hippocampal neurogenesis decreases interference or overlap or conflict between ensembles of similar contexts and promotes discrimination of a shock-associated context from a similar, neutral context. However, the impact of enhanced integration of adult-born neurons on hippocampal network activity or downstream circuits such as the dorsolateral septum that mediate defensive behavioral responses is poorly understood. Here, we first replicated our finding that genetic expansion of the population of adult-born dentate granule cells (8 weeks and younger) promotes contextual fear discrimination. We found that enhanced contextual fear discrimination is associated with greater c-Fos expression in discrete hippocampal subfields along the proximo-distal and dorsoventral axis. Examination of the dorsolateral septum revealed an increase in activation of somatostatin expressing neurons consistent with recent characterization of these cells as calibrators of defensive behavior. Together, these findings begin to shed light on how genetically enhancing adult hippocampal neurogenesis affects activity of hippocampal-dorsolateral septal circuits.
Collapse
Affiliation(s)
- Antoine Besnard
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA; Harvard Stem Cell Institute, Cambridge, MA, 02138, USA; Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Amar Sahay
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA; Harvard Stem Cell Institute, Cambridge, MA, 02138, USA; Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA; BROAD Institute of Harvard and MIT, Cambridge, MA, 02142, USA.
| |
Collapse
|
9
|
Adult-Born Hippocampal Neurons Undergo Extended Development and Are Morphologically Distinct from Neonatally-Born Neurons. J Neurosci 2020; 40:5740-5756. [PMID: 32571837 DOI: 10.1523/jneurosci.1665-19.2020] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 02/28/2020] [Accepted: 05/04/2020] [Indexed: 11/21/2022] Open
Abstract
During immature stages, adult-born neurons pass through critical periods for survival and plasticity. It is generally assumed that by 2 months of age adult-born neurons are mature and equivalent to the broader neuronal population, raising questions of how they might contribute to hippocampal function in old age when neurogenesis has declined. However, few have examined adult-born neurons beyond the critical period or directly compared them to neurons born in infancy. Here, we used a retrovirus to visualize functionally relevant morphological features of 2- to 24-week-old adult-born neurons in male rats. From 2 to 7 weeks, neurons grew and attained a relatively mature phenotype. However, several features of 7-week-old neurons suggested a later wave of growth: these neurons had larger nuclei, thicker dendrites, and more dendritic filopodia than all other groups. Indeed, between 7 and 24 weeks, adult-born neurons gained additional dendritic branches, formed a second primary dendrite, acquired more mushroom spines, and had enlarged mossy fiber presynaptic terminals. Compared with neonatal-born neurons, old adult-born neurons had greater spine density, larger presynaptic terminals, and more putative efferent filopodial contacts onto inhibitory neurons. By integrating rates of cell birth and growth across the life span, we estimate that adult neurogenesis ultimately produces half of the cells and the majority of spines in the dentate gyrus. Critically, protracted development contributes to the plasticity of the hippocampus through to the end of life, even after cell production declines. Persistent differences from neonatal-born neurons may additionally endow adult-born neurons with unique functions even after they have matured.SIGNIFICANCE STATEMENT Neurogenesis occurs in the hippocampus throughout adult life and contributes to memory and emotion. It is generally assumed that new neurons have the greatest impact on behavior when they are immature and plastic. However, since neurogenesis declines dramatically with age, it is unclear how they might contribute to behavior later in life when cell proliferation has slowed. Here we find that newborn neurons mature over many months in rats and may end up with distinct morphological features compared with neurons born in infancy. Using a mathematical model, we estimate that a large fraction of neurons is added in adulthood. Moreover, their extended growth produces a reserve of plasticity that persists even after neurogenesis has declined to low rates.
Collapse
|
10
|
Qi Z, Yang X, Sang Y, Liu Y, Li J, Xu B, Liu W, He M, Xu Z, Deng Y, Zhu J. Fluoxetine and Riluzole Mitigates Manganese-Induced Disruption of Glutamate Transporters and Excitotoxicity via Ephrin-A3/GLAST-GLT-1/Glu Signaling Pathway in Striatum of Mice. Neurotox Res 2020; 38:508-523. [PMID: 32472497 DOI: 10.1007/s12640-020-00209-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 04/09/2020] [Accepted: 04/14/2020] [Indexed: 01/05/2023]
Abstract
Manganese (Mn) is an essential element required for many biological processes and systems in the human body. Mn intoxication increases brain glutamate (Glu) levels causing neuronal damage. Recent studies have reported that ephrin-A3 regulates this glutamate transporter. However, none has explored the role of this crucial molecule in Mn-induced excitotoxicity. The present study investigated whether ephrin-A3/GLAST-GLT-1/Glu signaling pathway participates in Mn-induced excitotoxicity using astrocytes and Kunming mice. The mechanisms were explored using fluoxetine (ephrin-A3 inhibitor) and riluzole (a Glu release inhibitor). Firstly, we demonstrated that Mn exposure (500 μM or 50 mg/kg MnCl2) significantly increased Mn, ephrin-A3, and Glu levels, and inhibited Na+-K+ ATPase activity, as well as mRNA and protein levels of GLAST and GLT-1. Secondly, we found that astrocytes and mice pretreated with fluoxetine (100 μM or 15 mg/kg) and riluzole (100 μM or 32 μmol/kg) prior to Mn exposure had lower ephrin-A3 and Glu levels, but higher Na+-K+ ATPase activity, expression levels of GLAST and GLT-1 than those exposed to 500 μM or 50 mg/kg MnCl2. Moreover, the morphology of cells and the histomorphology of mice striatum were injured. Results showed that pretreatment with fluoxetine and riluzole attenuated the Mn-induced motor dysfunctions. Together, these results suggest that the ephrin-A3/GLAST-GLT-1/Glu signaling pathway participates in Mn-induced excitotoxicity, and fluoxetine and riluzole can mitigate the Mn-induced excitotoxicity in mice brain.
Collapse
Affiliation(s)
- Zhipeng Qi
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Xinxin Yang
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Yanqi Sang
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Yanan Liu
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Jiashuo Li
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Bin Xu
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Wei Liu
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Miao He
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Zhaofa Xu
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Yu Deng
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China.
| | - Jinghai Zhu
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China.
| |
Collapse
|
11
|
Short AK, Baram TZ. Early-life adversity and neurological disease: age-old questions and novel answers. Nat Rev Neurol 2019; 15:657-669. [PMID: 31530940 PMCID: PMC7261498 DOI: 10.1038/s41582-019-0246-5] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2019] [Indexed: 12/24/2022]
Abstract
Neurological illnesses, including cognitive impairment, memory decline and dementia, affect over 50 million people worldwide, imposing a substantial burden on individuals and society. These disorders arise from a combination of genetic, environmental and experiential factors, with the latter two factors having the greatest impact during sensitive periods in development. In this Review, we focus on the contribution of adverse early-life experiences to aberrant brain maturation, which might underlie vulnerability to cognitive brain disorders. Specifically, we draw on recent robust discoveries from diverse disciplines, encompassing human studies and experimental models. These discoveries suggest that early-life adversity, especially in the perinatal period, influences the maturation of brain circuits involved in cognition. Importantly, new findings suggest that fragmented and unpredictable environmental and parental signals comprise a novel potent type of adversity, which contributes to subsequent vulnerabilities to cognitive illnesses via mechanisms involving disordered maturation of brain 'wiring'.
Collapse
Affiliation(s)
- Annabel K Short
- Departments of Anatomy and Neruobiology, University of California-Irvine, Irvine, CA, USA
- Departments of Pediatrics, University of California-Irvine, Irvine, CA, USA
| | - Tallie Z Baram
- Departments of Anatomy and Neruobiology, University of California-Irvine, Irvine, CA, USA.
- Departments of Pediatrics, University of California-Irvine, Irvine, CA, USA.
- Departments of Neurology, University of California-Irvine, Irvine, CA, USA.
| |
Collapse
|
12
|
Cai D, Wei D, Chen S, Chen X, Li S, Chen W, He W. MiR-145 protected the cell viability of human cerebral cortical neurons after oxygen-glucose deprivation by downregulating EPHA4. Life Sci 2019; 231:116517. [PMID: 31150684 DOI: 10.1016/j.lfs.2019.05.073] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 05/23/2019] [Accepted: 05/28/2019] [Indexed: 02/05/2023]
Abstract
Our previous study indicated that microRNA 145 (miR-145) and its predicated target, erythropoietin-producing hepatoma (EPH) receptor A4 (EPHA4), was closely associated with ischemic stroke. In this study, we aimed to further explore their function in a model of oxygen-glucose deprivation (OGD). The expression of miR-145 in the blood of 44 patients with ischemic stroke and 37 normal controls was detected by qRT-PCR. After transfection with either the wild- or mutant-type pGL3-promoter EPHA4 3'UTR into the miR-145 mimic and miR-145 inhibitor, a dual-luciferase reporter assay was performed to explore the interaction between miR-145 and EPHA4. qRT-PCR and Western blot were performed to further explore the effects of miR-145 on EPHA4 expression after an miR-145 mimic, an miR-145 inhibitor or LV-sh-EPHA4 was transfected into cerebral cortical neurons. The expression of miR-145 was significantly upregulated in the blood of patients with ischemic stroke compared to that of normal controls. Dual-luciferase reporter assay, qRT-PCR and Western blot results indicated that miR-145 indeed targets EPHA4 through its 3'-UTR and regulates the expression level of EPHA4 at both the mRNA and protein levels. Moreover, the OGD model was successfully constructed, and miR-145 exerted a protective effects in cell viability in the OGD model by downregulating EPHA4. The expression of LOC105376244 could be regulated by the miR-145-EPHA4 interaction. MiR-145 exerted a protective effects in cell viability in the OGD model by downregulating EPHA4, which suggested their potential roles in ischemic stroke and requires further research.
