1
|
Meza-Sosa KF, Valle-Garcia D, González-Conchillos H, Blanco-Ayala T, Salazar A, Flores I, Gómez-Manzo S, González Esquivel DF, Pérez de la Cruz G, Pineda B, Pérez de la Cruz V. Molecular Mimicry between Toxoplasma gondii B-Cell Epitopes and Neurodevelopmental Proteins: An Immunoinformatic Approach. Biomolecules 2024; 14:933. [PMID: 39199321 PMCID: PMC11352964 DOI: 10.3390/biom14080933] [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: 06/19/2024] [Revised: 07/27/2024] [Accepted: 07/28/2024] [Indexed: 09/01/2024] Open
Abstract
Epidemiological studies and meta-analyses have shown a strong association between high seroprevalence of Toxoplasma gondii (T. gondii) and schizophrenia. Schizophrenic patients showed higher levels of anti-Toxoplasma immunoglobulins M and G (IgM and IgG) when compared to healthy controls. Previously, in a rat model, we demonstrated that the progeny of mothers immunized with T. gondii lysates before gestation had behavioral and social impairments during adulthood. Therefore, we suggested that T. gondii infection can trigger autoreactivity by molecularly mimicking host brain proteins. Here, we aimed to identify the occurrence of antigenic mimicry between T. gondii epitopes and host brain proteins. Using a bioinformatic approach, we predicted T. gondii RH-88 B cell epitopes and compared them to human cell-surface proteins involved in brain development and differentiation (BrainS). Five different algorithms for B-cell-epitope prediction were used and compared, resulting in 8584 T. gondii epitopes. We then compared T. gondii predicted epitopes to BrainS proteins by local sequence alignments using BLASTP. T. gondii immunogenic epitopes significantly overlapped with 42 BrainS proteins. Among these overlapping proteins essential for brain development and differentiation, we identified HSP90 and NOTCH receptors as the proteins most likely to be targeted by the maternally generated pathogenic antibodies due to their topological overlap at the extracellular region of their sequence. This analysis highlights the relevance of pregestational clinical surveillance and screening for potential pathogenic anti-T. gondii antibodies. It also identifies potential targets for the design of vaccines that could prevent behavioral and cognitive impairments associated with pre-gestational T. gondii exposure.
Collapse
Affiliation(s)
- Karla F. Meza-Sosa
- Neurochemistry and Behavior Laboratory, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico; (K.F.M.-S.); (T.B.-A.); (D.F.G.E.)
| | - David Valle-Garcia
- Neuroimmunology Department, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico; (D.V.-G.); (H.G.-C.); (A.S.); (I.F.)
| | - Hugo González-Conchillos
- Neuroimmunology Department, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico; (D.V.-G.); (H.G.-C.); (A.S.); (I.F.)
| | - Tonali Blanco-Ayala
- Neurochemistry and Behavior Laboratory, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico; (K.F.M.-S.); (T.B.-A.); (D.F.G.E.)
| | - Alelí Salazar
- Neuroimmunology Department, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico; (D.V.-G.); (H.G.-C.); (A.S.); (I.F.)
| | - Itamar Flores
- Neuroimmunology Department, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico; (D.V.-G.); (H.G.-C.); (A.S.); (I.F.)
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Manuel Carpio, Plutarco Elías Calles, Miguel Hidalgo, Mexico City 11350, Mexico
| | - Saúl Gómez-Manzo
- Laboratorio de Bioquímica Genética, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico;
| | - Dinora Fabiola González Esquivel
- Neurochemistry and Behavior Laboratory, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico; (K.F.M.-S.); (T.B.-A.); (D.F.G.E.)
| | - Gonzalo Pérez de la Cruz
- Department of Mathematics, Faculty of Sciences, Universidad Nacional Autónoma de Mexico (UNAM), Mexico City 04510, Mexico;
| | - Benjamín Pineda
- Neuroimmunology Department, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico; (D.V.-G.); (H.G.-C.); (A.S.); (I.F.)
| | - Verónica Pérez de la Cruz
- Neurochemistry and Behavior Laboratory, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico; (K.F.M.-S.); (T.B.-A.); (D.F.G.E.)
| |
Collapse
|
2
|
Belkozhayev A, Niyazova R, Kamal MA, Ivashchenko A, Sharipov K, Wilson CM. Differential microRNA expression in the SH-SY5Y human cell model as potential biomarkers for Huntington's disease. Front Cell Neurosci 2024; 18:1399742. [PMID: 39049823 PMCID: PMC11267620 DOI: 10.3389/fncel.2024.1399742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/26/2024] [Indexed: 07/27/2024] Open
Abstract
Huntington's disease (HD) is caused by an expansion of CAG trinucleotide repeat in the HTT gene; the exact pathogenesis of HD currently remains unclear. One of the promising directions in the study of HDs is to determine the molecular mechanism underlying the development and role of microRNAs (miRNAs). This study aimed to identify the profile of miRNAs in an HD human cell line model as diagnostic biomarkers for HD. To study HD, the human SH-SY5Y HD cell model is based on the expression of two different forms: pEGFP-Q23 and pEGFP-Q74 of HTT. The expression of Htt protein was confirmed using aggregation assays combined with immunofluorescence and Western blotting methods. miRNA levels were measured in SH-SY5Y neuronal cell model samples stably expressing Q23 and Q74 using the extraction-free HTG EdgeSeq protocol. A total of 2083 miRNAs were detected, and 354 (top 18 miRNAs) miRNAs were significantly differentially expressed (DE) (p < 0.05) in Q23 and Q74 cell lines. A majority of the miRNAs were downregulated in the HD cell model. Moreover, we revealed that six DE miRNAs target seven genes (ATN1, GEMIN4, EFNA5, CSMD2, CREBBP, ATXN1, and B3GNT) that play important roles in neurodegenerative disorders and showed significant expression differences in mutant Htt (Q74) when compared to wild-type Htt (Q23) using RT-qPCR (p < 0.05 and 0.01). We demonstrated the most important DE miRNA-mRNA profiles, interaction binding sites, and their related pathways in HD using experimental and bioinformatics methods. This will allow the development of novel diagnostic strategies and provide alternative therapeutic routes for treating HD.
Collapse
Affiliation(s)
- Ayaz Belkozhayev
- Life Sciences Industry Liaison Lab, School of Psychology and Life Sciences, Canterbury Christ Church University, Sandwich, United Kingdom
- M.A. Aitkhozhin Institute of Molecular Biology and Biochemistry, Almaty, Kazakhstan
- Department of Chemical and Biochemical Engineering, Geology and Oil-Gas Business Institute Named after K. Turyssov, Satbayev University, Almaty, Kazakhstan
- Faculty of Biology and Biotechnology, Al-Farabi Kazakh National University, Almaty, Kazakhstan
- Novel Global Community Educational Foundation, Hebersham, NSW, Australia
| | - Raigul Niyazova
- Faculty of Biology and Biotechnology, Al-Farabi Kazakh National University, Almaty, Kazakhstan
| | - Mohammad Amjad Kamal
- Novel Global Community Educational Foundation, Hebersham, NSW, Australia
- Center for High Altitude Medicine, Institutes for Systems Genetics, West China School of Nursing, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Pharmacy, Faculty of Health and Life Sciences, Daffodil International University, Dhaka, Bangladesh
- Centre for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, India
- Enzymoics, Hebersham, NSW, Australia
| | | | - Kamalidin Sharipov
- M.A. Aitkhozhin Institute of Molecular Biology and Biochemistry, Almaty, Kazakhstan
- Department of Biochemistry, Asfendiyarov Kazakh National Medical University, Almaty, Kazakhstan
| | - Cornelia M. Wilson
- Life Sciences Industry Liaison Lab, School of Psychology and Life Sciences, Canterbury Christ Church University, Sandwich, United Kingdom
- Novel Global Community Educational Foundation, Hebersham, NSW, Australia
- University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
3
|
Zhang S, Zhao J, Sha WM, Zhang XP, Mai JY, Bartlett PF, Hou ST. Inhibition of EphA4 reduces vasogenic edema after experimental stroke in mice by protecting the blood-brain barrier integrity. J Cereb Blood Flow Metab 2024; 44:419-433. [PMID: 37871622 PMCID: PMC10870966 DOI: 10.1177/0271678x231209607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/07/2023] [Accepted: 09/22/2023] [Indexed: 10/25/2023]
Abstract
Cerebral vasogenic edema, a severe complication of ischemic stroke, aggravates neurological deficits. However, therapeutics to reduce cerebral edema still represent a significant unmet medical need. Brain microvascular endothelial cells (BMECs), vital for maintaining the blood-brain barrier (BBB), represent the first defense barrier for vasogenic edema. Here, we analyzed the proteomic profiles of the cultured mouse BMECs during oxygen-glucose deprivation and reperfusion (OGD/R). Besides the extensively altered cytoskeletal proteins, ephrin type-A receptor 4 (EphA4) expressions and its activated phosphorylated form p-EphA4 were significantly increased. Blocking EphA4 using EphA4-Fc, a specific and well-tolerated inhibitor shown in our ongoing human phase I trial, effectively reduced OGD/R-induced BMECs contraction and tight junction damage. EphA4-Fc did not protect OGD/R-induced neuronal and astrocytic death. However, administration of EphA4-Fc, before or after the onset of transient middle cerebral artery occlusion (tMCAO), reduced brain edema by about 50%, leading to improved neurological function recovery. The BBB permeability test also confirmed that cerebral BBB integrity was well maintained in tMCAO brains treated with EphA4-Fc. Therefore, EphA4 was critical in signaling BMECs-mediated BBB breakdown and vasogenic edema during cerebral ischemia. EphA4-Fc is promising for the treatment of clinical post-stroke edema.
Collapse
Affiliation(s)
- Shuai Zhang
- Brain Research Centre, Department of Biology, School of Life Science, Southern University of Science and Technology, Shenzhen, P. R. China
| | - Jing Zhao
- Queensland Brain Institute, University of Queensland, Brisbane, Australia
| | - Wei-Meng Sha
- Brain Research Centre, Department of Biology, School of Life Science, Southern University of Science and Technology, Shenzhen, P. R. China
| | - Xin-Pei Zhang
- Brain Research Centre, Department of Biology, School of Life Science, Southern University of Science and Technology, Shenzhen, P. R. China
| | - Jing-Yuan Mai
- Brain Research Centre, Department of Biology, School of Life Science, Southern University of Science and Technology, Shenzhen, P. R. China
| | - Perry F Bartlett
- Queensland Brain Institute, University of Queensland, Brisbane, Australia
| | - Sheng-Tao Hou
- Brain Research Centre, Department of Biology, School of Life Science, Southern University of Science and Technology, Shenzhen, P. R. China
| |
Collapse
|
4
|
Paladini A, Vallejo R, Guerrero M, Pasqualucci A, Peppin JF, Pergolizzi J, Varrassi G. Answering Big Questions in Pain Medicine. Cureus 2023; 15:e43561. [PMID: 37719539 PMCID: PMC10502917 DOI: 10.7759/cureus.43561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 08/16/2023] [Indexed: 09/19/2023] Open
Abstract
The future of pain medicine is marked by many questions. What can other nations around the world learn from the opioid crisis that is still affecting the United States? The American opioid experience was mischaracterized and wrongly described, and its causes were misdiagnosed from the outset, leading to its mismanagement and the abandonment of many chronic pain patients to their suffering. There are a few new drugs in the analgesic armamentarium. What new targets do we have in pain medicine? There are many breakthroughs, discoveries, and potential new targets that could add to our analgesic prescribing choices. These include sigma receptors, d-amino acid oxidase, endoplasmic reticulum stress receptors, histone deacetylase, and others. Neuromodulation had been used with varying degrees of success for years, but with a simplistic approach based on the gate theory of pain. Despite our familiarity with neuromodulation and spinal cord stimulators, neuromodulation research indicates that the activation of glial cells may activate the immune system and enhance analgesia. Neuromodulation studies have concentrated on how electricity affects neuronal activity rather than how electrical activity could reduce pain. There are still more frontiers in our battle against pain and some promising avenues for treatments. This narrative review will try to summarize what can be done from the perspective of recent technological and pharmacological developments.
Collapse
Affiliation(s)
- Antonella Paladini
- Department of Life, Health & Environmental Sciences (MESVA), University of L'Aquila, L'Aquila, ITA
| | - Ricardo Vallejo
- Department of Research, Millennium Pain Center, Bloomington, USA
| | - Marixa Guerrero
- Department of Pain Medicine/ Pain Management, Clínica del Country, Bogota, COL
| | - Alberto Pasqualucci
- Department of Anesthesia and Critical Care, University of Perugia, Perugia, ITA
| | - John F Peppin
- Department of Osteopathic Medicine, Marian University, Indianapolis, USA
| | - Joseph Pergolizzi
- Department of Anesthesiology, Pain Medicine, and Critical Care Medicine, Nema Research, Naples, USA
| | | |
Collapse
|
5
|
The fornix acts as a permissive corridor for septal neuron migration beyond the diencephalic-telencephalic boundary. Sci Rep 2020; 10:8315. [PMID: 32433594 PMCID: PMC7239880 DOI: 10.1038/s41598-020-65284-7] [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: 12/02/2019] [Accepted: 04/28/2020] [Indexed: 11/08/2022] Open
Abstract
Neuronal migration is essential for constructing functional neural networks. Two posterior septal (PS) nuclei, the triangular septal nucleus and bed nuclei of the anterior commissure, are involved in fear and anxiety. During development, glutamatergic PS neurons undergo long-distance rostrodorsal migration from the thalamic eminence (TE) of the diencephalon, then settle in the caudalmost telencephalon. However, the developmental behavior of PS neurons and the guidance structures facilitating their migration remain unknown. We previously demonstrated the migration of PS neurons along the fornix, a major efferent pathway from the hippocampal formation. Here, we show that the postcommissural fornix is essential for PS neuron migration which is largely confined to its axonal tract, which grows in the opposite direction as PS neuron migration. Fornical axons reach the TE prior to initiation of PS neuron rostrodorsal migration. Ectopic expression of Semaphorin 3 A in the dorsomedial cortex resulted in defective fornix formation. Furthermore, loss of the postcommissural fornix stalled PS neuron migration resulting in abnormal accumulation near their origin. This suggests that PS neurons utilize the postcommissural fornix as a permissive corridor during migration beyond the diencephalic-telencephalic boundary. This axonal support is essential for the functional organization of the heterogeneous septal nuclear complex.
Collapse
|
6
|
Xiao L, Yang X, Loh YP. Neurotrophic, Gene Regulation, and Cognitive Functions of Carboxypeptidase E-Neurotrophic Factor-α1 and Its Variants. Front Neurosci 2019; 13:243. [PMID: 30941009 PMCID: PMC6433828 DOI: 10.3389/fnins.2019.00243] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 03/01/2019] [Indexed: 12/13/2022] Open
Abstract
Carboxypeptidase E, also known as neurotrophic factor-α1 (CPE-NFα1), was first discovered as an exopeptidase and is known to work by cleaving C-terminal basic amino acids from prohormone intermediates to produce mature peptide hormones and neuropeptides in the endocrine and central nervous systems, respectively. CPE-NFα1 also plays a critical role in prohormone sorting and secretory vesicle transportation. Recently, emerging studies have indicated that CPE-NFα1 exerts multiple non-enzymatic physiological roles in maintaining normal central nervous system function and in neurodevelopment. This includes potent neuroprotective and anti-depressant activities, as well as stem cell differentiation functions. In addition, N-terminal truncated variants of CPE-NFα1 have been identified to regulate expression of important neurodevelopmental genes. This mini-review summarizes recent advances in understanding the mechanisms underlying CPE-NFα1’s function in neuroprotection during stress and aspects of neurodevelopment.
Collapse
Affiliation(s)
- Lan Xiao
- Section on Cellular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Xuyu Yang
- Section on Cellular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Y Peng Loh
- Section on Cellular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
7
|
Watanabe K, Irie K, Hanashima C, Takebayashi H, Sato N. Diencephalic progenitors contribute to the posterior septum through rostral migration along the hippocampal axonal pathway. Sci Rep 2018; 8:11728. [PMID: 30082833 PMCID: PMC6078977 DOI: 10.1038/s41598-018-30020-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 07/23/2018] [Indexed: 12/21/2022] Open
Abstract
Septal nuclei are telencephalic structures associated with a variety of brain functions as part of the limbic system. The two posterior septal nuclei, the triangular septal nucleus (TS) and the bed nuclei of the anterior commissure (BAC), are involved in fear and anxiety through their projections to the medial habenular nucleus. However, the development of both the TS and BAC remains unclear. Here, we found a novel caudal origin and putative migratory stream of mouse posterior septal neurons arising from the thalamic eminence (TE), a transient developmental structure at the rostral end of the rodent diencephalon. TE-derived cells, which have glutamatergic identity, migrated rostrally and entered the telencephalic territory by passing beneath the third ventricle. Subsequently, they turned dorsally toward the posterior septum. We also observed that TS and BAC neurons in the postnatal septum were labeled with GFP by in utero electroporation into the TE, suggesting a shared origin. Furthermore, TE-derived septal neurons migrated along the fornix, an efferent pathway from the hippocampus. These results demonstrate that posterior septal neurons have a distinct extratelencephalic origin from other septal nuclei. This heterogeneous origin may contribute to neuronal diversity of the septal nuclear complex.
Collapse
Affiliation(s)
- Keisuke Watanabe
- Division of Gross Anatomy and Morphogenesis, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 951-8510, Japan.
| | - Koichiro Irie
- Faculty of Education and Integrated Arts and Sciences, Waseda University, Shinjuku-ku, Tokyo, 162-8480, Japan
| | - Carina Hanashima
- Faculty of Education and Integrated Arts and Sciences, Waseda University, Shinjuku-ku, Tokyo, 162-8480, Japan.,Graduate School of Advanced Science and Engineering, Waseda University, Shinjuku-ku, Tokyo, 162-8480, Japan
| | - Hirohide Takebayashi
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 951-8510, Japan
| | - Noboru Sato
- Division of Gross Anatomy and Morphogenesis, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 951-8510, Japan
| |
Collapse
|
8
|
Mata A, Gil V, Pérez-Clausell J, Dasilva M, González-Calixto MC, Soriano E, García-Verdugo JM, Sanchez-Vives MV, Del Río JA. New functions of Semaphorin 3E and its receptor PlexinD1 during developing and adult hippocampal formation. Sci Rep 2018; 8:1381. [PMID: 29358640 PMCID: PMC5777998 DOI: 10.1038/s41598-018-19794-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 01/08/2018] [Indexed: 12/19/2022] Open
Abstract
The development and maturation of cortical circuits relies on the coordinated actions of long and short range axonal guidance cues. In this regard, the class 3 semaphorins and their receptors have been seen to be involved in the development and maturation of the hippocampal connections. However, although the role of most of their family members have been described, very few data about the participation of Semaphorin 3E (Sema3E) and its receptor PlexinD1 during the development and maturation of the entorhino-hippocampal (EH) connection are available. In the present study, we focused on determining their roles both during development and in adulthood. We determined a relevant role for Sema3E/PlexinD1 in the layer-specific development of the EH connection. Indeed, mice lacking Sema3E/PlexinD1 signalling showed aberrant layering of entorhinal axons in the hippocampus during embryonic and perinatal stages. In addition, absence of Sema3E/PlexinD1 signalling results in further changes in postnatal and adult hippocampal formation, such as numerous misrouted ectopic mossy fibers. More relevantly, we describe how subgranular cells express PlexinD1 and how the absence of Sema3E induces a dysregulation of the proliferation of dentate gyrus progenitors leading to the presence of ectopic cells in the molecular layer. Lastly, Sema3E mutant mice displayed increased network excitability both in the dentate gyrus and the hippocampus proper.
Collapse
Affiliation(s)
- Agata Mata
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Parc Científic de Barcelona, Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, (CIBERNED), Barcelona, Spain.,Institut de Neurociències de la Universitat de Barcelona, Barcelona, Spain
| | - Vanessa Gil
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Parc Científic de Barcelona, Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, (CIBERNED), Barcelona, Spain.,Institut de Neurociències de la Universitat de Barcelona, Barcelona, Spain
| | - Jeús Pérez-Clausell
- Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain
| | - Miguel Dasilva
- Systems Neuroscience, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Mari Carmen González-Calixto
- Laboratory of Comparative Neurobiology, Institute Cavanilles, University of Valencia, CIBERNED, 46980, Valencia, Spain
| | - Eduardo Soriano
- Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, (CIBERNED), Barcelona, Spain.,Institut de Neurociències de la Universitat de Barcelona, Barcelona, Spain.,ICREA, Barcelona, Spain.,Vall d'Hebrón Institut de Recerca (VHIR), Barcelona, Spain
| | - José Manuel García-Verdugo
- Laboratory of Comparative Neurobiology, Institute Cavanilles, University of Valencia, CIBERNED, 46980, Valencia, Spain
| | - Maria V Sanchez-Vives
- Systems Neuroscience, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,ICREA, Barcelona, Spain
| | - José Antonio Del Río
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Parc Científic de Barcelona, Barcelona, Spain. .,Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain. .,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, (CIBERNED), Barcelona, Spain. .,Institut de Neurociències de la Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
9
|
Increased EphA4-ephexin1 signaling in the medial prefrontal cortex plays a role in depression-like phenotype. Sci Rep 2017; 7:7133. [PMID: 28769056 PMCID: PMC5541046 DOI: 10.1038/s41598-017-07325-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 06/23/2017] [Indexed: 02/02/2023] Open
Abstract
Accumulating evidence suggests a role of the ephrin receptor EphA4 and the downstream protein ephexin1 in synaptic plasticity, which is implicated in depression. We examined whether EphA4–ephexin1 signaling plays a role in the pathophysiology of depression, and the antidepressant-like effect of EphA4 inhibitor rhynchophylline. We found increased ratios of p-EphA4/EphA4 and p-ephexin1/ephexin1 in the prefrontal cortex (PFC) and hippocampus but not in the nucleus accumbens (NAc), of susceptible mice after social defeat stress. Furthermore, the p-EphA4/EphA4 ratio was higher in the parietal cortex of depressed patients compared with controls. Systemic administration of rhynchophylline, produced a rapid antidepressant-like effect in a social defeat stress model by inhibiting EphA4–ephexin1 signaling and activating brain-derived neurotrophic factor-TrkB signaling in the PFC and hippocampus. Pretreatment with rhynchophylline before each social defeat stress could prevent the onset of the depression-like phenotype after repeated social defeat stress. Overexpression of EphA4 in the medial PFC owing to infection with an EphA4 adeno-associated virus caused the depression-like phenotype 3 weeks later and rhynchophylline had a rapid antidepressant-like effect in these mice. These findings suggest that increased EphA4–ephexin1 signaling in the PFC plays a role in the pathophysiology of depression.
Collapse
|
10
|
Hennig KM, Fass DM, Zhao WN, Sheridan SD, Fu T, Erdin S, Stortchevoi A, Lucente D, Cody JD, Sweetser D, Gusella JF, Talkowski ME, Haggarty SJ. WNT/β-Catenin Pathway and Epigenetic Mechanisms Regulate the Pitt-Hopkins Syndrome and Schizophrenia Risk Gene TCF4. MOLECULAR NEUROPSYCHIATRY 2017; 3:53-71. [PMID: 28879201 DOI: 10.1159/000475666] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 04/07/2017] [Indexed: 12/11/2022]
Abstract
Genetic variation within the transcription factor TCF4 locus can cause the intellectual disability and developmental disorder Pitt-Hopkins syndrome (PTHS), whereas single-nucleotide polymorphisms within noncoding regions are associated with schizophrenia. These genetic findings position TCF4 as a link between transcription and cognition; however, the neurobiology of TCF4 remains poorly understood. Here, we quantitated multiple distinct TCF4 transcript levels in human induced pluripotent stem cell-derived neural progenitors and differentiated neurons, and PTHS patient fibroblasts. We identify two classes of pharmacological treatments that regulate TCF4 expression: WNT pathway activation and inhibition of class I histone deacetylases. In PTHS fibroblasts, both of these perturbations upregulate a subset of TCF4 transcripts. Finally, using chromatin immunoprecipitation sequencing in conjunction with genome-wide transcriptome analysis, we identified TCF4 target genes that may mediate the effect of TCF4 loss on neuroplasticity. Our studies identify new pharmacological assays, tools, and targets for the development of therapeutics for cognitive disorders.