Collapse
Affiliation(s)
- De Cai
- Department of Pharmacy, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Duncan Wei
- Department of Pharmacy, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Siqia Chen
- Department of Neurology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Xianguang Chen
- Department of Neurology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Shunxian Li
- Department of Neurology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Wenjie Chen
- Department of Neurology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Wenzhen He
- Department of Pharmacy, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China.
| |
Collapse
|
13
|
Recalibrating the Relevance of Adult Neurogenesis. Trends Neurosci 2019; 42:164-178. [PMID: 30686490 DOI: 10.1016/j.tins.2018.12.001] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 11/28/2018] [Accepted: 12/10/2018] [Indexed: 10/27/2022]
Abstract
Conflicting reports about whether adult hippocampal neurogenesis occurs in humans raise questions about its significance for human health and the relevance of animal models. Drawing upon published data, I review species' neurogenesis rates across the lifespan and propose that accelerated neurodevelopmental timing is consistent with lower rates of neurogenesis in adult primates and humans. Nonetheless, protracted neurogenesis may produce populations of neurons that retain plastic properties for long intervals, and have distinct functions depending on when in the lifespan they were born. With some conceptual recalibration we may therefore be able to reconcile seemingly disparate findings and continue to ask how adult neurogenesis, as studied in animals, is relevant for human health.
Collapse
|
14
|
Saha N, Robev D, Mason EO, Himanen JP, Nikolov DB. Therapeutic potential of targeting the Eph/ephrin signaling complex. Int J Biochem Cell Biol 2018; 105:123-133. [PMID: 30343150 DOI: 10.1016/j.biocel.2018.10.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 10/09/2018] [Accepted: 10/16/2018] [Indexed: 12/27/2022]
Abstract
The Eph-ephrin signaling pathway mediates developmental processes and the proper functioning of the adult human body. This distinctive bidirectional signaling pathway includes a canonical downstream signal cascade inside the Eph-bearing cells, as well as a reverse signaling in the ephrin-bearing cells. The signaling is terminated by ADAM metalloproteinase cleavage, internalization, and degradation of the Eph/ephrin complexes. Consequently, the Eph-ephrin-ADAM signaling cascade has emerged as a key target with immense therapeutic potential particularly in the context of cancer. An interesting twist was brought forth by the emergence of ephrins as the entry receptors for the pathological Henipaviruses, which has spurred new studies to target the viral entry. The availability of high-resolution structures of the multi-modular Eph receptors in complexes with ephrins and other binding partners, such as peptides, small molecule inhibitors and antibodies, offers a wealth of information for the structure-guided development of therapeutic intervention. Furthermore, genomic data mining of Eph mutants involved in cancer provides information for targeted drug development. In this review we summarize the distinct avenues for targeting the Eph-ephrin signaling pathway, including its termination by ADAM proteinases. We highlight the latest developments in Eph-related pharmacology in the context of Eph-ephrin-ADAM-based antibodies and small molecules. Finally, the future prospects of genomics- and proteomics-based medicine are discussed.
Collapse
Affiliation(s)
- Nayanendu Saha
- Sloan-Kettering Institute for Cancer Research, Structural Biology Program, 1275 York Avenue, New York, NY 10065, United States
| | - Dorothea Robev
- Sloan-Kettering Institute for Cancer Research, Structural Biology Program, 1275 York Avenue, New York, NY 10065, United States
| | - Emilia O Mason
- Sloan-Kettering Institute for Cancer Research, Structural Biology Program, 1275 York Avenue, New York, NY 10065, United States
| | - Juha P Himanen
- Sloan-Kettering Institute for Cancer Research, Structural Biology Program, 1275 York Avenue, New York, NY 10065, United States.
| | - Dimitar B Nikolov
- Sloan-Kettering Institute for Cancer Research, Structural Biology Program, 1275 York Avenue, New York, NY 10065, United States
| |
Collapse
|
15
|
Kassab R, Alexandre F. Pattern separation in the hippocampus: distinct circuits under different conditions. Brain Struct Funct 2018; 223:2785-2808. [PMID: 29637298 DOI: 10.1007/s00429-018-1659-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 03/26/2018] [Indexed: 10/17/2022]
Abstract
Pattern separation is a fundamental hippocampal process thought to be critical for distinguishing similar episodic memories, and has long been recognized as a natural function of the dentate gyrus (DG), supporting autoassociative learning in CA3. Understanding how neural circuits within the DG-CA3 network mediate this process has received much interest, yet the exact mechanisms behind remain elusive. Here, we argue for the case that sparse coding is necessary but not sufficient to ensure efficient separation and, alternatively, propose a possible interaction of distinct circuits which, nevertheless, act in synergy to produce a unitary function of pattern separation. The proposed circuits involve different functional granule-cell populations, a primary population mediates sparsification and provides recurrent excitation to the other populations which are related to additional pattern separation mechanisms with higher degrees of robustness against interference in CA3. A variety of top-down and bottom-up factors, such as motivation, emotion, and pattern similarity, control the selection of circuitry depending on circumstances. According to this framework, a computational model is implemented and tested against model variants in a series of numerical simulations and biological experiments. The results demonstrate that the model combines fast learning, robust pattern separation and high storage capacity. It also accounts for the controversy around the involvement of the DG during memory recall, explains other puzzling findings, and makes predictions that can inform future investigations.
Collapse
Affiliation(s)
- Randa Kassab
- INRIA, Bordeaux Sud-Ouest, Talence, France. .,Institut des Maladies Neurodégénératives, University of Bordeaux, CNRS UMR 5293-Case 28, Centre Broca Nouvelle-Aquitaine, 146 rue Léo Saignat, 33076, Bordeaux, France. .,LaBRI, UMR 5800, CNRS, Bordeaux INP, University of Bordeaux, Talence, France.
| | - Frédéric Alexandre
- INRIA, Bordeaux Sud-Ouest, Talence, France.,Institut des Maladies Neurodégénératives, University of Bordeaux, CNRS UMR 5293-Case 28, Centre Broca Nouvelle-Aquitaine, 146 rue Léo Saignat, 33076, Bordeaux, France.,LaBRI, UMR 5800, CNRS, Bordeaux INP, University of Bordeaux, Talence, France
| |
Collapse
|
16
|
Khalaf-Nazzal R, Stouffer MA, Olaso R, Muresan L, Roumegous A, Lavilla V, Carpentier W, Moutkine I, Dumont S, Albaud B, Cagnard N, Roest Crollius H, Francis F. Early born neurons are abnormally positioned in the doublecortin knockout hippocampus. Hum Mol Genet 2017; 26:90-108. [PMID: 28007902 DOI: 10.1093/hmg/ddw370] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 10/24/2016] [Indexed: 01/29/2023] Open
Abstract
Human doublecortin (DCX) mutations are associated with severe brain malformations leading to aberrant neuron positioning (heterotopia), intellectual disability and epilepsy. The Dcx protein plays a key role in neuronal migration, and hippocampal pyramidal neurons in Dcx knockout (KO) mice are disorganized. The single CA3 pyramidal cell layer observed in wild type (WT) is present as two abnormal layers in the KO, and CA3 KO pyramidal neurons are more excitable than WT. Dcx KO mice also exhibit spontaneous epileptic activity originating in the hippocampus. It is unknown, however, how hyperexcitability arises and why two CA3 layers are observed.Transcriptome analyses were performed to search for perturbed postnatal gene expression, comparing Dcx KO CA3 pyramidal cell layers with WT. Gene expression changes common to both KO layers indicated mitochondria and Golgi apparatus anomalies, as well as increased cell stress. Intriguingly, gene expression analyses also suggested that the KO layers differ significantly from each other, particularly in terms of maturity. Layer-specific molecular markers and BrdU birthdating to mark the final positions of neurons born at distinct timepoints revealed inverted layering of the CA3 region in Dcx KO animals. Notably, many early-born 'outer boundary' neurons are located in an inner position in the Dcx KO CA3, superficial to other pyramidal neurons. This abnormal positioning likely affects cell morphology and connectivity, influencing network function. Dissecting this Dcx KO phenotype sheds light on coordinated developmental mechanisms of neuronal subpopulations, as well as gene expression patterns contributing to a bi-layered malformation associated with epilepsy.
Collapse
Affiliation(s)
- Reham Khalaf-Nazzal
- INSERM UMR-S 839, Paris.,Sorbonne Universités, Université Pierre et Marie Curie, Paris.,Institut du Fer à Moulin, Paris, France
| | - Melissa A Stouffer
- INSERM UMR-S 839, Paris.,Sorbonne Universités, Université Pierre et Marie Curie, Paris.,Institut du Fer à Moulin, Paris, France
| | - Robert Olaso
- Plateforme de Transcriptomique, Laboratoire de Recherche Translationnelle, CEA/DSV/IG-Centre National de Genotypage, 2 rue Gaston Crémieux, Evry, France
| | - Leila Muresan
- Ecole Normale Supérieure, Institut de Biologie de l'ENS, IBENS, Paris, France.,INSERM, U1024, Paris, France.,CNRS, UMR 8197, Paris, France
| | - Audrey Roumegous
- INSERM UMR-S 839, Paris.,Sorbonne Universités, Université Pierre et Marie Curie, Paris.,Institut du Fer à Moulin, Paris, France
| | - Virginie Lavilla
- Plateforme de Transcriptomique, Laboratoire de Recherche Translationnelle, CEA/DSV/IG-Centre National de Genotypage, 2 rue Gaston Crémieux, Evry, France
| | - Wassila Carpentier
- Plateforme post-génomique de la Pitié-Salpêtrière, Faculty of Medicine, Paris
| | - Imane Moutkine
- INSERM UMR-S 839, Paris.,Sorbonne Universités, Université Pierre et Marie Curie, Paris.,Institut du Fer à Moulin, Paris, France
| | - Sylvie Dumont
- Sorbonne Universités, UPMC Paris 06, UMS30 LUMIC, plateforme d'histomorphologie, St Antoine, Paris
| | - Benoit Albaud
- Plateforme Affymetrix, Institut Curie, Hospital St Louis, Paris
| | - Nicolas Cagnard
- Plateforme Bio-informatique Paris Descartes, Faculté de Necker, 156 rue de Vaugirard, Paris
| | - Hugues Roest Crollius
- Ecole Normale Supérieure, Institut de Biologie de l'ENS, IBENS, Paris, France.,INSERM, U1024, Paris, France.,CNRS, UMR 8197, Paris, France
| | - Fiona Francis
- INSERM UMR-S 839, Paris.,Sorbonne Universités, Université Pierre et Marie Curie, Paris.,Institut du Fer à Moulin, Paris, France
| |
Collapse
|
17
|
Gulisano W, Bizzoca A, Gennarini G, Palmeri A, Puzzo D. Role of the adhesion molecule F3/Contactin in synaptic plasticity and memory. Mol Cell Neurosci 2016; 81:64-71. [PMID: 28038945 DOI: 10.1016/j.mcn.2016.12.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Revised: 12/07/2016] [Accepted: 12/22/2016] [Indexed: 12/14/2022] Open
Abstract
Cell adhesion molecules (CAMs) have a pivotal role in building and maintaining synaptic structures during brain development participating in axonal elongation and pathfinding, glial guidance of neuronal migration, as well as myelination. CAMs expression persists in the adult brain particularly in structures undergoing postnatal neurogenesis and involved in synaptic plasticity and memory as the hippocampus. Among the neural CAMs, we have recently focused on F3/Contactin, a glycosylphosphatidyl inositol-anchored glycoprotein belonging to the immunoglobulin superfamily, involved in neuronal development, synaptic maintenance and organization of neuronal networks. Here, we discuss our recent data suggesting that F3/Contactin exerts a role in hippocampal synaptic plasticity and memory in adult and aged mice. In particular, we have studied long-term potentiation (LTP), spatial and object recognition memory, and phosphorylation of the transcription factor cAMP-Responsive-Element Binding protein (CREB) in a transgenic mouse model of F3/Contactin overexpression. We also investigated whether F3/Contactin might influence neuronal apoptosis and the production of amyloid-beta peptide (Aβ), known to be one of the main pathogenetic hallmarks of Alzheimer's disease (AD). In conclusion, a further understanding of F3/Contactin role in synaptic plasticity and memory might have interesting clinical outcomes in cognitive disorders, such as aging and AD, offering innovative therapeutic opportunities.
Collapse
Affiliation(s)
- Walter Gulisano
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Antonella Bizzoca
- Section of Physiology, Department of Basic Medical Sciences, Neuroscience and Sensory Organs, University of Bari, Bari, Italy
| | - Gianfranco Gennarini
- Section of Physiology, Department of Basic Medical Sciences, Neuroscience and Sensory Organs, University of Bari, Bari, Italy
| | - Agostino Palmeri
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.
| | - Daniela Puzzo
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.
| |
Collapse
|
18
|
Park KA, Ribic A, Laage Gaupp FM, Coman D, Huang Y, Dulla CG, Hyder F, Biederer T. Excitatory Synaptic Drive and Feedforward Inhibition in the Hippocampal CA3 Circuit Are Regulated by SynCAM 1. J Neurosci 2016; 36:7464-75. [PMID: 27413156 PMCID: PMC4945666 DOI: 10.1523/jneurosci.0189-16.2016] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 06/01/2016] [Accepted: 06/02/2016] [Indexed: 01/24/2023] Open
Abstract
UNLABELLED Select adhesion proteins control the development of synapses and modulate their structural and functional properties. Despite these important roles, the extent to which different synapse-organizing mechanisms act across brain regions to establish connectivity and regulate network properties is incompletely understood. Further, their functional roles in different neuronal populations remain to be defined. Here, we applied diffusion tensor imaging (DTI), a modality of magnetic resonance imaging (MRI), to map connectivity changes in knock-out (KO) mice lacking the synaptogenic cell adhesion protein SynCAM 1. This identified reduced fractional anisotropy in the hippocampal CA3 area in absence of SynCAM 1. In agreement, mossy fiber refinement in CA3 was impaired in SynCAM 1 KO mice. Mossy fibers make excitatory inputs onto postsynaptic specializations of CA3 pyramidal neurons termed thorny excrescences and these structures were smaller in the absence of SynCAM 1. However, the most prevalent targets of mossy fibers are GABAergic interneurons and SynCAM 1 loss unexpectedly reduced the number of excitatory terminals onto parvalbumin (PV)-positive interneurons in CA3. SynCAM 1 KO mice additionally exhibited lower postsynaptic GluA1 expression in these PV-positive interneurons. These synaptic imbalances in SynCAM 1 KO mice resulted in CA3 disinhibition, in agreement with reduced feedforward inhibition in this network in the absence of SynCAM 1-dependent excitatory drive onto interneurons. In turn, mice lacking SynCAM 1 were impaired in memory tasks involving CA3. Our results support that SynCAM 1 modulates excitatory mossy fiber inputs onto both interneurons and principal neurons in the hippocampal CA3 area to balance network excitability. SIGNIFICANCE STATEMENT This study advances our understanding of synapse-organizing mechanisms on two levels. First, the data support that synaptogenic proteins guide connectivity and can function in distinct brain regions even if they are expressed broadly. Second, the results demonstrate that a synaptogenic process that controls excitatory inputs to both pyramidal neurons and interneurons can balance excitation and inhibition. Specifically, the study reveals that hippocampal CA3 connectivity is modulated by the synapse-organizing adhesion protein SynCAM 1 and identifies a novel, SynCAM 1-dependent mechanism that controls excitatory inputs onto parvalbumin-positive interneurons. This enables SynCAM 1 to regulate feedforward inhibition and set network excitability. Further, we show that diffusion tensor imaging is sensitive to these cellular refinements affecting neuronal connectivity.
Collapse
Affiliation(s)
- Kellie A Park
- Department of Anesthesiology, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Adema Ribic
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Fabian M Laage Gaupp
- Institute of Clinical Neuroimmunology, Ludwig-Maximilians-University, 80539 Munich, Germany
| | - Daniel Coman
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut 06520, and
| | - Yuegao Huang
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut 06520, and
| | - Chris G Dulla
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Fahmeed Hyder
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut 06520, and Department of Biomedical Engineering, Yale University, School of Engineering and Applied Science, New Haven, Connecticut 06520
| | - Thomas Biederer
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111,
| |
Collapse
|
19
|
Wiera G, Mozrzymas JW. Extracellular proteolysis in structural and functional plasticity of mossy fiber synapses in hippocampus. Front Cell Neurosci 2015; 9:427. [PMID: 26582976 PMCID: PMC4631828 DOI: 10.3389/fncel.2015.00427] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 10/09/2015] [Indexed: 02/04/2023] Open
Abstract
Brain is continuously altered in response to experience and environmental changes. One of the underlying mechanisms is synaptic plasticity, which is manifested by modification of synapse structure and function. It is becoming clear that regulated extracellular proteolysis plays a pivotal role in the structural and functional remodeling of synapses during brain development, learning and memory formation. Clearly, plasticity mechanisms may substantially differ between projections. Mossy fiber synapses onto CA3 pyramidal cells display several unique functional features, including pronounced short-term facilitation, a presynaptically expressed long-term potentiation (LTP) that is independent of NMDAR activation, and NMDA-dependent metaplasticity. Moreover, structural plasticity at mossy fiber synapses ranges from the reorganization of projection topology after hippocampus-dependent learning, through intrinsically different dynamic properties of synaptic boutons to pre- and postsynaptic structural changes accompanying LTP induction. Although concomitant functional and structural plasticity in this pathway strongly suggests a role of extracellular proteolysis, its impact only starts to be investigated in this projection. In the present report, we review the role of extracellular proteolysis in various aspects of synaptic plasticity in hippocampal mossy fiber synapses. A growing body of evidence demonstrates that among perisynaptic proteases, tissue plasminogen activator (tPA)/plasmin system, β-site amyloid precursor protein-cleaving enzyme 1 (BACE1) and metalloproteinases play a crucial role in shaping plastic changes in this projection. We discuss recent advances and emerging hypotheses on the roles of proteases in mechanisms underlying mossy fiber target specific synaptic plasticity and memory formation.