Collapse
Affiliation(s)
- Krista M Hennig
- Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital, Massachusetts, USA.,Department of Neurology, Harvard Medical School, Boston, Massachusetts, USA.,Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA
| | - Daniel M Fass
- Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital, Massachusetts, USA.,Department of Neurology, Harvard Medical School, Boston, Massachusetts, USA.,Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA.,Stanley Center for Psychiatric Research, Broad Institute, Cambridge, Massachusetts, USA
| | - Wen-Ning Zhao
- Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital, Massachusetts, USA.,Department of Neurology, Harvard Medical School, Boston, Massachusetts, USA.,Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA
| | - Steven D Sheridan
- Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital, Massachusetts, USA.,Department of Neurology, Harvard Medical School, Boston, Massachusetts, USA.,Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA
| | - Ting Fu
- Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital, Massachusetts, USA.,Department of Neurology, Harvard Medical School, Boston, Massachusetts, USA.,Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA
| | - Serkan Erdin
- Molecular Neurogenetics Unit, Massachusetts General Hospital, Boston, Massachusetts, USA.,Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Alexei Stortchevoi
- Molecular Neurogenetics Unit, Massachusetts General Hospital, Boston, Massachusetts, USA.,Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Diane Lucente
- Molecular Neurogenetics Unit, Massachusetts General Hospital, Boston, Massachusetts, USA.,Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jannine D Cody
- Chromosome 18 Clinical Research Center, Department of Pediatrics, University of Texas Health Sciences Center, San Antonio, Texas, USA.,The Chromosome 18 Registry and Research Society, San Antonio, Texas, USA
| | - David Sweetser
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA.,Divisions of Pediatric Hematology/Oncology and Medical Genetics, Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - James F Gusella
- Molecular Neurogenetics Unit, Massachusetts General Hospital, Boston, Massachusetts, USA.,Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael E Talkowski
- Department of Neurology, Harvard Medical School, Boston, Massachusetts, USA.,Molecular Neurogenetics Unit, Massachusetts General Hospital, Boston, Massachusetts, USA.,Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Stephen J Haggarty
- Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital, Massachusetts, USA.,Department of Neurology, Harvard Medical School, Boston, Massachusetts, USA.,Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA.,Stanley Center for Psychiatric Research, Broad Institute, Cambridge, Massachusetts, USA.,Molecular Neurogenetics Unit, Massachusetts General Hospital, Boston, Massachusetts, USA.,Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
11
|
EphA4 Regulates Neuroblast and Astrocyte Organization in a Neurogenic Niche. J Neurosci 2017; 37:3331-3341. [PMID: 28258169 DOI: 10.1523/jneurosci.3738-16.2017] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 02/10/2017] [Accepted: 02/13/2017] [Indexed: 11/21/2022] Open
Abstract
Significant migration cues are required to guide and contain newly generated rodent subventricular zone (SVZ) neuroblasts as they transit along the lateral ventricles and then through the anterior forebrain to their ultimate site of differentiation in the olfactory bulbs (OBs). These cues enforce strict neuroblast spatial boundaries within the dense astroglial meshwork of the SVZ and rostral migratory stream (RMS), yet are permissive to large-scale neuroblast migration. Therefore, the molecular mechanisms that define these cues and control dynamic interactions between migratory neuroblasts and surrounding astrocytes are of particular interest. We found that deletion of EphA4 and specifically ablation of EphA4 kinase activity resulted in misaligned neuroblasts and disorganized astrocytes in the RMS/SVZ, linking EphA4 forward signaling to SVZ and RMS spatial organization, orientation, and regulation. In addition, within a 3 week period, there was a significant reduction in the number of neuroblasts that reached the OB and integrated into the periglomerular layer, revealing a crucial role for EphA4 in facilitating efficient neuroblast migration to the OB. Single-cell analysis revealed that EPHA4 and its EFN binding partners are expressed by subpopulations of neuroblasts and astrocytes within the SVZ/RMS/OB system resulting in a cell-specific mosaic, suggesting complex EphA4 signaling involving both homotypic and heterotypic cell-cell interactions. Together, our studies reveal a novel molecular mechanism involving EphA4 signaling that functions in stem cell niche organization and ultimately neuroblast migration in the anterior forebrain.SIGNIFICANCE STATEMENT The subventricular zone neurogenic stem cell niche generates highly migratory neuroblasts that transit the anterior forebrain along a defined pathway to the olfactory bulb. Postnatal and adult brain organization dictates strict adherence to a narrow migration corridor. Subventricular zone neuroblasts are aligned in tightly bundled chains within a meshwork of astrocytes; however, the cell-cell cues that organize this unique, cell-dense migration pathway are largely unknown. Our studies show that forward signaling through the EphA4 tyrosine kinase receptor, mediated by ephrins expressed by subpopulations of neuroblasts and astrocytes, is required for compact, directional organization of neuroblasts and astrocytes within the pathway and efficient transit of neuroblasts through the anterior forebrain to the olfactory bulb.
Collapse
|
12
|
Wei W, Wang H, Ji S. Paradoxes of the EphB1 receptor in malignant brain tumors. Cancer Cell Int 2017; 17:21. [PMID: 28194092 PMCID: PMC5299699 DOI: 10.1186/s12935-017-0384-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 01/20/2017] [Indexed: 02/07/2023] Open
Abstract
Eph receptors are a subfamily of receptor tyrosine kinases. Eph receptor-mediated forward and ephrin ligand-mediated reverse signalings are termed bidirectional signaling. Increasing evidence shows that Eph/ephrin signaling regulates cell migration, adhesion, morphological changes, differentiation, proliferation and survival through cell–cell communication. Some recent studies have started to implicate Eph/ephrin signaling in tumorigenesis, metastasis, and angiogenesis. Previous studies have shown that EphB1 receptor and its ephrin ligands are expressed in the central nervous system. EphB1/ephrin signaling plays an important role in the regulation of synapse formation and maturation, migration of neural progenitors, establishment of tissue patterns, and the development of immune organs. Besides, various recent studies have detected the abnormal expression of EphB1 receptor in different brain tumors. However, the underlying molecular mechanisms of EphB1/ephrins signaling in the development of these tumors are not fully understood. This review focuses on EphB1 that has both tumor-suppressing and -promoting roles in some brain tumors. Understanding the intracellular mechanisms of EphB1 in tumorigenesis and metastasis of brain tumors might provide a foundation for the development of EphB1-targeted therapies.
Collapse
Affiliation(s)
- Wenqiang Wei
- Laboratory of Cell Signal Transduction, Medical School, Henan University, Kaifeng, 475004 China.,Department of Microbiology, Medical School, Henan University, Kaifeng, 475004 China
| | - Hongju Wang
- Department of Anatomy, Medical School, Henan University, Kaifeng, 475004 China
| | - Shaoping Ji
- Laboratory of Cell Signal Transduction, Medical School, Henan University, Kaifeng, 475004 China.,Department of Oncology, The First Affiliated Hospital, Henan University, Kaifeng, 475001 China
| |
Collapse
|
13
|
Chitramuthu B, Bateman A. Progranulin and the receptor tyrosine kinase EphA2, partners in crime? J Cell Biol 2016; 215:603-605. [PMID: 27903608 PMCID: PMC5147006 DOI: 10.1083/jcb.201610097] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 10/26/2016] [Indexed: 11/22/2022] Open
Abstract
Chitramuthu and Bateman highlight recent work proposing that the receptor tyrosine kinase EphA2 functions as receptor for the growth factor progranulin. Progranulin is a secreted protein with roles in tumorigenesis, inflammation, and neurobiology, but its signaling receptors have remained unclear. In this issue, Neill et al. (2016. J. Cell Biol. https://doi.org/10.1083/jcb.201603079) identify the tyrosine kinase EphA2 as a strong candidate for such a receptor, providing insight into progranulin and EphA2 signaling.
Collapse
Affiliation(s)
- Babykumari Chitramuthu
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada.,Centre for Translational Biology, Platform in Experimental Therapeutics and Metabolism, McGill University Health Centre Research Institute, Montreal, QC H4A 3J1, Canada
| | - Andrew Bateman
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada.,Centre for Translational Biology, Platform in Experimental Therapeutics and Metabolism, McGill University Health Centre Research Institute, Montreal, QC H4A 3J1, Canada
| |
Collapse
|
14
|
Inverse Expression Levels of EphrinA3 and EphrinA5 Contribute to Dopaminergic Differentiation of Human SH-SY5Y Cells. J Mol Neurosci 2016; 59:483-92. [PMID: 27217159 DOI: 10.1007/s12031-016-0759-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 04/22/2016] [Indexed: 10/21/2022]
Abstract
Two key principles underlying successful cellular therapies for Parkinson's disease (PD) are appropriate differentiation of dopaminergic (DA) neurons from transplanted cells and precise axon growth. EphrinAs, a subclass of ephrins, act as axon guidance molecules and are highly expressed in DA brain regions. Existing evidences indicate that they act as either repulsion or attraction signals to guide axon growth. This study investigated whether ephrinAs are involved in DA neuron differentiation. Data from miRCURY™ LNA mRNAs/microRNAs microarrays and quantitative real-time polymerase chain reaction (qRT-PCR) showed upregulated ephrinA3 mRNA (EFNA3) and downregulated ephrinA5 mRNA (EFNA5) during DA neuron differentiation. In addition, hsa-miR-4271 was downregulated, which could influence EFNA3 translation. Furthermore, immunofluorescence (IF) and western blotting confirmed the mRNA results and showed increased ephrinA3 and decreased ephrinA5 protein levels in differentiating DA neurons. Taken together, our results indicate that inverse expression levels of ephrinA3 and ephrinA5, which are possibly influenced by microRNAs, contribute to DA neuron differentiation by guiding axon growth.
Collapse
|
15
|
Chen X, Yang H, Zhou X, Zhang L, Lu X. MiR-93 Targeting EphA4 Promotes Neurite Outgrowth from Spinal Cord Neurons. J Mol Neurosci 2016; 58:517-24. [PMID: 26798048 DOI: 10.1007/s12031-015-0709-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Accepted: 12/28/2015] [Indexed: 11/26/2022]
Abstract
The failure of neurite outgrowth in the adult mammalian spinal cord injury is thought to be attributed to the intrinsic growth ability of mature neurons. Ephrin/Eph system is a major growth regulator of many axonal guidance processes. EphA4 is expressed specifically in traumatic central nervous system (CNS) and dynamically regulate target gene expression, suggesting that it may be associated with neural regeneration. Here, we found an alteration in temporal expression of miR-93 following a contusive spinal cord injury (SCI) in adult rats. The messenger RNA (mRNA) expression level of miR-93 was upregulated and the protein expression levels of EphA4, p-Ephexin, and active RhoA were all decreased in traumatic spinal cord relative to those with an intact spinal cord. Infection of cultured spinal cord neurons (SCNs) with miR-93 mimic led to neuronal growth promotion and decreased levels of EphA4, p-Ephexin, and active RhoA protein expression. Dual-luciferase reporter assay confirmed that miR-93 bound to the three prime untranslated region (3' UTR) of EphA4 and inhibited the expression of EphA4 mRNA. These findings provide evidence that miR-93 inhibits EphA4 expression, decreased EphA4 expression could promote neurite outgrowth in SCNs due to reduced levels of p-Ephexin and active RhoA.
Collapse
Affiliation(s)
- Xiaogang Chen
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China
- Department of Orthopedic Surgery, Huai'an NO.2 People's Hospital, Affiliated Huai'an Hospital of Xuzhou Medical College, Huai'an, 223002, Jiangsu Province, China
| | - Huilin Yang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China.
| | - Xiaoqing Zhou
- Department of Orthopedic Surgery, Huai'an NO.2 People's Hospital, Affiliated Huai'an Hospital of Xuzhou Medical College, Huai'an, 223002, Jiangsu Province, China
| | - Lin Zhang
- Department of Orthopedic Surgery, Huai'an NO.2 People's Hospital, Affiliated Huai'an Hospital of Xuzhou Medical College, Huai'an, 223002, Jiangsu Province, China
| | - Xiaoqing Lu
- Department of Orthopedic Surgery, Huai'an NO.2 People's Hospital, Affiliated Huai'an Hospital of Xuzhou Medical College, Huai'an, 223002, Jiangsu Province, China
| |
Collapse
|
16
|
The Development of the Calvarial Bones and Sutures and the Pathophysiology of Craniosynostosis. Curr Top Dev Biol 2015; 115:131-56. [PMID: 26589924 DOI: 10.1016/bs.ctdb.2015.07.004] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The skull vault is a complex, exquisitely patterned structure that plays a variety of key roles in vertebrate life, ranging from the acquisition of food to the support of the sense organs for hearing, smell, sight, and taste. During its development, it must meet the dual challenges of protecting the brain and accommodating its growth. The bones and sutures of the skull vault are derived from cranial neural crest and head mesoderm. The frontal and parietal bones develop from osteogenic rudiments in the supraorbital ridge. The coronal suture develops from a group of Shh-responsive cells in the head mesoderm that are collocated, with the osteogenic precursors, in the supraorbital ridge. The osteogenic rudiments and the prospective coronal suture expand apically by cell migration. A number of congenital disorders affect the skull vault. Prominent among these is craniosynostosis, the fusion of the bones at the sutures. Analysis of the pathophysiology underling craniosynostosis has identified a variety of cellular mechanisms, mediated by a range of signaling pathways and effector transcription factors. These cellular mechanisms include loss of boundary integrity, altered sutural cell specification in embryos, and loss of a suture stem cell population in adults. Future work making use of genome-wide transcriptomic approaches will address the deep structure of regulatory interactions and cellular processes that unify these seemingly diverse mechanisms.