Collapse
Affiliation(s)
- Grzegorz Wiera
- Department of Animal Molecular Physiology, Institute of Experimental Biology, Wroclaw University Wroclaw, Poland ; Laboratory of Neuroscience, Department of Biophysics, Wroclaw Medical University Wroclaw, Poland
| | - Jerzy W Mozrzymas
- Department of Animal Molecular Physiology, Institute of Experimental Biology, Wroclaw University Wroclaw, Poland ; Laboratory of Neuroscience, Department of Biophysics, Wroclaw Medical University Wroclaw, Poland
| |
Collapse
|
20
|
Hass J, Walton E, Wright C, Beyer A, Scholz M, Turner J, Liu J, Smolka MN, Roessner V, Sponheim SR, Gollub RL, Calhoun VD, Ehrlich S. Associations between DNA methylation and schizophrenia-related intermediate phenotypes - a gene set enrichment analysis. Prog Neuropsychopharmacol Biol Psychiatry 2015; 59:31-39. [PMID: 25598502 PMCID: PMC4346504 DOI: 10.1016/j.pnpbp.2015.01.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 01/06/2015] [Accepted: 01/13/2015] [Indexed: 12/18/2022]
Abstract
Multiple genetic approaches have identified microRNAs as key effectors in psychiatric disorders as they post-transcriptionally regulate expression of thousands of target genes. However, their role in specific psychiatric diseases remains poorly understood. In addition, epigenetic mechanisms such as DNA methylation, which affect the expression of both microRNAs and coding genes, are critical for our understanding of molecular mechanisms in schizophrenia. Using clinical, imaging, genetic, and epigenetic data of 103 patients with schizophrenia and 111 healthy controls of the Mind Clinical Imaging Consortium (MCIC) study of schizophrenia, we conducted gene set enrichment analysis to identify markers for schizophrenia-associated intermediate phenotypes. Genes were ranked based on the correlation between DNA methylation patterns and each phenotype, and then searched for enrichment in 221 predicted microRNA target gene sets. We found the predicted hsa-miR-219a-5p target gene set to be significantly enriched for genes (EPHA4, PKNOX1, ESR1, among others) whose methylation status is correlated with hippocampal volume independent of disease status. Our results were strengthened by significant associations between hsa-miR-219a-5p target gene methylation patterns and hippocampus-related neuropsychological variables. IPA pathway analysis of the respective predicted hsa-miR-219a-5p target genes revealed associated network functions in behavior and developmental disorders. Altered methylation patterns of predicted hsa-miR-219a-5p target genes are associated with a structural aberration of the brain that has been proposed as a possible biomarker for schizophrenia. The (dys)regulation of microRNA target genes by epigenetic mechanisms may confer additional risk for developing psychiatric symptoms. Further study is needed to understand possible interactions between microRNAs and epigenetic changes and their impact on risk for brain-based disorders such as schizophrenia.
Collapse
Affiliation(s)
- Johanna Hass
- Translational Developmental Neuroscience Section, Department of Child and Adolescent Psychiatry, Faculty of Medicine of the TU Dresden, Dresden, Germany
| | - Esther Walton
- Translational Developmental Neuroscience Section, Department of Child and Adolescent Psychiatry, Faculty of Medicine of the TU Dresden, Dresden, Germany
| | - Carrie Wright
- Department of Neurosciences, Health Sciences Center, University of New Mexico, Albuquerque, NM, USA,The Mind Research Network, Albuquerque, NM USA
| | - Andreas Beyer
- Cellular Networks and Systems Biology, Biotechnology Center, TU Dresden, Dresden, Germany,University of Cologne, CECAD, Cologne, Germany
| | - Markus Scholz
- Institute for Medical Informatics, Statistics and Epidemiology (IMISE), University of Leipzig, Leipzig, Germany,LIFE (Leipzig Interdisciplinary Research Cluster of Genetic Factors, Phenotypes and Environment), University of Leipzig, Leipzig, Germany
| | - Jessica Turner
- The Mind Research Network, Albuquerque, NM USA,Psychology Department, University of New Mexico, Albuquerque, NM, USA
| | - Jingyu Liu
- The Mind Research Network, Albuquerque, NM USA,Department of Electrical and Computer Engineering, University of New Mexico, Albuquerque, NM USA
| | - Michael N. Smolka
- Department of Psychiatry, Faculty of Medicine of the TU Dresden, Dresden, Germany
| | - Veit Roessner
- Translational Developmental Neuroscience Section, Department of Child and Adolescent Psychiatry, Faculty of Medicine of the TU Dresden, Dresden, Germany
| | - Scott R. Sponheim
- Department of Psychiatry and the Center for magnetic Resonance Research, University of Minnesota, Minneapolis, MN USA
| | - Randy L. Gollub
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA USA,MGH/MIT/HMS Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA USA
| | - Vince D. Calhoun
- The Mind Research Network, Albuquerque, NM USA,Department of Electrical and Computer Engineering, University of New Mexico, Albuquerque, NM USA
| | - Stefan Ehrlich
- Translational Developmental Neuroscience Section, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Dresden, Germany; Department of Psychiatry, Massachusetts General Hospital, Boston, MA USA; MGH/MIT/HMS Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA USA.
| |
Collapse
|
21
|
Pieraut S, Gounko N, Sando R, Dang W, Rebboah E, Panda S, Madisen L, Zeng H, Maximov A. Experience-dependent remodeling of basket cell networks in the dentate gyrus. Neuron 2015; 84:107-122. [PMID: 25277456 DOI: 10.1016/j.neuron.2014.09.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2014] [Indexed: 11/19/2022]
Abstract
The structural organization of neural circuits is strongly influenced by experience, but the underlying mechanisms are incompletely understood. We found that, in the developing dentate gyrus (DG), excitatory drive promotes the somatic innervation of principal granule cells (GCs) by parvalbumin (PV)-positive basket cells. In contrast, presynaptic differentiation of GCs and interneuron subtypes that inhibit GC dendrites is largely resistant to loss of glutamatergic neurotransmission. The networks of PV basket cells in the DG are regulated by vesicular release from projection entorhinal cortical neurons and, at least in part, by NMDA receptors in interneurons. Finally, we present evidence that glutamatergic inputs and NMDA receptors regulate these networks through a presynaptic mechanism that appears to control the branching of interneuron axons. Our results provide insights into how cortical activity tunes the inhibition in a subcortical circuit and reveal new principles of interneuron plasticity.
Collapse
Affiliation(s)
- Simon Pieraut
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, CA 92037, USA; The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Natalia Gounko
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, CA 92037, USA; The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Richard Sando
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, CA 92037, USA; The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA; The Kellogg School of Science and Technology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Westley Dang
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, CA 92037, USA; The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA; The Kellogg School of Science and Technology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Elisabeth Rebboah
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, CA 92037, USA; The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Satchidananda Panda
- Regulatory Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Linda Madisen
- Allen Institute for Brain Science, Seattle, WA 98103, USA
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA 98103, USA
| | - Anton Maximov
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, CA 92037, USA; The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
22
|
Puzzo D, Bizzoca A, Loreto C, Guida CA, Gulisano W, Frasca G, Bellomo M, Castorina S, Gennarini G, Palmeri A. Role of F3/contactin expression profile in synaptic plasticity and memory in aged mice. Neurobiol Aging 2015; 36:1702-1715. [PMID: 25659859 DOI: 10.1016/j.neurobiolaging.2015.01.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 12/30/2014] [Accepted: 01/03/2015] [Indexed: 12/14/2022]
Abstract
We have recently shown that overexpression of the F3/contactin adhesive glycoprotein (also known as Contactin-1) promotes neurogenesis in adult hippocampus, which correlates with improved synaptic plasticity and memory. Because F3/contactin levels physiologically decrease with age, here, we aim at investigating whether its overexpression might counteract the cognitive decline in aged animals. For this we use 20- to 24-month-old TAG/F3 transgenic mice in which F3/contactin overexpression is driven by regulatory sequences from the gene encoding the transient axonal glycoprotein TAG-1 throughout development. We show that aged TAG/F3 mice display improved hippocampal long-term potentiation and memory compared with wild-type littermates. The same mice undergo a decrease of neuronal apoptosis at the hippocampal level, which correlated to a decrease of active caspase-3; by contrast, procaspase-3 and Bax as well as the anti-apoptotic and plasticity-related pathway BDNF/CREB/Bcl-2 were rather increased. Interestingly, amyloid-precursor protein processing was shifted toward sAPPα generation, with a decrease of sAPPβ and amyloid-beta levels. Our data confirm that F3/contactin plays a role in hippocampal synaptic plasticity and memory also in aged mice, suggesting that it acts on molecular pathways related to apoptosis and amyloid-beta production.
Collapse
Affiliation(s)
- Daniela Puzzo
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Antonella Bizzoca
- Section of Physiology, Department of Basic Medical Sciences, Neuroscience and Sensory Organs, University of Bari, Bari, Italy
| | - Carla Loreto
- Section of Anatomy, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Chiara A Guida
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Walter Gulisano
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Giuseppina Frasca
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Maria Bellomo
- Faculty of Psychology and Educational Sciences, University "Kore", Enna, Italy
| | - Sergio Castorina
- Section of Anatomy, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Gianfranco Gennarini
- Section of Physiology, Department of Basic Medical Sciences, Neuroscience and Sensory Organs, University of Bari, Bari, Italy.
| | - Agostino Palmeri
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.
| |
Collapse
|
23
|
Riedl SJ, Pasquale EB. Targeting the Eph System with Peptides and Peptide Conjugates. Curr Drug Targets 2015; 16:1031-47. [PMID: 26212263 PMCID: PMC4861043 DOI: 10.2174/1389450116666150727115934] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 02/02/2015] [Accepted: 07/20/2015] [Indexed: 01/06/2023]
Abstract
Eph receptor tyrosine kinases and ephrin ligands constitute an important cell communication system that controls development, tissue homeostasis and many pathological processes. Various Eph receptors/ephrins are present in essentially all cell types and their expression is often dysregulated by injury and disease. Thus, the 14 Eph receptors are attracting increasing attention as a major class of potential drug targets. In particular, agents that bind to the extracellular ephrin-binding pocket of these receptors show promise for medical applications. This pocket comprises a broad and shallow groove surrounded by several flexible loops, which makes peptides particularly suitable to target it with high affinity and selectivity. Accordingly, a number of peptides that bind to Eph receptors with micromolar affinity have been identified using phage display and other approaches. These peptides are generally antagonists that inhibit ephrin binding and Eph receptor/ ephrin signaling, but some are agonists mimicking ephrin-induced Eph receptor activation. Importantly, some of the peptides are exquisitely selective for single Eph receptors. Most identified peptides are linear, but recently the considerable advantages of cyclic scaffolds have been recognized, particularly in light of potential optimization towards drug leads. To date, peptide improvements have yielded derivatives with low nanomolar Eph receptor binding affinity, high resistance to plasma proteases and/or long in vivo half-life, exemplifying the merits of peptides for Eph receptor targeting. Besides their modulation of Eph receptor/ephrin function, peptides can also serve to deliver conjugated imaging and therapeutic agents or various types of nanoparticles to tumors and other diseased tissues presenting target Eph receptors.
Collapse
Affiliation(s)
| | - Elena B Pasquale
- Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Rd., La Jolla, CA 92037, USA.
| |
Collapse
|
24
|
Lamberto I, Lechtenberg BC, Olson EJ, Mace PD, Dawson PE, Riedl SJ, Pasquale EB. Development and structural analysis of a nanomolar cyclic peptide antagonist for the EphA4 receptor. ACS Chem Biol 2014; 9:2787-95. [PMID: 25268696 PMCID: PMC4273976 DOI: 10.1021/cb500677x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The EphA4 receptor is highly expressed in the nervous system, and recent findings suggest that its signaling activity hinders neural repair and exacerbates certain neurodegenerative processes. EphA4 has also been implicated in cancer progression. Thus, EphA4 inhibitors represent potential therapeutic leads and useful research tools to elucidate the role of EphA4 in physiology and disease. Here, we report the structure of a cyclic peptide antagonist, APY, in complex with the EphA4 ligand-binding domain (LBD), which represents the first structure of a cyclic peptide bound to a receptor tyrosine kinase. The structure shows that the dodecameric APY efficiently occupies the ephrin ligand-binding pocket of EphA4 and promotes a "closed" conformation of the surrounding loops. Structure-guided relaxation of the strained APY β-turn and amidation of the C terminus to allow an additional intrapeptide hydrogen bond yielded APY-βAla8.am, an improved APY derivative that binds to EphA4 with nanomolar affinity. APY-βAla8.am potently inhibits ephrin-induced EphA4 activation in cells and EphA4-dependent neuronal growth cone collapse, while retaining high selectivity for EphA4. The two crystal structures of APY and APY-βAla8.am bound to EphA4, in conjunction with secondary phage display screens, highlighted peptide residues that are essential for EphA4 binding as well as residues that can be modified. Thus, the APY scaffold represents an exciting prototype, particularly since cyclic peptides have potentially favorable metabolic stability and are emerging as an important class of molecules for disruption of protein-protein interactions.
Collapse
Affiliation(s)
- Ilaria Lamberto
- Cancer Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Bernhard C. Lechtenberg
- Cancer Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Erika J. Olson
- Department
of Chemistry and Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Peter D. Mace
- Cancer Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Philip E. Dawson
- Department
of Chemistry and Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Stefan J. Riedl
- Cancer Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Elena B. Pasquale
- Cancer Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, California 92037, United States
- Pathology
Department, University of California San Diego, La Jolla, California 92093, United States
| |
Collapse
|
25
|
Optic flow instructs retinotopic map formation through a spatial to temporal to spatial transformation of visual information. Proc Natl Acad Sci U S A 2014; 111:E5105-13. [PMID: 25385606 DOI: 10.1073/pnas.1416953111] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Retinotopic maps are plastic in response to changes in sensory input; however, the experience-dependent instructive cues that organize retinotopy are unclear. In animals with forward-directed locomotion, the predominant anterior to posterior optic flow activates retinal ganglion cells in a stereotyped temporal to nasal sequence. Here we imaged retinotectal axon arbor location and structural plasticity to assess map refinement in vivo while exposing Xenopus tadpoles to visual stimuli. We show that the temporal sequence of retinal activity driven by natural optic flow organizes retinotopy by regulating axon arbor branch dynamics, whereas the opposite sequence of retinal activity prevents map refinement. Our study demonstrates that a spatial to temporal to spatial transformation of visual information controls experience-dependent topographic map plasticity. This organizational principle is likely to apply to other sensory modalities and projections in the brain.
Collapse
|
26
|
Frotscher M, Studer D, Graber W, Chai X, Nestel S, Zhao S. Fine structure of synapses on dendritic spines. Front Neuroanat 2014; 8:94. [PMID: 25249945 PMCID: PMC4158982 DOI: 10.3389/fnana.2014.00094] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2014] [Accepted: 08/24/2014] [Indexed: 12/16/2022] Open
Abstract
Camillo Golgi's "Reazione Nera" led to the discovery of dendritic spines, small appendages originating from dendritic shafts. With the advent of electron microscopy (EM) they were identified as sites of synaptic contact. Later it was found that changes in synaptic strength were associated with changes in the shape of dendritic spines. While live-cell imaging was advantageous in monitoring the time course of such changes in spine structure, EM is still the best method for the simultaneous visualization of all cellular components, including actual synaptic contacts, at high resolution. Immunogold labeling for EM reveals the precise localization of molecules in relation to synaptic structures. Previous EM studies of spines and synapses were performed in tissue subjected to aldehyde fixation and dehydration in ethanol, which is associated with protein denaturation and tissue shrinkage. It has remained an issue to what extent fine structural details are preserved when subjecting the tissue to these procedures. In the present review, we report recent studies on the fine structure of spines and synapses using high-pressure freezing (HPF), which avoids protein denaturation by aldehydes and results in an excellent preservation of ultrastructural detail. In these studies, HPF was used to monitor subtle fine-structural changes in spine shape associated with chemically induced long-term potentiation (cLTP) at identified hippocampal mossy fiber synapses. Changes in spine shape result from reorganization of the actin cytoskeleton. We report that cLTP was associated with decreased immunogold labeling for phosphorylated cofilin (p-cofilin), an actin-depolymerizing protein. Phosphorylation of cofilin renders it unable to depolymerize F-actin, which stabilizes the actin cytoskeleton. Decreased levels of p-cofilin, in turn, suggest increased actin turnover, possibly underlying the changes in spine shape associated with cLTP. The findings reviewed here establish HPF as an appropriate method for studying the fine structure and molecular composition of synapses on dendritic spines.
Collapse
Affiliation(s)
- Michael Frotscher
- Institute for Structural Neurobiology, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf Hamburg Germany
| | - Daniel Studer
- Institute of Anatomy, University of Bern Bern Switzerland
| | - Werner Graber
- Institute of Anatomy, University of Bern Bern Switzerland
| | - Xuejun Chai
- Institute for Structural Neurobiology, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf Hamburg Germany
| | - Sigrun Nestel
- Institute of Anatomy and Cell Biology, University of Freiburg Freiburg Germany
| | - Shanting Zhao
- Institute for Structural Neurobiology, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf Hamburg Germany
| |
Collapse
|
27
|
Yang J, Luo X, Huang X, Ning Q, Xie M, Wang W. Ephrin-A3 reverse signaling regulates hippocampal neuronal damage and astrocytic glutamate transport after transient global ischemia. J Neurochem 2014; 131:383-94. [PMID: 25040798 DOI: 10.1111/jnc.12819] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 07/01/2014] [Accepted: 07/03/2014] [Indexed: 12/01/2022]
Abstract
Increasing evidence indicates that the Eph receptors and their ephrin ligands are involved in the regulation of interactions between neurons and astrocytes. Moreover, astrocytic ephrin-A3 reverse signaling mediated by EphA4 receptors is necessary for controlling the abundance of glial glutamate transporters. However, the role of ephrin-A3 reverse signaling in astrocytic function and neuronal death under ischemic conditions remains unclear. In the present study, we found that the EphA4 receptor and its ephrin-A3 ligand, which were distributed in neurons and astrocytes, respectively, in the hippocampus showed a coincident up-regulation of protein expression in the early stage of ischemia. Application of clustered EphA4 decreased the expressions of astrocytic glutamate transporters together with astrocytic glutamate uptake capacity through activating ephrin-A3 reverse signaling. In consequence, neuronal loss was aggravated in the CA1 region of the hippocampus accompanied by impaired hippocampus-dependent spatial memory when clustered EphA4 treatment was administered prior to transient global ischemia. These findings indicate that EphA4-mediated ephrin-A3 reverse signaling is a crucial mechanism for astrocytes to control glial glutamate transporters and prevent glutamate excitotoxicity under pathological conditions. Astrocytic ephrin-A3 reverse signaling mediated by EphA4 receptor is necessary for controlling the abundance of glial glutamate transporters under physiological conditions. However, the role of ephrin-A3 reverse signaling in astrocytic function and neuronal death under ischemic conditions remains unclear. We found EphA4-mediated ephrin-A3 reverse signaling to be a crucial mechanism for astrocytes to control glial glutamate transporters and protect hippocampal neurons from glutamate excitotoxicity under ischemic conditions, this cascade representing a potential therapeutic target for stroke.