Collapse
|
17
|
Schneider A, Puechberty J, Ng BL, Coubes C, Gatinois V, Tournaire M, Girard M, Dumont B, Bouret P, Magnetto J, Baghdadli A, Pellestor F, Geneviève D. Identification of disrupted AUTS2 and EPHA6 genes by array painting in a patient carrying a de novo balanced translocation t(3;7) with intellectual disability and neurodevelopment disorder. Am J Med Genet A 2015; 167A:3031-7. [PMID: 26333717 DOI: 10.1002/ajmg.a.37350] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 08/12/2015] [Indexed: 11/09/2022]
Abstract
Intellectual disability (ID) is a frequent feature but is highly clinically and genetically heterogeneous. The establishment of the precise diagnosis in patients with ID is challenging due to this heterogeneity but crucial for genetic counseling and appropriate care for the patients. Among the etiologies of patients with ID, apparently balanced de novo rearrangements represent 0.6%. Several mechanisms explain the ID in patients with apparently balanced de novo rearrangement. Among them, disruption of a disease gene at the breakpoint, is frequently evoked. In this context, technologies recently developed are used to characterize precisely such chromosomal rearrangements. Here, we report the case of a boy with ID, facial features and autistic behavior who is carrying a de novo balanced reciprocal translocation t(3;7)(q11.2;q11.22)dn. Using microarray analysis, array painting (AP) technology combined with molecular study, we have identified the interruption of the autism susceptibility candidate 2 gene (AUTS2) and EPH receptor A6 gene (EPHA6). We consider that the disruption of AUTS2 explains the phenotype of the patient; the exact role of EPHA6 in human pathology is not well defined. Based on the observation of recurrent germinal and somatic translocations involving AUTS2 and the molecular environment content, we put forward the hypothesis that the likely chromosomal mechanism responsible for the translocation could be due either to replicative stress or to recombination-based mechanisms.
Collapse
Affiliation(s)
- Anouck Schneider
- Laboratoire de Génétique Chromosomique, Plateforme de puces à ADN, CHRU de Montpellier, France
| | | | - Bee Ling Ng
- Cytometry Core Facility, The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | | | - Vincent Gatinois
- Laboratoire de Génétique Chromosomique, Plateforme de puces à ADN, CHRU de Montpellier, France
| | - Magali Tournaire
- Laboratoire de Génétique Chromosomique, Plateforme de puces à ADN, CHRU de Montpellier, France
| | - Manon Girard
- Laboratoire de Génétique Chromosomique, Plateforme de puces à ADN, CHRU de Montpellier, France
| | - Bruno Dumont
- Laboratoire de Génétique Chromosomique, Plateforme de puces à ADN, CHRU de Montpellier, France
| | - Pauline Bouret
- Laboratoire de Génétique Chromosomique, Plateforme de puces à ADN, CHRU de Montpellier, France
| | - Julia Magnetto
- CRA, Département de Psychiatrie de l'Enfant et de l'Adolescent, Centre de Ressources Autisme, CHRU de Montpellier, France
| | - Amaria Baghdadli
- CRA, Département de Psychiatrie de l'Enfant et de l'Adolescent, Centre de Ressources Autisme, CHRU de Montpellier, France
| | - Franck Pellestor
- Laboratoire de Génétique Chromosomique, Plateforme de puces à ADN, CHRU de Montpellier, France
| | - David Geneviève
- Laboratoire de Génétique Chromosomique, Plateforme de puces à ADN, CHRU de Montpellier, France.,Département de Génétique Médicale, CHRU de Montpellier, France
| |
Collapse
|
18
|
Li J, Cui J, Wang X, Ma J, Niu H, Ma X, Zhang X, Liu S. An association study between DLGAP1 rs11081062 and EFNA5 rs26728 polymorphisms with obsessive-compulsive disorder in a Chinese Han population. Neuropsychiatr Dis Treat 2015; 11:897-905. [PMID: 25897225 PMCID: PMC4389912 DOI: 10.2147/ndt.s75009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND A recent genome-wide association study indicated that the single nucleotide polymorphisms (SNPs) rs11081062 in DLGAP1 and rs26728 in EFNA5 were associated with obsessive-compulsive disorder (OCD) in Caucasians. The present case-control association study assessed the global relevance of these two SNPs with respect to OCD subtypes in a Chinese Han population. METHODS We recruited 320 OCD patients and 431 age- and sex-matched controls from a Chinese Han population. rs11081062 and rs26728 SNPs were genotyped by real-time TaqMan polymerase chain reaction, and the chi-squared test was used to compare allele and genotype frequencies of variants between the two groups. RESULTS No significant differences were found in allele or genotype frequencies of DLGAP1 rs11081062 and EFNA5 rs26728 between the OCD and control groups. Moreover, consistently negative results were observed when classifying by sex, onset age, and comorbidity. However, on analyzing OCD subphenotypes, significant associations were observed between rs11081062 and the presence of contamination obsessions and cleaning compulsions (χ (2)=7.724, P=0.021 by genotype; χ (2)=3.745, P=0.053 by allele; and χ (2)=0.821, P=0.365 by genotype, χ (2)=27.809, P=0.000 by allele, respectively), and rs26728 with the presence of repeating compulsions (χ (2)=8.285, P=0.004 by genotype; χ (2)=7.512, P=0.006 by allele). CONCLUSION Although we found no association between DLGAP1 rs11081062 and EFNA5 rs26728 SNPs with OCD in a Chinese Han population, obvious associations were observed with OCD subphenotypes. Therefore, it appears to be useful to divide OCD into more homogeneous subphenotypes to help understand the complex genetic basis of this disorder. Further investigations are needed to replicate these findings using larger sample sizes, different populations, and other polymorphisms.
Collapse
Affiliation(s)
- Jiang Li
- Physiatry Department, The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Jiajia Cui
- Department of Psychiatry, Qingdao University, Qingdao, People's Republic of China
| | - Xiuhai Wang
- Department of Genetics, Medical College, Qingdao University, Qingdao, People's Republic of China
| | - Jianhua Ma
- The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Haitao Niu
- The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Xu Ma
- Graduate School, Peking Union Medical College, Beijing, People's Republic of China ; National Research Institute for Family Planning, Beijing, People's Republic of China ; World Health Organization Collaborating Centre for Research in Human Reproduction, Beijing, People's Republic of China
| | - Xinhua Zhang
- Department of Psychiatry, Qingdao University, Qingdao, People's Republic of China ; The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Shiguo Liu
- Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| |
Collapse
|
19
|
The Binding Receptors of Aβ: an Alternative Therapeutic Target for Alzheimer's Disease. Mol Neurobiol 2014; 53:455-471. [PMID: 25465238 DOI: 10.1007/s12035-014-8994-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 11/06/2014] [Indexed: 01/18/2023]
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative disorders, which causes the deterioration of memory and other cognitive abilities of the elderly. Previous lines of research have shown that Aβ is an essential factor in AD pathology and the soluble oligomeric species of Aβ peptide is presumed to be the drivers of synaptic impairment in AD. However, the exact mechanisms underlying Aβ-induced synapse dysfunction are still not fully understood. Recently, increasing evidence suggests that some potential receptors which bind specifically with Aβ may play important roles in inducing the toxicity of the neurons in AD pathology. These receptors include the cellular prion protein (PrPc), the α7 nicotinic acetylcholine receptor (α7nAChR), the p75 neurotrophin receptor (p75(NTR)), the beta-adrenergic receptors (β-ARs), the Eph receptors, the paired immunoglobulin-like receptor B (PirB), the PirB's human ortholog receptor (LilrB2), and the Fcγ receptor II-b (FcγRIIb). This review summarizes the characters of these prominent receptors and how the bindings of them with Aβ inhibit the LTP, decrease the number of dendritic spine, damage the neurons, and so on in AD pathogenesis. Blocking or rescuing these receptors may have significant importance for AD treatments.
Collapse
|
20
|
Nakagawa M, Inokuchi M, Takagi Y, Kato K, Sugita H, Otsuki S, Kojima K, Uetake H, Sugihara K. Erythropoietin-Producing Hepatocellular A1 is an Independent Prognostic Factor for Gastric Cancer. Ann Surg Oncol 2014; 22:2329-35. [DOI: 10.1245/s10434-014-4231-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Indexed: 12/25/2022]
|
21
|
Cissé M, Checler F. Eph receptors: new players in Alzheimer's disease pathogenesis. Neurobiol Dis 2014; 73:137-49. [PMID: 25193466 DOI: 10.1016/j.nbd.2014.08.028] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 08/01/2014] [Accepted: 08/22/2014] [Indexed: 12/23/2022] Open
Abstract
Alzheimer's disease (AD) is devastating and leads to permanent losses of memory and other cognitive functions. Although recent genetic evidences strongly argue for a causative role of Aβ in AD onset and progression (Jonsson et al., 2012), its role in AD etiology remains a matter of debate. However, even if not the sole culprit or pathological trigger, genetic and anatomical evidences in conjunction with numerous pharmacological studies, suggest that Aβ peptides, at least contribute to the disease. How Aβ contributes to memory loss remains largely unknown. Soluble Aβ species referred to as Aβ oligomers have been shown to be neurotoxic and induce network failure and cognitive deficits in animal models of the disease. In recent years, several proteins were described as potential Aβ oligomers receptors, amongst which are the receptor tyrosine kinases of Eph family. These receptors together with their natural ligands referred to as ephrins have been involved in a plethora of physiological and pathological processes, including embryonic neurogenesis, learning and memory, diabetes, cancers and anxiety. Here we review recent discoveries on Eph receptors-mediated protection against Aβ oligomers neurotoxicity as well as their potential as therapeutic targets in AD pathogenesis.
Collapse
Affiliation(s)
- Moustapha Cissé
- Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275 CNRS/UNS, "Labex Distalz", 660 route des Lucioles, 06560, Sophia-Antipolis, Valbonne, France..
| | - Frédéric Checler
- Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275 CNRS/UNS, "Labex Distalz", 660 route des Lucioles, 06560, Sophia-Antipolis, Valbonne, France..
| |
Collapse
|
22
|
Fukusumi Y, Miyauchi N, Hashimoto T, Saito A, Kawachi H. Therapeutic target for nephrotic syndrome: Identification of novel slit diaphragm associated molecules. World J Nephrol 2014; 3:77-84. [PMID: 25332898 PMCID: PMC4202494 DOI: 10.5527/wjn.v3.i3.77] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 05/15/2014] [Accepted: 06/27/2014] [Indexed: 02/06/2023] Open
Abstract
The slit diaphragm bridging the neighboring foot processes functions as a final barrier of glomerular capillary wall for preventing the leak of plasma proteins into primary urine. It is now accepted that the dysfunction of the sit diaphragm contributes to the development of proteinuria in several glomerular diseases. Nephrin, a gene product of NPHS1, a gene for a congenital nephrotic syndrome of Finnish type, constitutes an extracellular domain of the slit diaphragm. Podocin was identified as a gene product of NPHS2, a gene for a familial steroid-resistant nephrotic syndrome of French. Podocin binds the cytoplasmic domain of nephrin. After then, CD2 associated protein, NEPH1 and transient receptor potential-6 were also found as crucial molecules of the slit diaphragm. In order to explore other novel molecules contributing to the development of proteinuria, we performed a subtraction hybridization assay with a normal rat glomerular RNA and a glomerular RNA of rats with a puromycin aminonucleoside nephropathy, a mimic of a human minimal change type nephrotic syndrome. Then we have found that synaptic vesicle protein 2B, ephrin-B1 and neurexin were already downregulated at the early stage of puromycin aminonucleoside nephropathy, and that these molecules were localized close to nephrin. It is conceivable that these molecules are the slit diaphragm associated molecules, which participate in the regulation of the barrier function. These molecules could be targets to establish a novel therapy for nephrotic syndrome.
Collapse
|
23
|
Rudolph J, Gerstmann K, Zimmer G, Steinecke A, Döding A, Bolz J. A dual role of EphB1/ephrin-B3 reverse signaling on migrating striatal and cortical neurons originating in the preoptic area: should I stay or go away? Front Cell Neurosci 2014; 8:185. [PMID: 25100946 PMCID: PMC4103172 DOI: 10.3389/fncel.2014.00185] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 06/16/2014] [Indexed: 11/23/2022] Open
Abstract
During embryonic development the preoptic area (POA) gives rise to two populations of neurons which are generated at the same time, cortical interneurons and striatal cells. POA-derived cortical interneurons take a superficial path and avoid the developing striatum (Str) when they migrate to their target region. We found that EphB1, which is expressed in the striatal anlage, prevents cortical interneurons from entering the Str via ephrin-B3 reverse signaling. In contrast, for striatal neurons which also express ephrin-B3, EphB1 acts as a stop signal. This dual role of EphB1 is due to differences in ephrin-B3 reverse signaling cascades. For striatal neurons, binding of EphB1 to ephrin-B3 reduces endogenously high levels of pSrc and pFAK, which then causes the cells to stop migration. In contrast, in cortical interneurons EphB1-ephrin-B3 reverse signaling leads to phosphorylation of Src and focal adhesion kinase (FAK) which then mediates repulsion. Consistent with these in vitro findings, in an ephrin-B3 knockout mouse line, we discovered misrouted cortical interneurons in the Str and an over-migration of striatal neurons in their target region. Thus, EphB1/ephrin-B3 reverse signaling has a different impact on two sets of neurons which are generated at the same time and place: it can act as a repulsive cue for migrating neurons or it can terminate neuronal migration, a novel role of the Eph/ephrin system.
Collapse
Affiliation(s)
- Judith Rudolph
- Institut für Allgemeine Zoologie und Tierphysiologie, Universität Jena Jena, Germany
| | - Katrin Gerstmann
- Institut für Allgemeine Zoologie und Tierphysiologie, Universität Jena Jena, Germany
| | - Geraldine Zimmer
- Institut für Allgemeine Zoologie und Tierphysiologie, Universität Jena Jena, Germany
| | - André Steinecke
- Institut für Allgemeine Zoologie und Tierphysiologie, Universität Jena Jena, Germany
| | - Annika Döding
- Institut für Allgemeine Zoologie und Tierphysiologie, Universität Jena Jena, Germany
| | - Jürgen Bolz
- Institut für Allgemeine Zoologie und Tierphysiologie, Universität Jena Jena, Germany
| |
Collapse
|
24
|
Copy number variation distribution in six monozygotic twin pairs discordant for schizophrenia. Twin Res Hum Genet 2014; 17:108-20. [PMID: 24556202 DOI: 10.1017/thg.2014.6] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
We have evaluated copy number variants (CNVs) in six monozygotic twin pairs discordant for schizophrenia. The data from Affymetrix® Human SNP 6.0 arrays™ were analyzed using Affymetrix® Genotyping Console™, Partek® Genomics Suite™, PennCNV, and Golden Helix SVS™. This yielded both program-specific and overlapping results. Only CNVs called by Affymetrix Genotyping Console, Partek Genomics Suite, and PennCNV were used in further analysis. This analysis included an assessment of calls in each of the six twin pairs towards identification of unique CNVs in affected and unaffected co-twins. Real time polymerase chain reaction (PCR) experiments confirmed one CNV loss at 7q11.21 that was found in the affected patient but not in the unaffected twin. The results identified CNVs and genes that were previously implicated in mental abnormalities in four of the six twin pairs. It included PYY (twin pairs 1 and 5), EPHA3 (twin pair 3), KIAA1211L (twin pair 4), and GPR139 (twin pair 5). They represent likely candidate genes and CNVs for the discordance of four of the six monozygotic twin pairs for this heterogeneous neurodevelopmental disorder. An explanation for these differences is ontogenetic de novo events that differentiate in the monozygotic twins during development.
Collapse
|
25
|
Xi HQ, Wu XS, Wei B, Chen L. Eph receptors and ephrins as targets for cancer therapy. J Cell Mol Med 2014; 16:2894-909. [PMID: 22862837 PMCID: PMC4393718 DOI: 10.1111/j.1582-4934.2012.01612.x] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Accepted: 07/13/2012] [Indexed: 12/15/2022] Open
Abstract
Eph receptor tyrosine kinases and their ephrin ligands are involved in various signalling pathways and mediate critical steps of a wide variety of physiological and pathological processes. Increasing experimental evidence demonstrates that both Eph receptor and ephrin ligands are overexpressed in a number of human tumours, and are associated with tumour growth, invasiveness and metastasis. In this regard, the Eph/ephrin system provides the foundation for potentially exciting new targets for anticancer therapies for Eph-expressing tumours. The purpose of this review is to outline current advances in the role of Eph receptors and ephrin ligands in cancer, and to discuss novel therapeutic approaches of anticancer therapies.
Collapse
Affiliation(s)
- Hong-Qing Xi
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | | | | | | |
Collapse
|
26
|
Hu Y, Li S, Jiang H, Li MT, Zhou JW. Ephrin-B2/EphA4 forward signaling is required for regulation of radial migration of cortical neurons in the mouse. Neurosci Bull 2014; 30:425-32. [PMID: 24477991 DOI: 10.1007/s12264-013-1404-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 06/08/2013] [Indexed: 11/26/2022] Open
Abstract
Postmitotic neurons in the neocortex migrate to appropriate positions and form layered structures of nascent cortex during brain development. The migration of these neurons requires precise control and coordination of a large number of molecules such as axon guidance cues. The Eph-ephrin signaling pathway plays important roles in the development of the nervous system in a wide variety of ways, including cell segregation, axon pathfinding, and neuron migration. However, the role of ephrin-B2/EphA4 signaling in cortical neuron migration remains elusive. Here we demonstrated that ephrin-B2 and its receptor EphA4 were expressed in complementary and overlapping patterns in the developing neocortex. Deletion of the EphA4 gene in the embryonic cerebral cortex resulted in faster migration of cortical neurons, whereas knockdown or overexpression of ephrin-B2 did not alter the normal process of migration. These results suggest that ephrin-B2 forward signaling through EphA4 is required for the precise control of cortical neuron migration.
Collapse
Affiliation(s)
- Yan Hu
- Department of Pharmacology and Proteomics Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | | | | | | | | |
Collapse
|
27
|
Steinecke A, Gampe C, Zimmer G, Rudolph J, Bolz J. EphA/ephrin A reverse signaling promotes the migration of cortical interneurons from the medial ganglionic eminence. Development 2014; 141:460-71. [DOI: 10.1242/dev.101691] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Inhibitory interneurons control the flow of information and synchronization in the cerebral cortex at the circuit level. During embryonic development, multiple subtypes of cortical interneurons are generated in different regions of the ventral telencephalon, such as the medial and caudal ganglionic eminence (MGE and CGE), as well as the preoptic area (POA). These neurons then migrate over long distances towards their cortical target areas. Diverse families of diffusible and cell-bound signaling molecules, including the Eph/ephrin system, regulate and orchestrate interneuron migration. Ephrin A3 and A5, for instance, are expressed at the borders of the pathway of MGE-derived interneurons and prevent these cells from entering inappropriate regions via EphA4 forward signaling. We found that MGE-derived interneurons, in addition to EphA4, also express ephrin A and B ligands, suggesting Eph/ephrin forward and reverse signaling in the same cell. In vitro and in vivo approaches showed that EphA4-induced reverse signaling in MGE-derived interneurons promotes their migration and that this effect is mediated by ephrin A2 ligands. In EphA4 mutant mice, as well as after ephrin A2 knockdown using in utero electroporation, we found delayed interneuron migration at embryonic stages. Thus, besides functions in guiding MGE-derived interneurons to the cortex through forward signaling, here we describe a novel role of the ephrins in driving these neurons to their target via reverse signaling.
Collapse
Affiliation(s)
- André Steinecke
- Universität Jena, Institut für Allgemeine Zoologie und Tierphysiologie, 07743 Jena, Germany
| | - Christin Gampe
- Universität Jena, Institut für Allgemeine Zoologie und Tierphysiologie, 07743 Jena, Germany
| | - Geraldine Zimmer
- Universität Jena, Institut für Allgemeine Zoologie und Tierphysiologie, 07743 Jena, Germany
| | - Judith Rudolph
- Universität Jena, Institut für Allgemeine Zoologie und Tierphysiologie, 07743 Jena, Germany
| | - Jürgen Bolz
- Universität Jena, Institut für Allgemeine Zoologie und Tierphysiologie, 07743 Jena, Germany
| |
Collapse
|
28
|
The role of astrocytes in the regulation of synaptic plasticity and memory formation. Neural Plast 2013; 2013:185463. [PMID: 24369508 PMCID: PMC3867861 DOI: 10.1155/2013/185463] [Citation(s) in RCA: 135] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 10/07/2013] [Accepted: 11/05/2013] [Indexed: 12/22/2022] Open
Abstract
Astrocytes regulate synaptic transmission and play a role in the formation of new memories, long-term potentiation (LTP), and functional synaptic plasticity. Specifically, astroglial release of glutamate, ATP, and cytokines likely alters the survivability and functioning of newly formed connections. Among these pathways, regulation of glutamate appears to be most directly related to the promotion of LTP, which is highly dependent on the synchronization of synaptic receptors through the regulation of excitatory postsynaptic potentials. Moreover, regulation of postsynaptic glutamate receptors, particularly AMPA receptors, is dependent on signaling by ATP synthesized in astrocytes. Finally, cytokine signaling is also implicated in regulating LTP, but is likely most important in plasticity following tissue damage. Despite the role of these signaling factors in regulating LTP and functional plasticity, an integrative model of these factors has not yet been elucidated. In this review, we seek to summarize the current body of evidence on astrocytic mechanisms for regulation of LTP and functional plasticity, and provide an integrative model of the processes.
Collapse
|
29
|
Fang Y, Cho KS, Tchedre K, Lee SW, Guo C, Kinouchi H, Fried S, Sun X, Chen DF. Ephrin-A3 suppresses Wnt signaling to control retinal stem cell potency. Stem Cells 2013; 31:349-59. [PMID: 23165658 DOI: 10.1002/stem.1283] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Accepted: 10/23/2012] [Indexed: 11/09/2022]
Abstract
The ciliary epithelium (CE) of adult mammals has been reported to provide a source of retinal stem cells (RSCs) that can give rise to all retinal cell types in vitro. A recent study, however, suggests that CE-derived cells possess properties of pigmented ciliary epithelial cells and display little neurogenic potential. Here we show that the neurogenic potential of CE-derived cells is negatively regulated by ephrin-A3, which is upregulated in the CE of postnatal mice and presents a strong prohibitory niche for adult RSCs. Addition of ephrin-A3 inhibits proliferation of CE-derived RSCs and increases pigment 349 cell 359. In contrast, absence of ephrin-A3 promotes proliferation and increases expression of neural progenitor cell markers and photoreceptor progeny. The negative effects of ephrin-A3 on CE-derived RSCs are mediated through activation of an EphA4 receptor and suppression of Wnt3a/β-catenin signaling. Together, our data suggest that CE-derived RSCs contain the intrinsic machinery to generate photoreceptors and other retinal neurons, while the CE of adult mice expresses negative regulators that prohibit the proliferation and neural differentiation of RSCs. Manipulating ephrin and Wnt/β-catenin signaling may, thus, represent a viable approach in activating the endogenous neurogenic potential of CE-derived RSCs for treating photoreceptor damage and retinal degenerative disorders.
Collapse
Affiliation(s)
- Yuan Fang
- Department of Ophthalmology and Visual Science, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Geng D, Kang L, Su Y, Jia J, Ma J, Li S, Du J, Cui H. Protective effects of EphB2 on Aβ1-42 oligomer-induced neurotoxicity and synaptic NMDA receptor signaling in hippocampal neurons. Neurochem Int 2013; 63:283-90. [PMID: 23831214 DOI: 10.1016/j.neuint.2013.06.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Revised: 06/14/2013] [Accepted: 06/15/2013] [Indexed: 12/19/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized pathologically by the abnormal deposition of extracellular amyloid-β (Aβ) oligomers. However, the nature and precise mechanism of the toxicity of Aβ oligomers are not clearly understood. Aβ oligomers have been previously shown to cause a major loss of EphB2, a member of the EphB family of receptor tyrosine kinases. To determine the effect of EphB2 on Aβ oligomer-induced neurotoxicity and the underlying molecular mechanisms, we examined the EphB2 gene in cultured hippocampal neurons. Using a cellular model of AD, Aβ1-42 oligomers were confirmed to induce neurotoxicity in a time-dependent manner and result in a major decrease of EphB2. EphB2 overexpression could prevent the neurotoxicity of hippocampal neurons from exposure to Aβ1-42 oligomers for 1h. Further analysis revealed that EphB2 overexpression increased synaptic NR1 and NR2B expression in Aβ1-42 oligomer-treated neurons. Moreover, EphB2 overexpression prevented Aβ1-42 oligomer-induced downregulation of dephosphorylated p38 MAPK and phosphorylated CREB. Together, these results suggest that EphB2 is a factor which protects hippocampal neurons against the toxicity of Aβ1-42 oligomers, and we infer that the protection of EphB2 is achieved by increasing the synaptic NMDA receptor level and downstream p38 MAPK and CREB signaling in hippocampal neurons. This study provides new molecular insights into the neuroprotective effect of EphB2 and highlights its potential therapeutic role in the management of AD.