Collapse
Affiliation(s)
- Jinshan Yang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | | | | | | | |
Collapse
|
28
|
Affiliation(s)
- Bethany Powell Gray
- Department of Internal Medicine and The Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390-8807, United States
| | - Kathlynn C. Brown
- Department of Internal Medicine and The Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390-8807, United States
| |
Collapse
|
29
|
Mizumoto K, Shen K. Two Wnts instruct topographic synaptic innervation in C. elegans. Cell Rep 2013; 5:389-96. [PMID: 24139806 DOI: 10.1016/j.celrep.2013.09.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Revised: 07/18/2013] [Accepted: 09/09/2013] [Indexed: 11/26/2022] Open
Abstract
Gradients of topographic cues play essential roles in the organization of sensory systems by guiding axonal growth cones. Little is known about whether there are additional mechanisms for precise topographic mapping of synaptic connections. Whereas the C. elegans DA8 and DA9 neurons have similar axonal trajectories, their synapses are positioned in distinct but adjacent domains in the anterior-posterior axis. We found that two Wnts, LIN-44 and EGL-20, are responsible for this spatial organization of synapses. Both Wnts form putative posterior-high, anterior-low gradients. The posteriorly expressed LIN-44 inhibits synapse formation in both DA9 and DA8, and creates a synapse-free domain on both axons via LIN-17 /Frizzled. EGL-20, a more anteriorly expressed Wnt, inhibits synapse formation through MIG-1/Frizzled, which is expressed in DA8 but not in DA9. The Wnt-Frizzled specificity and selective Frizzled expression dictate the stereotyped, topographic positioning of synapses between these two neurons.
Collapse
Affiliation(s)
- Kota Mizumoto
- Howard Hughes Medical Institute, Department of Biology, Stanford University, 385 Serra Mall, Stanford, CA 94305, USA
| | | |
Collapse
|
30
|
Proenca CC, Stoehr N, Bernhard M, Seger S, Genoud C, Roscic A, Paganetti P, Liu S, Murphy LO, Kuhn R, Bouwmeester T, Galimberti I. Atg4b-dependent autophagic flux alleviates Huntington's disease progression. PLoS One 2013; 8:e68357. [PMID: 23861892 PMCID: PMC3704647 DOI: 10.1371/journal.pone.0068357] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Accepted: 05/29/2013] [Indexed: 01/24/2023] Open
Abstract
The accumulation of aggregated mutant huntingtin (mHtt) inclusion bodies is involved in Huntigton’s disease (HD) progression. Medium sized-spiny neurons (MSNs) in the corpus striatum are highly vulnerable to mHtt aggregate accumulation and degeneration, but the mechanisms and pathways involved remain elusive. Here we have developed a new model to study MSNs degeneration in the context of HD. We produced organotypic cortico-striatal slice cultures (CStS) from HD transgenic mice mimicking specific features of HD progression. We then show that induction of autophagy using catalytic inhibitors of mTOR prevents MSNs degeneration in HD CStS. Furthermore, disrupting autophagic flux by overexpressing Atg4b in neurons and slice cultures, accelerated mHtt aggregation and neuronal death, suggesting that Atg4b-dependent autophagic flux influences HD progression. Under these circumstances induction of autophagy using catalytic inhibitors of mTOR was inefficient and did not affect mHtt aggregate accumulation and toxicity, indicating that mTOR inhibition alleviates HD progression by inducing Atg4b-dependent autophagic flux. These results establish modulators of Atg4b-dependent autophagic flux as new potential targets in the treatment of HD.
Collapse
Affiliation(s)
- Catia C. Proenca
- Developmental and Molecular Pathways, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Natacha Stoehr
- Developmental and Molecular Pathways, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Mario Bernhard
- Developmental and Molecular Pathways, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | | | | | | | | | - Shanming Liu
- Developmental and Molecular Pathways, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Leon O. Murphy
- Developmental and Molecular Pathways, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | | | - Tewis Bouwmeester
- Developmental and Molecular Pathways, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Ivan Galimberti
- Developmental and Molecular Pathways, Novartis Institutes for Biomedical Research, Basel, Switzerland
- * E-mail:
| |
Collapse
|
31
|
Khalaf-Nazzal R, Francis F. Hippocampal development - old and new findings. Neuroscience 2013; 248:225-42. [PMID: 23756184 DOI: 10.1016/j.neuroscience.2013.05.061] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Revised: 05/14/2013] [Accepted: 05/31/2013] [Indexed: 01/26/2023]
Abstract
The hippocampus, derived from medial regions of the telencephalon, constitutes a remarkable brain structure. It is part of the limbic system, and it plays important roles in information encoding, related to short-term and long-term memory, and spatial navigation. It has also attracted the attention of many clinicians and neuroscientists for its involvement in a wide spectrum of pathological conditions, including epilepsy, intellectual disability, Alzheimer disease and others. Here we address the topic of hippocampal development. As well as original landmark findings, modern techniques such as large-scale in situ hybridizations, in utero electroporation and the study of mouse mutants with hippocampal phenotypes, add further detail to our knowledge of the finely regulated processes which form this intricate structure. Molecular signatures are being revealed related to field, intra-field and laminar cell identity, as well as, cell compartments expressing surface proteins instrumental for connectivity. We summarize here old and new findings, and highlight elegant tools used to fine-study hippocampal development.
Collapse
Affiliation(s)
- R Khalaf-Nazzal
- INSERM, UMR-S 839, Paris 75005, France; Université Pierre et Marie Curie, Paris 75005, France; Institut du Fer à Moulin, Paris 75005, France
| | - F Francis
- INSERM, UMR-S 839, Paris 75005, France; Université Pierre et Marie Curie, Paris 75005, France; Institut du Fer à Moulin, Paris 75005, France.
| |
Collapse
|
32
|
Cang J, Feldheim DA. Developmental mechanisms of topographic map formation and alignment. Annu Rev Neurosci 2013; 36:51-77. [PMID: 23642132 DOI: 10.1146/annurev-neuro-062012-170341] [Citation(s) in RCA: 177] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Brain connections are organized into topographic maps that are precisely aligned both within and across modalities. This alignment facilitates coherent integration of different categories of sensory inputs and allows for proper sensorimotor transformations. Topographic maps are established and aligned by multistep processes during development, including interactions of molecular guidance cues expressed in gradients; spontaneous activity-dependent axonal and dendritic remodeling; and sensory-evoked plasticity driven by experience. By focusing on the superior colliculus, a major site of topographic map alignment for different sensory modalities, this review summarizes current understanding of topographic map development in the mammalian visual system and highlights recent advances in map alignment studies. A major goal looking forward is to reveal the molecular and synaptic mechanisms underlying map alignment and to understand the physiological and behavioral consequences when these mechanisms are disrupted at various scales.
Collapse
Affiliation(s)
- Jianhua Cang
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA.
| | | |
Collapse
|
33
|
Abstract
Eph receptors and their membrane-tethered ligands have important functions in development. Trans interactions of Eph receptors with ephrins at cell-cell interfaces promote a variety of cellular responses, including repulsion, attraction and migration. Eph-ephrin signalling can be bi-directional and controls actin cytoskeleton dynamics, thereby leading to changes in cellular shape. This article provides an overview of the general structures and signalling mechanisms, and of typical developmental functions along with cell biological principles.
Collapse
Affiliation(s)
- Rüdiger Klein
- Max-Planck Institute of Neurobiology, Department of Molecular Neurobiology, Am Klopferspitz 18, Munich-Martinsried, Germany.
| |
Collapse
|
34
|
Zhao S, Studer D, Chai X, Graber W, Brose N, Nestel S, Young C, Rodriguez EP, Saetzler K, Frotscher M. Structural plasticity of spines at giant mossy fiber synapses. Front Neural Circuits 2012; 6:103. [PMID: 23264762 PMCID: PMC3524460 DOI: 10.3389/fncir.2012.00103] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Accepted: 11/22/2012] [Indexed: 11/13/2022] Open
Abstract
The granule cells of the dentate gyrus give rise to thin unmyelinated axons, the mossy fibers. They form giant presynaptic boutons impinging on large complex spines on the proximal dendritic portions of hilar mossy cells and CA3 pyramidal neurons. While these anatomical characteristics have been known for some time, it remained unclear whether functional changes at mossy fiber synapses such as long-term potentiation (LTP) are associated with structural changes. Since subtle structural changes may escape a fine-structural analysis when the tissue is fixed by using aldehydes and is dehydrated in ethanol, rapid high-pressure freezing (HPF) of the tissue was applied. Slice cultures of hippocampus were prepared and incubated in vitro for 2 weeks. Then, chemical LTP (cLTP) was induced by the application of 25 mM tetraethylammonium (TEA) for 10 min. Whole-cell patch-clamp recordings from CA3 pyramidal neurons revealed a highly significant potentiation of mossy fiber synapses when compared to control conditions before the application of TEA. Next, the slice cultures were subjected to HPF, cryosubstitution, and embedding in Epon for a fine-structural analysis. When compared to control tissue, we noticed a significant decrease of synaptic vesicles in mossy fiber boutons and a concomitant increase in the length of the presynaptic membrane. On the postsynaptic side, we observed the formation of small, finger-like protrusions, emanating from the large complex spines. These short protrusions gave rise to active zones that were shorter than those normally found on the thorny excrescences. However, the total number of active zones was significantly increased. Of note, none of these cLTP-induced structural changes was observed in slice cultures from Munc13-1 deficient mouse mutants showing severely impaired vesicle priming and docking. In conclusion, application of HPF allowed us to monitor cLTP-induced structural reorganization of mossy fiber synapses.