Collapse
Affiliation(s)
- Dandan Geng
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Prenatal inhibition of the tryptophan–kynurenine pathway alters synaptic plasticity and protein expression in the rat hippocampus. Brain Res 2013; 1504:1-15. [DOI: 10.1016/j.brainres.2013.01.031] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Revised: 01/08/2013] [Accepted: 01/18/2013] [Indexed: 11/19/2022]
|
32
|
Axon guidance mechanisms for establishment of callosal connections. Neural Plast 2013; 2013:149060. [PMID: 23533817 PMCID: PMC3595665 DOI: 10.1155/2013/149060] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Revised: 12/30/2012] [Accepted: 01/21/2013] [Indexed: 01/03/2023] Open
Abstract
Numerous studies have investigated the formation of interhemispheric connections which are involved in high-ordered functions of the cerebral cortex in eutherian animals, including humans. The development of callosal axons, which transfer and integrate information between the right/left hemispheres and represent the most prominent commissural system, must be strictly regulated. From the beginning of their growth, until reaching their targets in the contralateral cortex, the callosal axons are guided mainly by two environmental cues: (1) the midline structures and (2) neighboring? axons. Recent studies have shown the importance of axona guidance by such cues and the underlying molecular mechanisms. In this paper, we review these guidance mechanisms during the development of the callosal neurons. Midline populations express and secrete guidance molecules, and "pioneer" axons as well as interactions between the medial and lateral axons are also involved in the axon pathfinding of the callosal neurons. Finally, we describe callosal dysgenesis in humans and mice, that results from a disruption of these navigational mechanisms.
Collapse
|
33
|
Activation of EphA receptors mediates the recruitment of the adaptor protein Slap, contributing to the downregulation of N-methyl-D-aspartate receptors. Mol Cell Biol 2013; 33:1442-55. [PMID: 23382070 DOI: 10.1128/mcb.01618-12] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Regulation of the activity of N-methyl-d-aspartate receptors (NMDARs) at glutamatergic synapses is essential for certain forms of synaptic plasticity underlying learning and memory and is also associated with neurotoxicity and neurodegenerative diseases. In this report, we investigate the role of Src-like adaptor protein (Slap) in NMDA receptor signaling. We present data showing that in dissociated neuronal cultures, activation of ephrin (Eph) receptors by chimeric preclustered eph-Fc ligands leads to recruitment of Slap and NMDA receptors at the sites of Eph receptor activation. Interestingly, our data suggest that prolonged activation of EphA receptors is as efficient in recruiting Slap and NMDA receptors as prolonged activation of EphB receptors. Using established heterologous systems, we examined whether Slap is an integral part of NMDA receptor signaling. Our results showed that Slap does not alter baseline activity of NMDA receptors and does not affect Src-dependent potentiation of NMDA receptor currents in Xenopus oocytes. We also demonstrate that Slap reduces excitotoxic cell death triggered by activation of NMDARs in HEK293 cells. Finally, we present evidence showing reduced levels of NMDA receptors in the presence of Slap occurring in an activity-dependent manner, suggesting that Slap is part of a mechanism that homeostatically modulates the levels of NMDA receptors.
Collapse
|
34
|
Gael B, Julie D, Shao Z, Xuan Z, Ren Y, Xu J, Arbez N, Mauger G, Bruban J, Georgakopoulos A, Shioi J, Robakis NK. Presenilin mediates neuroprotective functions of ephrinB and brain-derived neurotrophic factor and regulates ligand-induced internalization and metabolism of EphB2 and TrkB receptors. Neurobiol Aging 2013; 34:499-510. [PMID: 22475621 PMCID: PMC3394882 DOI: 10.1016/j.neurobiolaging.2012.02.024] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Revised: 02/21/2012] [Accepted: 02/24/2012] [Indexed: 10/28/2022]
Abstract
Activation of EphB receptors by ephrinB (efnB) ligands on neuronal cell surface regulates important functions, including neurite outgrowth, axonal guidance, and synaptic plasticity. Here, we show that efnB rescues primary cortical neuronal cultures from necrotic cell death induced by glutamate excitotoxicity and that this function depends on EphB receptors. Importantly, the neuroprotective function of the efnB/EphB system depends on presenilin 1 (PS1), a protein that plays crucial roles in Alzheimer's disease (AD) neurodegeneration. Furthermore, absence of one PS1 allele results in significantly decreased neuroprotection, indicating that both PS1 alleles are necessary for full expression of the neuroprotective activity of the efnB/EphB system. We also show that the ability of brain-derived neurotrophic factor (BDNF) to protect neuronal cultures from glutamate-induced cell death depends on PS1. Neuroprotective functions of both efnB and BDNF, however, were independent of γ-secretase activity. Absence of PS1 decreases cell surface expression of neuronal TrkB and EphB2 without affecting total cellular levels of the receptors. Furthermore, PS1-knockout neurons show defective ligand-dependent internalization and decreased ligand-induced degradation of TrkB and Eph receptors. Our data show that PS1 mediates the neuroprotective activities of efnB and BDNF against excitotoxicity and regulates surface expression and ligand-induced metabolism of their cognate receptors. Together, our observations indicate that PS1 promotes neuronal survival by regulating neuroprotective functions of ligand-receptor systems.
Collapse
Affiliation(s)
- Barthet Gael
- Center for Molecular Biology and Genetics of Neurodegeneration, Departments of Psychiatry and Neuroscience, Mount Sinai School of Medicine, New York, NY 10029
| | - Dunys Julie
- Center for Molecular Biology and Genetics of Neurodegeneration, Departments of Psychiatry and Neuroscience, Mount Sinai School of Medicine, New York, NY 10029
| | - Zhiping Shao
- Center for Molecular Biology and Genetics of Neurodegeneration, Departments of Psychiatry and Neuroscience, Mount Sinai School of Medicine, New York, NY 10029
| | - Zhao Xuan
- Center for Molecular Biology and Genetics of Neurodegeneration, Departments of Psychiatry and Neuroscience, Mount Sinai School of Medicine, New York, NY 10029
| | - Ymin Ren
- Center for Molecular Biology and Genetics of Neurodegeneration, Departments of Psychiatry and Neuroscience, Mount Sinai School of Medicine, New York, NY 10029
| | - Jindong Xu
- Center for Molecular Biology and Genetics of Neurodegeneration, Departments of Psychiatry and Neuroscience, Mount Sinai School of Medicine, New York, NY 10029
| | - Nicolas Arbez
- Center for Molecular Biology and Genetics of Neurodegeneration, Departments of Psychiatry and Neuroscience, Mount Sinai School of Medicine, New York, NY 10029
| | - Gweltas Mauger
- Center for Molecular Biology and Genetics of Neurodegeneration, Departments of Psychiatry and Neuroscience, Mount Sinai School of Medicine, New York, NY 10029
| | - Julien Bruban
- Center for Molecular Biology and Genetics of Neurodegeneration, Departments of Psychiatry and Neuroscience, Mount Sinai School of Medicine, New York, NY 10029
| | - Anastasios Georgakopoulos
- Center for Molecular Biology and Genetics of Neurodegeneration, Departments of Psychiatry and Neuroscience, Mount Sinai School of Medicine, New York, NY 10029
| | - Junichi Shioi
- Center for Molecular Biology and Genetics of Neurodegeneration, Departments of Psychiatry and Neuroscience, Mount Sinai School of Medicine, New York, NY 10029
| | - Nikolaos K. Robakis
- Center for Molecular Biology and Genetics of Neurodegeneration, Departments of Psychiatry and Neuroscience, Mount Sinai School of Medicine, New York, NY 10029
| |
Collapse
|
35
|
Glutamate-receptor-like molecule GluRδ2 involved in synapse formation at parallel fiber-Purkinje neuron synapses. THE CEREBELLUM 2012; 11:71-7. [PMID: 20387025 DOI: 10.1007/s12311-010-0170-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Glutamate-receptor-like molecule δ2 (GluRδ2, GluD2) has been classified as an ionotropic glutamate receptor subunit. It is selectively expressed on the postsynaptic membrane at parallel fiber-Purkinje neuron synapses in the cerebellum. Mutant mice deficient in GluRδ2 show impaired synaptic plasticity, the decrease in the number of parallel fiber-Purkinje neuron synapses, multiple innervation of climbing fibers on a Purkinje neuron, and defects in motor control and learning. Thus, GluRδ2 plays crucial roles in the cerebellar function. Recent studies on GluRδ2 have shown that it has synaptogenic activity. GluRδ2 expressed in a non-neuronal cell induces presynaptic differentiation of granule neurons in a co-culture preparation. This synaptogenic activity depends on an extracellular N-terminal leucine/isoleucine/valine binding protein-like domain of GluRδ2. GluRδ2 plays critical roles in formation, maturation, and/or maintenance of granule neuron-Purkinje neuron synapses.
Collapse
|
36
|
Pinato G, Cojoc D, Lien LT, Ansuini A, Ban J, D'Este E, Torre V. Less than 5 Netrin-1 molecules initiate attraction but 200 Sema3A molecules are necessary for repulsion. Sci Rep 2012; 2:675. [PMID: 22997549 PMCID: PMC3447186 DOI: 10.1038/srep00675] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Accepted: 09/03/2012] [Indexed: 11/09/2022] Open
Abstract
Guidance molecules, such as Sema3A or Netrin-1, induce growth cone (GC) repulsion or attraction. In order to determine the speed of action and efficiency of these guidance cues we developed an experimental procedure to deliver controlled amounts of these molecules. Lipid vesicles encapsulating 10-10(4) molecules of Sema3A or Netrin-1 were manipulated with high spatial and temporal resolution by optical tweezers and their photolysis triggered by laser pulses. Guidance molecules released from the vesicles diffused and reached the GC membrane in a few seconds. Following their arrival, GCs retracted or grew in 20-120 s. By determining the number of guidance molecules trapped inside vesicles and estimating the fraction of guidance molecules reaching the GC, we show that the arrival of less than 5 Netrin-1 molecules on the GC membrane is sufficient to induce growth. In contrast, the arrival of about 200 Sema3A molecules is necessary to induce filopodia repulsion.
Collapse
Affiliation(s)
- Giulietta Pinato
- Istituto Officina dei Materiali (IOM-CNR), Area Science Park, Trieste, Italy.
| | | | | | | | | | | | | |
Collapse
|
37
|
Astrocytes regulate adult hippocampal neurogenesis through ephrin-B signaling. Nat Neurosci 2012; 15:1399-406. [PMID: 22983209 PMCID: PMC3458152 DOI: 10.1038/nn.3212] [Citation(s) in RCA: 169] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Accepted: 08/09/2012] [Indexed: 12/18/2022]
Abstract
Neurogenesis in the adult hippocampus involves activation of quiescent neural stem cells (NSCs) to yield transiently amplifying NSCs and progenitors, and ultimately neurons that affect learning and memory. This process is tightly controlled by microenvironmental cues, though few endogenous factors are known to regulate neuronal differentiation. While astrocytes have been implicated, their role in juxtacrine (i.e. cell-cell contact-dependent) signaling within NSC niches has not been investigated. We show that ephrin-B2 presented from rodent hippocampal astrocytes regulates neurogenesis in vivo. Furthermore, clonal analysis in NSC fate-mapping studies reveals a novel role for ephrin-B2 in instructing neuronal differentiation. Additionally, ephrin-B2 signaling, transduced by EphB4 receptors on NSCs, activates β-catenin in vitro and in vivo independent of Wnt signaling and upregulates proneural transcription factors. Ephrin-B2+ astrocytes thus promote neuronal differentiation of adult NSCs through juxtacrine signaling, findings that advance our understanding of adult neurogenesis and may have future regenerative medicine implications.
Collapse
|
38
|
GABA through the ages: regulation of cortical function and plasticity by inhibitory interneurons. Neural Plast 2012; 2012:892784. [PMID: 22792496 PMCID: PMC3390141 DOI: 10.1155/2012/892784] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Revised: 03/30/2012] [Accepted: 03/30/2012] [Indexed: 11/17/2022] Open
Abstract
Inhibitory interneurons comprise only about 20% of cortical neurons and thus constitute a clear minority compared to the vast number of excitatory projection neurons. They are, however, an influential minority with important roles in cortical maturation, function, and plasticity. In this paper, we will highlight the functional importance of cortical inhibition throughout brain development, starting with the embryonal formation of the cortex, proceeding by the regulation of sensory cortical plasticity in adulthood, and finishing with the GABA involvement in sensory information processing in old age.