Collapse
Affiliation(s)
- Shanting Zhao
- Center for Molecular Neurobiology Hamburg, Institute for Structural Neurobiology Hamburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Zhao S, Studer D, Chai X, Graber W, Brose N, Nestel S, Young C, Rodriguez EP, Saetzler K, Frotscher M. Structural plasticity of hippocampal mossy fiber synapses as revealed by high-pressure freezing. J Comp Neurol 2012; 520:2340-51. [PMID: 22237743 DOI: 10.1002/cne.23040] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Despite recent progress in fluorescence microscopy techniques, electron microscopy (EM) is still superior in the simultaneous analysis of all tissue components at high resolution. However, it is unclear to what extent conventional fixation for EM using aldehydes results in tissue alteration. Here we made an attempt to minimize tissue alteration by using rapid high-pressure freezing (HPF) of hippocampal slice cultures. We used this approach to monitor fine-structural changes at hippocampal mossy fiber synapses associated with chemically induced long-term potentiation (LTP). Synaptic plasticity in LTP has been known to involve structural changes at synapses including reorganization of the actin cytoskeleton and de novo formation of spines. While LTP-induced formation and growth of postsynaptic spines have been reported, little is known about associated structural changes in presynaptic boutons. Mossy fiber synapses are assumed to exhibit presynaptic LTP expression and are easily identified by EM. In slice cultures from wildtype mice, we found that chemical LTP increased the length of the presynaptic membrane of mossy fiber boutons, associated with a de novo formation of small spines and an increase in the number of active zones. Of note, these changes were not observed in slice cultures from Munc13-1 knockout mutants exhibiting defective vesicle priming. These findings show that activation of hippocampal mossy fibers induces pre- and postsynaptic structural changes at mossy fiber synapses that can be monitored by EM.
Collapse
Affiliation(s)
- Shanting Zhao
- Department for Structural Neurobiology, Center for Molecular Neurobiology Hamburg-ZMNH, University of Hamburg, D-20246 Hamburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Hitt B, Riordan SM, Kukreja L, Eimer WA, Rajapaksha TW, Vassar R. β-Site amyloid precursor protein (APP)-cleaving enzyme 1 (BACE1)-deficient mice exhibit a close homolog of L1 (CHL1) loss-of-function phenotype involving axon guidance defects. J Biol Chem 2012; 287:38408-25. [PMID: 22988240 DOI: 10.1074/jbc.m112.415505] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACE1 is the β-secretase enzyme that initiates production of the β-amyloid peptide involved in Alzheimer disease. However, little is known about the functions of BACE1. BACE1-deficient mice exhibit mild but complex neurological phenotypes suggesting therapeutic BACE1 inhibition may not be completely free of mechanism-based side effects. Recently, we have reported that BACE1 null mice have axon guidance defects in olfactory sensory neuron projections to glomeruli in the olfactory bulb. Here, we show that BACE1 deficiency also causes an axon guidance defect in the hippocampus, a shortened and disorganized infrapyramidal bundle of the mossy fiber projection from the dentate gyrus to CA3. Although we observed that a classical axon guidance molecule, EphA4, was cleaved by BACE1 when co-expressed with BACE1 in HEK293 cells, we could find no evidence of BACE1 processing of EphA4 in the brain. Remarkably, we discovered that the axon guidance defects of BACE1(-/-) mice were strikingly similar to those of mice deficient in a recently identified BACE1 substrate, the neural cell adhesion molecule close homolog of L1 (CHL1) that is involved in neurite outgrowth. CHL1 undergoes BACE1-dependent processing in BACE1(+/+), but not BACE1(-/-), hippocampus, and olfactory bulb, indicating that CHL1 is a BACE1 substrate in vivo. Finally, BACE1 and CHL1 co-localize in the terminals of hippocampal mossy fibers, olfactory sensory neuron axons, and growth cones of primary hippocampal neurons. We conclude that BACE1(-/-) axon guidance defects are likely the result of abrogated BACE1 processing of CHL1 and that BACE1 deficiency produces a CHL1 loss-of-function phenotype. Our results imply the possibility that axon mis-targeting may occur in adult neurogenic and/or regenerating neurons as a result of chronic BACE1 inhibition and add a note of caution to BACE1 inhibitor development.
Collapse
Affiliation(s)
- Brian Hitt
- Department of Cell and Molecular Biology, The Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | | | | | | | | | | |
Collapse
|
37
|
Distinctive binding of three antagonistic peptides to the ephrin-binding pocket of the EphA4 receptor. Biochem J 2012; 445:47-56. [PMID: 22489865 DOI: 10.1042/bj20120408] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The EphA4 receptor tyrosine kinase interacts with ephrin ligands to regulate many processes, ranging from axon guidance and nerve regeneration to cancer malignancy. Thus antagonists that inhibit ephrin binding to EphA4 could be useful for a variety of research and therapeutic applications. In the present study we characterize the binding features of three antagonistic peptides (KYL, APY and VTM) that selectively target EphA4 among the Eph receptors. Isothermal titration calorimetry analysis demonstrated that all three peptides bind to the ephrin-binding domain of EphA4 with low micromolar affinity. Furthermore, the effects of a series of EphA4 mutations suggest that the peptides interact in different ways with the ephrin-binding pocket of EphA4. Chemical-shift changes observed by NMR spectroscopy upon binding of the KYL peptide involve many EphA4 residues, consistent with extensive interactions and possibly receptor conformational changes. Additionally, systematic replacement of each of the 12 amino acids of KYL and VTM identify the residues critical for EphA4, binding. The peptides exhibit a long half-life in cell culture medium which, with their substantial binding affinity and selectivity for EphA4, makes them excellent research tools to modulate EphA4 function.
Collapse
|
38
|
Alignment of multimodal sensory input in the superior colliculus through a gradient-matching mechanism. J Neurosci 2012; 32:5264-71. [PMID: 22496572 DOI: 10.1523/jneurosci.0240-12.2012] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The superior colliculus (SC) is a midbrain structure that integrates visual, somatosensory, and auditory inputs to direct head and eye movements. Each of these modalities is topographically mapped and aligned with the others to ensure precise behavioral responses to multimodal stimuli. While it is clear that neural activity is instructive for topographic alignment of inputs from the visual cortex (V1) and auditory system with retinal axons in the SC, there is also evidence that activity-independent mechanisms are used to establish topographic alignment between modalities. Here, we show that the topography of the projection from primary somatosensory cortex (S1) to the SC is established during the first postnatal week. Unlike V1-SC projections, the S1-SC projection does not bifurcate when confronted with a duplicated retinocollicular map, showing that retinal input in the SC does not influence the topography of the S1-SC projection. However, S1-SC topography is disrupted in mice lacking ephrin-As, which we find are expressed in graded patterns along with their binding partners, the EphA4 and EphA7, in both S1 and the somatosensory recipient layer of the SC. Together, these data support a model in which somatosensory inputs into the SC map topographically and establish alignment with visual inputs in the SC using a gradient-matching mechanism.
Collapse
|
39
|
Dieni S, Matsumoto T, Dekkers M, Rauskolb S, Ionescu MS, Deogracias R, Gundelfinger ED, Kojima M, Nestel S, Frotscher M, Barde YA. BDNF and its pro-peptide are stored in presynaptic dense core vesicles in brain neurons. ACTA ACUST UNITED AC 2012; 196:775-88. [PMID: 22412021 PMCID: PMC3308691 DOI: 10.1083/jcb.201201038] [Citation(s) in RCA: 263] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Contrasting with the long-established retrograde model for neurotrophin function, specific immunohistochemical localization of brain-derived neurotrophic factor in the central nervous system supports the alternative model of presynaptic localization and anterograde function. Although brain-derived neurotrophic factor (BDNF) regulates numerous and complex biological processes including memory retention, its extremely low levels in the mature central nervous system have greatly complicated attempts to reliably localize it. Using rigorous specificity controls, we found that antibodies reacting either with BDNF or its pro-peptide both stained large dense core vesicles in excitatory presynaptic terminals of the adult mouse hippocampus. Both moieties were ∼10-fold more abundant than pro-BDNF. The lack of postsynaptic localization was confirmed in Bassoon mutants, a seizure-prone mouse line exhibiting markedly elevated levels of BDNF. These findings challenge previous conclusions based on work with cultured neurons, which suggested activity-dependent dendritic synthesis and release of BDNF. They instead provide an ultrastructural basis for an anterograde mode of action of BDNF, contrasting with the long-established retrograde model derived from experiments with nerve growth factor in the peripheral nervous system.
Collapse
Affiliation(s)
- Sandra Dieni
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Brennaman LH, Zhang X, Guan H, Triplett JW, Brown A, Demyanenko GP, Manis PB, Landmesser L, Maness PF. Polysialylated NCAM and ephrinA/EphA regulate synaptic development of GABAergic interneurons in prefrontal cortex. ACTA ACUST UNITED AC 2012; 23:162-77. [PMID: 22275477 DOI: 10.1093/cercor/bhr392] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A novel function for the neural cell adhesion molecule (NCAM) was identified in ephrinA/EphA-mediated repulsion as an important regulatory mechanism for development of GABAergic inhibitory synaptic connections in mouse prefrontal cortex. Deletion of NCAM, EphA3, or ephrinA2/3/5 in null mutant mice increased the numbers and size of perisomatic synapses between GABAergic basket interneurons and pyramidal cells in the developing cingulate cortex (layers II/III). A functional consequence of NCAM loss was increased amplitudes and faster kinetics of miniature inhibitory postsynaptic currents in NCAM null cingulate cortex. NCAM and EphA3 formed a molecular complex and colocalized with the inhibitory presynaptic marker vesicular GABA transporter (VGAT) in perisomatic puncta and neuropil in the cingulate cortex. EphrinA5 treatment promoted axon remodeling of enhanced green fluorescent protein-labeled basket interneurons in cortical slice cultures and induced growth cone collapse in wild-type but not NCAM null mutant neurons. NCAM modified with polysialic acid (PSA) was required to promote ephrinA5-induced axon remodeling of basket interneurons in cortical slices, likely by providing a permissive environment for ephrinA5/EphA3 signaling. These results reveal a new mechanism in which NCAM and ephrinAs/EphA3 coordinate to constrain GABAergic interneuronal arborization and perisomatic innervation, potentially contributing to excitatory/inhibitory balance in prefrontal cortical circuitry.