Collapse
|
39
|
Forrest CM, Khalil OS, Pisar M, Smith RA, Darlington LG, Stone TW. Prenatal activation of Toll-like receptors-3 by administration of the viral mimetic poly(I:C) changes synaptic proteins, N-methyl-D-aspartate receptors and neurogenesis markers in offspring. Mol Brain 2012; 5:22. [PMID: 22681877 PMCID: PMC3496691 DOI: 10.1186/1756-6606-5-22] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Accepted: 04/25/2012] [Indexed: 12/30/2022] Open
Abstract
Background There is mounting evidence for a neurodevelopmental basis for disorders such as autism and schizophrenia, in which prenatal or early postnatal events may influence brain development and predispose the young to develop these and related disorders. We have now investigated the effect of a prenatal immune challenge on brain development in the offspring. Pregnant rats were treated with the double-stranded RNA polyinosinic:polycytidylic acid (poly(I:C); 10 mg/kg) which mimics immune activation occurring after activation of Toll-like receptors-3 (TLR3) by viral infection. Injections were made in late gestation (embryonic days E14, E16 and E18), after which parturition proceeded naturally and the young were allowed to develop up to the time of weaning at postnatal day 21 (P21). The brains of these animals were then removed to assess the expression of 13 different neurodevelopmental molecules by immunoblotting. Results Measurement of cytokine levels in the maternal blood 5 hours after an injection of poly(I:C) showed significantly increased levels of monocyte chemoattractant protein-1 (MCP-1), confirming immune activation. In the P21 offspring, significant changes were detected in the expression of GluN1 subunits of NMDA receptors, with no difference in GluN2A or GluN2B subunits or the postsynaptic density protein PSD-95 and no change in the levels of the related small GTPases RhoA or RhoB, or the NMDA receptor modulator EphA4. Among presynaptic molecules, a significant increase in Vesicle Associated Membrane Protein-1 (VAMP-1; synaptobrevin) was seen, with no change in synaptophysin or synaptotagmin. Proliferating Cell Nuclear Antigen (PCNA), as well as the neurogenesis marker doublecortin were unchanged, although Sox-2 levels were increased, suggesting possible changes in the rate of new cell differentiation. Conclusions The results reveal the induction by prenatal poly(I:C) of selective molecular changes in the brains of P21 offspring, affecting primarily molecules associated with neuronal development and synaptic transmission. These changes may contribute to the behavioural abnormalities that have been reported in adult animals after exposure to poly(I:C) and which resemble symptoms seen in schizophrenia and related disorders.
Collapse
Affiliation(s)
- Caroline M Forrest
- Institute for Neuroscience and Psychology, University of Glasgow, West Medical Building, Glasgow, G12 8QQ, UK
| | | | | | | | | | | |
Collapse
|
40
|
Yu J, Xiang M, Wu H, Shen C. Ephrin A2 protein expression in the regeneration and plasticity of cochlear hair cells in chicken following kanamycin ototoxicity. Neural Regen Res 2012; 7:714-8. [PMID: 25745469 PMCID: PMC4347014 DOI: 10.3969/j.issn.1673-5374.2012.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2011] [Accepted: 12/24/2011] [Indexed: 11/18/2022] Open
Abstract
The results from this study showed that the thresholds of brainstem auditory-evoked potentials peak following 10 successive days of intramuscular injection of Roman chickens with kanamycin, starting 3 days after birth. Fluorescence immunohistochemistry analysis revealed few ganglion cells positively labeled for Ephrin A2 in the cochlea of experimental chickens from 2 days before until 7 days after the last kanamycin injection. The number of Ephrin A2-positive ganglion cell bodies was increased at 15 days after the last injection and was similar to that in normal chickens at 30 days following the cessation of kanamycin treatment. These experimental findings indicate that Ephrin A2 protein expression in the acoustic ganglia is synchronized with the connection damage and regeneration of cochlear hair cells after kanamycin exposure. Ephrin A2 may play an important role in the regeneration and plasticity of cochlear hair cells in the chick cochlea following kanamycin ototoxicity.
Collapse
Affiliation(s)
- Jia Yu
- Department of OtlaryngoIogy & Head and Neck Surgery of Xinhua Hospital, Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Mingliang Xiang
- Department of OtlaryngoIogy & Head and Neck Surgery of Xinhua Hospital, Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Hao Wu
- Department of OtlaryngoIogy & Head and Neck Surgery of Xinhua Hospital, Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Chenling Shen
- Department of OtlaryngoIogy & Head and Neck Surgery of Xinhua Hospital, Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| |
Collapse
|
41
|
Bidirectional ephrinB3/EphA4 signaling mediates the segregation of medial ganglionic eminence- and preoptic area-derived interneurons in the deep and superficial migratory stream. J Neurosci 2012; 31:18364-80. [PMID: 22171039 DOI: 10.1523/jneurosci.4690-11.2011] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The integration of interneuron subtypes into specific microcircuits is essential for proper cortical function. Understanding to what extent interneuron diversity is regulated and maintained during development might help to reveal the principles that govern their role as synchronizing elements as well as causes for dysfunction. Particular interneuron subtypes are generated in a temporally regulated manner in the medial ganglionic eminence (MGE), the caudal ganglionic eminence, and the preoptic area (POA) of the basal telencephalon. Long-range tangential migration from their site of origin to cortical targets is orchestrated by a variety of attractive, repulsive, membrane-bound, and secreted signaling molecules, to establish the critical balance of inhibition and excitation. It remains unknown whether interneurons deriving from distinct domains are predetermined to migrate in particular routes and whether this process underlies cell type-specific regulation. We found that POA- and MGE-derived cortical interneurons migrate within spatially segregated corridors. EphrinB3, expressed in POA-derived interneurons traversing the superficial route, acts as a repellent signal for deeply migrating interneurons born in the MGE, which is mediated by EphA4 forward signaling. In contrast, EphA4 induces repulsive ephrinB3 reverse signaling in interneurons generated in the POA, restricting this population to the superficial path. Perturbation of this bidirectional ephrinB3/EphA4 signaling in vitro and in vivo leads to a partial intermingling of cells in these segregated migratory pathways. Thus, we conclude that cell contact-mediated bidirectional ephrinB3/EphA4 signaling mediates the sorting of MGE- and POA-derived interneurons in the deep and superficial migratory stream.
Collapse
|
42
|
Abstract
The corpus callosum, composed of callosal axons, is the largest structure among commissural connections in eutherian animals. Axon pathfinding of callosal neurons has been shown to be guided by intermediate targets, such as midline glial structures. However, it has not yet been understood completely how axon-axon interactions, another major mechanism for axon pathfinding, are involved in the pathfinding of callosal neurons. Here, we show that callosal axons from the medial and lateral regions of the mouse cerebral cortex pass through the dorsal and ventral parts, respectively, of the corpus callosum. Using an explant culture system, we observed that the axons from the medial and lateral cortices were segregated from each other in vitro, and that this segregation was attenuated by inhibition of EphA3 signaling. We also found that knockdown of EphA3, which is preferentially expressed in the lateral cortex, resulted in disorganized segregation of the callosal axons and disrupted axon pathfinding in vivo. These results together suggest the role of axonal segregation in the corpus callosum, mediated at least in part by EphA3, in correct pathfinding of callosal neurons.
Collapse
|
43
|
Meier C, Anastasiadou S, Knöll B. Ephrin-A5 suppresses neurotrophin evoked neuronal motility, ERK activation and gene expression. PLoS One 2011; 6:e26089. [PMID: 22022520 PMCID: PMC3191169 DOI: 10.1371/journal.pone.0026089] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2011] [Accepted: 09/19/2011] [Indexed: 12/20/2022] Open
Abstract
During brain development, growth cones respond to attractive and repulsive axon guidance cues. How growth cones integrate guidance instructions is poorly understood. Here, we demonstrate a link between BDNF (brain derived neurotrophic factor), promoting axonal branching and ephrin-A5, mediating axonal repulsion via Eph receptor tyrosine kinase activation. BDNF enhanced growth cone filopodial dynamics and neurite branching of primary neurons. We show that ephrin-A5 antagonized this BDNF-evoked neuronal motility. BDNF increased ERK phosphorylation (P-ERK) and nuclear ERK entry. Ephrin-A5 suppressed BDNF-induced ERK activity and might sequester P-ERK in the cytoplasm. Neurotrophins are well established stimulators of a neuronal immediate early gene (IEG) response. This is confirmed in this study by e.g. c-fos, Egr1 and Arc upregulation upon BDNF application. This BDNF-evoked IEG response required the transcription factor SRF (serum response factor). Notably, ephrin-A5 suppressed a BDNF-evoked neuronal IEG response, suggesting a role of Eph receptors in modulating gene expression. In opposite to IEGs, long-term ephrin-A5 application induced cytoskeletal gene expression of tropomyosin and actinin. To uncover specific Eph receptors mediating ephrin-As impact on neurotrophin signaling, EphA7 deficient mice were analyzed. In EphA7 deficient neurons alterations in growth cone morphology were observed. However, ephrin-A5 still counteracted neurotrophin signaling suggesting that EphA7 is not required for ephrin and BDNF crosstalk. In sum, our data suggest an interaction of ephrin-As and neurotrophin signaling pathways converging at ERK signaling and nuclear gene activity. As ephrins are involved in development and function of many organs, such modulation of receptor tyrosine kinase signaling and gene expression by Ephs might not be limited to the nervous system.
Collapse
Affiliation(s)
- Christin Meier
- Neuronal Gene Expression Laboratory, Department of Molecular Biology, Interfaculty Institute for Cell Biology, Eberhard-Karls-University Tübingen, Tübingen, Germany
| | - Sofia Anastasiadou
- Neuronal Gene Expression Laboratory, Department of Molecular Biology, Interfaculty Institute for Cell Biology, Eberhard-Karls-University Tübingen, Tübingen, Germany
| | - Bernd Knöll
- Neuronal Gene Expression Laboratory, Department of Molecular Biology, Interfaculty Institute for Cell Biology, Eberhard-Karls-University Tübingen, Tübingen, Germany
- * E-mail:
| |
Collapse
|
44
|
Abstract
In the present review, we look back at the recent history of GWAS (genome-wide association studies) in AD (Alzheimer's disease) and integrate the major findings with current knowledge of biological processes and pathways. These topics are essential for the development of animal models, which will be fundamental to our complete understanding of AD.
Collapse
|
45
|
Kim SK, Jeon JW, Park JJ, Cha JM, Joo KR, Lee JI, Chung JH, Shin DH, Shin HP. Associations of EPHB1 polymorphisms with hepatocellular carcinoma in the Korean population. Hum Immunol 2011; 72:916-20. [PMID: 21763378 DOI: 10.1016/j.humimm.2011.06.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2010] [Revised: 06/10/2011] [Accepted: 06/27/2011] [Indexed: 11/29/2022]
Abstract
Hepatocellular carcinoma (HCC) is a type of hypervascular tumor, and angiogenesis is important for HCC tumor growth. Eph receptor B1 (EPHB1), a member of the Eph family, mediates embryonic vascular system development and adult angiogenesis. This receptor may be involved in carcinogenesis of the digestive tract. Our aim was to examine the relationships between EPHB1 polymorphisms and HCC in the Korean population. Genomic DNA was extracted from 182 patients with HCC and 266 healthy subjects. EPHB1 polymorphisms were determined by polymerase chain reaction and direct sequencing. Multiple logistic regression models (log-additive, dominant, and recessive models) were used for odds ratios, 95% confidence intervals, and p values. Five polymorphisms (rs11929692, rs7644369, rs6776570, rs3821502, and rs6766459) of the EPHB1 gene and alleles of 2 polymorphisms (rs1502174 and rs9877457) were associated with HCC (p < 0.05 for both). Our results suggest that EPHB1 polymorphisms may be associated with susceptibility to HCC in the Korean population.