Collapse
Affiliation(s)
- Leann H Brennaman
- Department of Biochemistry and Biophysics, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
McGonigal R, Tabatadze N, Routtenberg A. Selective presynaptic terminal remodeling induced by spatial, but not cued, learning: a quantitative confocal study. Hippocampus 2011; 22:1242-55. [PMID: 22180136 DOI: 10.1002/hipo.20998] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/03/2011] [Indexed: 01/09/2023]
Abstract
The hippocampal mossy fibers (MFs) are capable of behaviorally selective, use-dependent structural remodeling. Indeed, we previously observed a new layer of Timm's staining induced in the stratum oriens (SO) in CA3 after spatial but not cued water maze learning (Rekart et al., (2007) Learn Mem 14:416-421). This led to the prediction that there is a learning-specific induction of presynaptic terminal plasticity of MF axons. This study confirms this prediction demonstrating, at the confocal level of analysis, terminal-specific, and behavior-selective presynaptic structural plasticity linked to long-term memory. Male adult Wistar rats were trained for 5 days to locate a hidden or visible platform in a water maze and a retention test was performed 7 days later. MF terminal subtypes, specifically identified by an antibody to zinc transporter 3 (ZnT3), were counted from confocal z-stacks in the stratum lucidum (SL) and the SO. In hidden platform trained rats, there was a significant increase in the number of large MF terminals (LMTs, 2.5-10 μm diameter, >2 μm(2) area) compared to controls both in the proximal SL (P < 0.05) and in the SO (P < 0.01). Surprisingly, there was no detectable increase in small MF terminals (SMTs, 0.5-2 μm diameter, <2 μm(2) area) in either SL or SO as a consequence of training. This distinction of the two MF terminal types is functionally important as LMTs synapse on CA3 pyramidal neurons, while SMTs are known to target inhibitory interneurons. The present findings highlight the pivotal role in memory of presynaptic structural plasticity. Because the "sprouting" observed is specific to the LMT, with no detectable change in the number of the SMT, learning may enhance net excitatory input to CA3 pyramidal neurons. Given the sparse coding of the MF-CA3 connection, and the role that granule cells play in pattern separation, the remodeling observed here may be expected to have a major impact on the long-term integration of spatial context into memory.
Collapse
Affiliation(s)
- R McGonigal
- Department of Psychology, Northwestern University, 2029 Sheridan Rd., Evanston, IL 60208, USA
| | | | | |
Collapse
|
42
|
Noberini R, Lamberto I, Pasquale EB. Targeting Eph receptors with peptides and small molecules: progress and challenges. Semin Cell Dev Biol 2011; 23:51-7. [PMID: 22044885 DOI: 10.1016/j.semcdb.2011.10.023] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Accepted: 10/17/2011] [Indexed: 11/18/2022]
Abstract
The Eph receptors are a large family of receptor tyrosine kinases. Their kinase activity and downstream signaling ability are stimulated by the binding of cell surface-associated ligands, the ephrins. The ensuing signals are bidirectional because the ephrins can also transduce signals (known as reverse signals) following their interaction with Eph receptors. The ephrin-binding pocket in the extracellular N-terminal domain of the Eph receptors and the ATP-binding pocket in the intracellular kinase domain represent potential binding sites for peptides and small molecules. Indeed, a number of peptides and chemical compounds that target Eph receptors and inhibit ephrin binding or kinase activity have been identified. These molecules show promise as probes to study Eph receptor/ephrin biology, as lead compounds for drug development, and as targeting agents to deliver drugs or imaging agents to tumors. Current challenges are to find (1) small molecules that inhibit Eph receptor-ephrin interactions with high binding affinity and good lead-like properties and (2) selective kinase inhibitors that preferentially target the Eph receptor family or subsets of Eph receptors. Strategies that could also be explored include targeting additional Eph receptor interfaces and the ephrin ligands.
Collapse
Affiliation(s)
- Roberta Noberini
- Sanford-Burnham Medical Research Institute, 10901 N. Torrey Pines Rd., La Jolla, CA 92037, USA
| | | | | |
Collapse
|
43
|
Eph and ephrin signaling in the formation of topographic maps. Semin Cell Dev Biol 2011; 23:7-15. [PMID: 22044886 DOI: 10.1016/j.semcdb.2011.10.026] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2011] [Accepted: 10/17/2011] [Indexed: 11/20/2022]
Abstract
The axonal connections between the retina and its midbrain target, the superior colliculus (SC), is mapped topographically, such that the spatial relationships of cell bodies in the retina are maintained when terminating in the SC. Topographic map development uses a Cartesian mapping system such that each axis of the retina is mapped independently. Along the nasal-temporal mapping axis, EphAs and ephrin-As, are graded molecular cues required for topographic mapping while the dorsal-ventral axis is mapped in part via EphB and ephrin-Bs. Because both Ephs and ephrins are cell surface molecules they can signal in the forward and reverse directions. Eph/ephrin signaling leads to changes in cytoskeletal dynamics that lead to actin depolymerization and endocytosis guiding axons via attraction and repulsion.
Collapse
|
44
|
Eph receptors and ephrins in neuron-astrocyte communication at synapses. Glia 2011; 59:1567-78. [DOI: 10.1002/glia.21226] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2011] [Accepted: 07/05/2011] [Indexed: 12/24/2022]
|
45
|
Learning-related feedforward inhibitory connectivity growth required for memory precision. Nature 2011; 473:514-8. [PMID: 21532590 DOI: 10.1038/nature09946] [Citation(s) in RCA: 218] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2010] [Accepted: 02/17/2011] [Indexed: 01/10/2023]
|
46
|
Abstract
Axons are generally considered as reliable transmission cables in which stable propagation occurs once an action potential is generated. Axon dysfunction occupies a central position in many inherited and acquired neurological disorders that affect both peripheral and central neurons. Recent findings suggest that the functional and computational repertoire of the axon is much richer than traditionally thought. Beyond classical axonal propagation, intrinsic voltage-gated ionic currents together with the geometrical properties of the axon determine several complex operations that not only control signal processing in brain circuits but also neuronal timing and synaptic efficacy. Recent evidence for the implication of these forms of axonal computation in the short-term dynamics of neuronal communication is discussed. Finally, we review how neuronal activity regulates both axon morphology and axonal function on a long-term time scale during development and adulthood.
Collapse
Affiliation(s)
- Dominique Debanne
- Institut National de la Santé et de la Recherche Médicale U.641 and Université de la Méditerranée, Faculté de Médecine Secteur Nord, Marseille, France
| | - Emilie Campanac
- Institut National de la Santé et de la Recherche Médicale U.641 and Université de la Méditerranée, Faculté de Médecine Secteur Nord, Marseille, France
| | - Andrzej Bialowas
- Institut National de la Santé et de la Recherche Médicale U.641 and Université de la Méditerranée, Faculté de Médecine Secteur Nord, Marseille, France
| | - Edmond Carlier
- Institut National de la Santé et de la Recherche Médicale U.641 and Université de la Méditerranée, Faculté de Médecine Secteur Nord, Marseille, France
| | - Gisèle Alcaraz
- Institut National de la Santé et de la Recherche Médicale U.641 and Université de la Méditerranée, Faculté de Médecine Secteur Nord, Marseille, France
| |
Collapse
|
47
|
Bednarek E, Caroni P. β-Adducin Is Required for Stable Assembly of New Synapses and Improved Memory upon Environmental Enrichment. Neuron 2011; 69:1132-46. [DOI: 10.1016/j.neuron.2011.02.034] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/17/2011] [Indexed: 12/15/2022]
|
48
|
Temporally matched subpopulations of selectively interconnected principal neurons in the hippocampus. Nat Neurosci 2011; 14:495-504. [PMID: 21358645 DOI: 10.1038/nn.2768] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2010] [Accepted: 01/18/2011] [Indexed: 11/08/2022]
|
49
|
Gibson DA, Ma L. Developmental regulation of axon branching in the vertebrate nervous system. Development 2011; 138:183-95. [PMID: 21177340 DOI: 10.1242/dev.046441] [Citation(s) in RCA: 141] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
During nervous system development, axons generate branches to connect with multiple synaptic targets. As with axon growth and guidance, axon branching is tightly controlled in order to establish functional neural circuits, yet the mechanisms that regulate this important process are less well understood. Here, we review recent advances in the study of several common branching processes in the vertebrate nervous system. By focusing on each step in these processes we illustrate how different types of branching are regulated by extracellular cues and neural activity, and highlight some common principles that underlie the establishment of complex neural circuits in vertebrate development.
Collapse
Affiliation(s)
- Daniel A Gibson
- Zilkha Neurogenetic Institute, Department of Cell and Neurobiology, Keck School of Medicine, Neuroscience Graduate Program, University of Southern California, 1501 San Pablo Street, Los Angeles, CA 90089, USA
| | | |
Collapse
|
50
|
Genetic dissection of synaptic specificity. Curr Opin Neurobiol 2010; 21:93-9. [PMID: 21087855 DOI: 10.1016/j.conb.2010.10.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2010] [Revised: 10/04/2010] [Accepted: 10/21/2010] [Indexed: 11/22/2022]
Abstract
Nervous systems are built of a myriad of neurons connected by an even larger number of synapses. While it has been long known that neurons specifically select their synaptic partners among many possible choices during development, we only begin to understand how they make those decisions. Recent findings have started to elucidate the molecular mechanisms underlying synaptic target selection including positive as well as negative cues from synaptic partners, intermediate targets and surrounding tissues. Furthermore, emerging evidence suggests that synaptic connections are not only formed among specific sets of neurons, but also targeted to specific subcellular domains. Finally, spatial and temporal transcriptional regulation of these molecular cues represents an additional, versatile mechanism to provide wiring specificity.
Collapse
|