Collapse
Affiliation(s)
- Su Kang Kim
- Department of Pharmacology, Kyung Hee University School of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Hollingworth P, Harold D, Sims R, Gerrish A, Lambert JC, Carrasquillo MM, Abraham R, Hamshere ML, Pahwa JS, Moskvina V, Dowzell K, Jones N, Stretton A, Thomas C, Richards A, Ivanov D, Widdowson C, Chapman J, Lovestone S, Powell J, Proitsi P, Lupton MK, Brayne C, Rubinsztein DC, Gill M, Lawlor B, Lynch A, Brown KS, Passmore PA, Craig D, McGuinness B, Todd S, Holmes C, Mann D, Smith AD, Beaumont H, Warden D, Wilcock G, Love S, Kehoe PG, Hooper NM, Vardy ERLC, Hardy J, Mead S, Fox NC, Rossor M, Collinge J, Maier W, Jessen F, Rüther E, Schürmann B, Heun R, Kölsch H, van den Bussche H, Heuser I, Kornhuber J, Wiltfang J, Dichgans M, Frölich L, Hampel H, Gallacher J, Hüll M, Rujescu D, Giegling I, Goate AM, Kauwe JSK, Cruchaga C, Nowotny P, Morris JC, Mayo K, Sleegers K, Bettens K, Engelborghs S, De Deyn PP, Van Broeckhoven C, Livingston G, Bass NJ, Gurling H, McQuillin A, Gwilliam R, Deloukas P, Al-Chalabi A, Shaw CE, Tsolaki M, Singleton AB, Guerreiro R, Mühleisen TW, Nöthen MM, Moebus S, Jöckel KH, Klopp N, Wichmann HE, Pankratz VS, Sando SB, Aasly JO, Barcikowska M, Wszolek ZK, Dickson DW, Graff-Radford NR, Petersen RC, et alHollingworth P, Harold D, Sims R, Gerrish A, Lambert JC, Carrasquillo MM, Abraham R, Hamshere ML, Pahwa JS, Moskvina V, Dowzell K, Jones N, Stretton A, Thomas C, Richards A, Ivanov D, Widdowson C, Chapman J, Lovestone S, Powell J, Proitsi P, Lupton MK, Brayne C, Rubinsztein DC, Gill M, Lawlor B, Lynch A, Brown KS, Passmore PA, Craig D, McGuinness B, Todd S, Holmes C, Mann D, Smith AD, Beaumont H, Warden D, Wilcock G, Love S, Kehoe PG, Hooper NM, Vardy ERLC, Hardy J, Mead S, Fox NC, Rossor M, Collinge J, Maier W, Jessen F, Rüther E, Schürmann B, Heun R, Kölsch H, van den Bussche H, Heuser I, Kornhuber J, Wiltfang J, Dichgans M, Frölich L, Hampel H, Gallacher J, Hüll M, Rujescu D, Giegling I, Goate AM, Kauwe JSK, Cruchaga C, Nowotny P, Morris JC, Mayo K, Sleegers K, Bettens K, Engelborghs S, De Deyn PP, Van Broeckhoven C, Livingston G, Bass NJ, Gurling H, McQuillin A, Gwilliam R, Deloukas P, Al-Chalabi A, Shaw CE, Tsolaki M, Singleton AB, Guerreiro R, Mühleisen TW, Nöthen MM, Moebus S, Jöckel KH, Klopp N, Wichmann HE, Pankratz VS, Sando SB, Aasly JO, Barcikowska M, Wszolek ZK, Dickson DW, Graff-Radford NR, Petersen RC, Alzheimer's Disease Neuroimaging Initiative, van Duijn CM, Breteler MMB, Ikram MA, DeStefano AL, Fitzpatrick AL, Lopez O, Launer LJ, Seshadri S, CHARGE consortium, Berr C, Campion D, Epelbaum J, Dartigues JF, Tzourio C, Alpérovitch A, Lathrop M, EADI1 consortium, Feulner TM, Friedrich P, Riehle C, Krawczak M, Schreiber S, Mayhaus M, Nicolhaus S, Wagenpfeil S, Steinberg S, Stefansson H, Stefansson K, Snaedal J, Björnsson S, Jonsson PV, Chouraki V, Genier-Boley B, Hiltunen M, Soininen H, Combarros O, Zelenika D, Delepine M, Bullido MJ, Pasquier F, Mateo I, Frank-Garcia A, Porcellini E, Hanon O, Coto E, Alvarez V, Bosco P, Siciliano G, Mancuso M, Panza F, Solfrizzi V, Nacmias B, Sorbi S, Bossù P, Piccardi P, Arosio B, Annoni G, Seripa D, Pilotto A, Scarpini E, Galimberti D, Brice A, Hannequin D, Licastro F, Jones L, Holmans PA, Jonsson T, Riemenschneider M, Morgan K, Younkin SG, Owen MJ, O'Donovan M, Amouyel P, Williams J. Common variants at ABCA7, MS4A6A/MS4A4E, EPHA1, CD33 and CD2AP are associated with Alzheimer's disease. Nat Genet 2011; 43:429-35. [PMID: 21460840 PMCID: PMC3084173 DOI: 10.1038/ng.803] [Show More Authors] [Citation(s) in RCA: 1593] [Impact Index Per Article: 113.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2010] [Accepted: 03/10/2011] [Indexed: 11/09/2022]
Abstract
We sought to identify new susceptibility loci for Alzheimer's disease through a staged association study (GERAD+) and by testing suggestive loci reported by the Alzheimer's Disease Genetic Consortium (ADGC) in a companion paper. We undertook a combined analysis of four genome-wide association datasets (stage 1) and identified ten newly associated variants with P ≤ 1 × 10(-5). We tested these variants for association in an independent sample (stage 2). Three SNPs at two loci replicated and showed evidence for association in a further sample (stage 3). Meta-analyses of all data provided compelling evidence that ABCA7 (rs3764650, meta P = 4.5 × 10(-17); including ADGC data, meta P = 5.0 × 10(-21)) and the MS4A gene cluster (rs610932, meta P = 1.8 × 10(-14); including ADGC data, meta P = 1.2 × 10(-16)) are new Alzheimer's disease susceptibility loci. We also found independent evidence for association for three loci reported by the ADGC, which, when combined, showed genome-wide significance: CD2AP (GERAD+, P = 8.0 × 10(-4); including ADGC data, meta P = 8.6 × 10(-9)), CD33 (GERAD+, P = 2.2 × 10(-4); including ADGC data, meta P = 1.6 × 10(-9)) and EPHA1 (GERAD+, P = 3.4 × 10(-4); including ADGC data, meta P = 6.0 × 10(-10)).
Collapse
Affiliation(s)
- Paul Hollingworth
- Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Neurosciences and Mental Health Research Institute, Department of Psychological Medicine and Neurology, School of Medicine, Cardiff University, Cardiff, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Braskie MN, Ringman JM, Thompson PM. Neuroimaging measures as endophenotypes in Alzheimer's disease. Int J Alzheimers Dis 2011; 2011:490140. [PMID: 21547229 PMCID: PMC3087508 DOI: 10.4061/2011/490140] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Revised: 01/08/2011] [Accepted: 02/07/2011] [Indexed: 01/06/2023] Open
Abstract
Late onset Alzheimer's disease (AD) is moderately to highly heritable. Apolipoprotein E allele ε4 (APOE4) has been replicated consistently as an AD risk factor over many studies, and recently confirmed variants in other genes such as CLU, CR1, and PICALM each increase the lifetime risk of AD. However, much of the heritability of AD remains unexplained. AD is a complex disease that is diagnosed largely through neuropsychological testing, though neuroimaging measures may be more sensitive for detecting the incipient disease stages. Difficulties in early diagnosis and variable environmental contributions to the disease can obscure genetic relationships in traditional case-control genetic studies. Neuroimaging measures may be used as endophenotypes for AD, offering a reliable, objective tool to search for possible genetic risk factors. Imaging measures might also clarify the specific mechanisms by which proposed risk factors influence the brain.
Collapse
Affiliation(s)
- Meredith N Braskie
- Laboratory of Neuro Imaging, Department of Neurology, UCLA School of Medicine, 635 Charles Young Drive South, Suite 225, Los Angeles, CA 90095, USA
| | | | | |
Collapse
|
48
|
Bi C, Yue X, Zhou R, Plummer MR. EphA activation overrides the presynaptic actions of BDNF. J Neurophysiol 2011; 105:2364-74. [PMID: 21411563 DOI: 10.1152/jn.00564.2010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The adult pattern of neural connectivity is shaped by repulsive and attractive factors, many of which are modulated by activity. Although much is known about the actions of these factors when studied in isolation, little is known about how they interact. To address this question, we examined the effects of sequential or coapplication of brain-derived neurotrophic factor (BDNF) and Fc-conjugated ephrin-A5 or EphA5 in cultured embryonic hippocampal neurons. BDNF promotes neurite outgrowth and synapse formation, and when applied acutely, it elicits an increase in ongoing synaptic activity. Members of the ephrin family of ligands and receptors can be repulsive and prevent formation of synaptic contacts. Acute exposure to either ephrin-A5-Fc or EphA5-Fc transiently enhanced synaptic activity when applied alone, but when applied prior to BDNF, they dramatically reduced the electrophysiological effects of the neurotrophin. Conversely, BDNF had no effect on subsequently applied ephrin-A5-Fc or EphA5-Fc. Consistent with this, ephrin-A5-Fc also prevented BDNF-induced activation of p42/44 MAPK. The effect of ephrin-A5-Fc appears to be presynaptic, as it prevented the BDNF-induced increase in spontaneous miniature postsynaptic current frequency, whereas EphA5-Fc did not. These results suggest that these factors can be categorized differently, with the contact-mediated activation of EphA receptors by ephrin-A5 overriding the diffusion-mediated effect of BDNF.
Collapse
Affiliation(s)
- Caixia Bi
- Rutgers University, Department of Cell Biology & Neuroscience, Nelson Laboratories, 604 Allison Rd., Piscataway, NJ 08854-8082, USA
| | | | | | | |
Collapse
|
49
|
Schang AL, Ngô-Muller V, Bleux C, Granger A, Chenut MC, Loudes C, Magre S, Counis R, Cohen-Tannoudji J, Laverrière JN. GnRH receptor gene expression in the developing rat hippocampus: transcriptional regulation and potential roles in neuronal plasticity. Endocrinology 2011; 152:568-80. [PMID: 21123436 DOI: 10.1210/en.2010-0840] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
In the pituitary of mammals, the GnRH receptor (GnRHR) plays a primary role in the control of reproductive function. It is further expressed in the hippocampus, where its function, however, is not well defined. By quantitative RT-PCR analyses, we demonstrate herein that the onset of GnRHR gene (Gnrhr) expression in the rat hippocampus was unexpectedly delayed as compared to the pituitary and only occurred after birth. Using a previously described transgenic mouse model bearing the human placental alkaline phosphatase reporter gene under the control of the rat Gnrhr promoter, we established a positive correlation between the temporal pattern of Gnrhr mRNA levels and promoter activity in the hippocampal formation. The gradual appearance of human placental alkaline phosphatase transgene expression occurred simultaneously in the hippocampus and interconnected structures such as the lateral septum and the amygdala, coinciding with the establishment of hippocampo-septal projections. Analysis of transcription factors together with transient transfection assays in hippocampal neurons indicated that the combinatorial code governing the hippocampus-specific expression of the Gnrhr is distinct from the pituitary, likely involving transactivating factors such as NUR77, cyclic AMP response element binding protein, and Finkel-Biskis-Jinkins murine osteosarcoma virus oncogene homolog. A silencing transcription factor acting via the -3255/-1135 promoter region of the Gnrhr may be responsible for the transcriptional repression observed around birth. Finally, GnRH directly stimulated via activation of its receptor the expression of several marker genes of neuronal plasticity such as Egr1, synaptophysin, and spinophilin in hippocampal primary cultures, suggesting a role for GnRHR in neuronal plasticity. Further characterization of these mechanisms may help unravel important functions of GnRH/GnRHR signaling in the brain.
Collapse
Affiliation(s)
- Anne-Laure Schang
- Université Paris-Diderot Paris 7, Equipe d'Accueil Conventionnée Centre National de la Recherche Scientifique 4413, Physiologie de l'Axe Gonadotrope, Bâtiment Buffon, 4 rue MA Lagroua Weill-Hallé 75205 Paris cedex 13, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Hara Y, Nomura T, Yoshizaki K, Frisén J, Osumi N. Impaired hippocampal neurogenesis and vascular formation in ephrin-A5-deficient mice. Stem Cells 2010; 28:974-83. [PMID: 20474079 DOI: 10.1002/stem.427] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Neurogenesis occurs throughout the life in the mammalian brain. The hippocampal dentate gyrus (DG) is one of the major regions of the adult neurogenesis, where neural stem/progenitor cells continuously generate new granule neurons, although molecular mechanisms underlying generation and maintenance of newly born neurons are still elusive. Here we show that ephrin-A5, a ligand for Eph receptor tyrosine kinases, plays multiple roles in both neurogenesis and vascular formation in the adult hippocampus. In mice lacking ephrin-A5 function, cell proliferation and survival of newborn neurons were severely reduced in the hippocampus DG. Furthermore, ephrin-A5-deficient mice exhibited altered distribution of EphA4 receptor in the vascular endothelial cells and increased narrower capillaries in the hippocampus DG. EphA/ephrin-A signaling thus plays crucial roles in the establishment and/or maintenance of the brain vascular system, as an essential constituent of the adult neurogenic niche.
Collapse
Affiliation(s)
- Yoshinobu Hara
- Division of Developmental Neuroscience, Core Center for Neuroscience, ART, Tohoku University School of Medicine, Sendai, Japan
| | | | | | | | | |
Collapse
|