Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters










Publication year range
1.
J Virol ; 98(3): e0183823, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38426726

ABSTRACT

Nipah virus (NiV) is a highly lethal, zoonotic Henipavirus (HNV) that causes respiratory and neurological signs and symptoms in humans. Similar to other paramyxoviruses, HNVs mediate entry into host cells through the concerted actions of two surface glycoproteins: a receptor-binding protein (RBP) that mediates attachment and a fusion glycoprotein (F) that triggers fusion in an RBP-dependent manner. NiV uses ephrin-B2 (EFNB2) and ephrin-B3 (EFNB3) as entry receptors. Ghana virus (GhV), a novel HNV identified in a Ghanaian bat, uses EFNB2 but not EFNB3. In this study, we employ a structure-informed approach to identify receptor-interfacing residues and systematically introduce GhV-RBP residues into a NiV-RBP backbone to uncover the molecular determinants of EFNB3 usage. We reveal two regions that severely impair EFNB3 binding by NiV-RBP and EFNB3-mediated entry by NiV pseudotyped viral particles. Further analyses uncovered two-point mutations (NiVN557SGhV and NiVY581TGhV) pivotal for this phenotype. Moreover, we identify NiV interaction with Y120 of EFNB3 as important for the usage of this receptor. Beyond these EFNB3-related findings, we reveal two domains that restrict GhV binding of EFNB2, confirm the HNV-head as an immunodominant target for polyclonal and monoclonal antibodies, and describe putative epitopes for GhV- and NiV-specific monoclonal antibodies. Cumulatively, the work presented here generates useful reagents and tools that shed insight to residues important for NiV usage of EFNB3, reveal regions critical for GhV binding of EFNB2, and describe putative HNV antibody-binding epitopes. IMPORTANCE: Hendra virus and Nipah virus (NiV) are lethal, zoonotic Henipaviruses (HNVs) that cause respiratory and neurological clinical features in humans. Since their initial outbreaks in the 1990s, several novel HNVs have been discovered worldwide, including Ghana virus. Additionally, there is serological evidence of zoonotic transmission, lending way to concerns about future outbreaks. HNV infection of cells is mediated by the receptor-binding protein (RBP) and the Fusion protein (F). The work presented here identifies NiV RBP amino acids important for the usage of ephrin-B3 (EFNB3), a receptor highly expressed in neurons and predicted to be important for neurological clinical features caused by NiV. This study also characterizes epitopes recognized by antibodies against divergent HNV RBPs. Together, this sheds insight to amino acids critical for HNV receptor usage and antibody binding, which is valuable for future studies investigating determinants of viral pathogenesis and developing antibody therapies.


Subject(s)
Henipavirus Infections , Henipavirus , Receptors, Virus , Humans , Amino Acids/genetics , Antibodies, Monoclonal/metabolism , Carrier Proteins/metabolism , Ephrin-B3/genetics , Ephrin-B3/chemistry , Ephrin-B3/metabolism , Epitopes/genetics , Epitopes/metabolism , Ghana , Hendra Virus/metabolism , Henipavirus/classification , Henipavirus/genetics , Henipavirus/metabolism , Mutagenesis , Nipah Virus/metabolism , Viral Envelope Proteins/genetics , Virus Internalization , Receptors, Virus/metabolism
2.
J Orthop Surg Res ; 16(1): 499, 2021 Aug 14.
Article in English | MEDLINE | ID: mdl-34389038

ABSTRACT

BACKGROUND: Heterotopic ossification (HO) represents pathological lesions that refer to the development of heterotopic bone in extraskeletal tissues around joints. This study investigates the genetic characteristics of bone marrow mesenchymal stem cells (BMSCs) from HO tissues and explores the potential pathways involved in this ailment. METHODS: Gene expression profiles (GSE94683) were obtained from the Gene Expression Omnibus (GEO), including 9 normal specimens and 7 HO specimens, and differentially expressed genes (DEGs) were identified. Then, protein-protein interaction (PPI) networks and Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were performed for further analysis. RESULTS: In total, 275 DEGs were differentially expressed, of which 153 were upregulated and 122 were downregulated. In the biological process (BP) category, the majority of DEGs, including EFNB3, UNC5C, TMEFF2, PTH2, KIT, FGF13, and WISP3, were intensively enriched in aspects of cell signal transmission, including axon guidance, negative regulation of cell migration, peptidyl-tyrosine phosphorylation, and cell-cell signaling. Moreover, KEGG analysis indicated that the majority of DEGs, including EFNB3, UNC5C, FGF13, MAPK10, DDIT3, KIT, COL4A4, and DKK2, were primarily involved in the mitogen-activated protein kinase (MAPK) signaling pathway, Ras signaling pathway, phosphatidylinositol-3-kinase/protein kinase B (PI3K/Akt) signaling pathway, and Wnt signaling pathway. Ten hub genes were identified, including CX3CL1, CXCL1, ADAMTS3, ADAMTS16, ADAMTSL2, ADAMTSL3, ADAMTSL5, PENK, GPR18, and CALB2. CONCLUSIONS: This study presented novel insight into the pathogenesis of HO. Ten hub genes and most of the DEGs intensively involved in enrichment analyses may be new candidate targets for the prevention and treatment of HO in the future.


Subject(s)
ADAMTS Proteins/genetics , Ephrin-B3/genetics , Extracellular Matrix Proteins/genetics , Membrane Proteins/genetics , Neoplasm Proteins/genetics , Ossification, Heterotopic , Phosphatidylinositol 3-Kinases/genetics , Transcriptome , ADAMTS Proteins/chemistry , Computational Biology , Ephrin-B3/chemistry , Extracellular Matrix Proteins/chemistry , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , Membrane Proteins/chemistry , Neoplasm Proteins/chemistry , Ossification, Heterotopic/genetics , Phosphatidylinositol 3-Kinases/chemistry , Phosphatidylinositol 3-Kinases/metabolism , Protein Interaction Maps
3.
Sci Rep ; 10(1): 18162, 2020 10 23.
Article in English | MEDLINE | ID: mdl-33097800

ABSTRACT

PEA3 transcription factor subfamily is present in a variety of tissues with branching morphogenesis, and play a particularly significant role in neural circuit formation and specificity. Many target genes in axon guidance and cell-cell adhesion pathways have been identified for Pea3 transcription factor (but not for Erm or Er81); however it was not so far clear whether all Pea3 subfamily members regulate same target genes, or whether there are unique targets for each subfamily member that help explain the exclusivity and specificity of these proteins in neuronal circuit formation. In this study, using transcriptomics and qPCR analyses in SH-SY5Y neuroblastoma cells, hypothalamic and hippocampal cell line, we have identified cell type-specific and subfamily member-specific targets for PEA3 transcription factor subfamily. While Pea3 upregulates transcription of Sema3D and represses Sema5B, for example, Erm and Er81 upregulate Sema5A and Er81 regulates Unc5C and Sema4G while repressing EFNB3 in SH-SY5Y neuroblastoma cells. We furthermore present a molecular model of how unique sites within the ETS domain of each family member can help recognize specific target motifs. Such cell-context and member-specific combinatorial expression profiles help identify cell-cell and cell-extracellular matrix communication networks and how they establish specific connections.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental , Neuronal Outgrowth/genetics , Proto-Oncogene Proteins c-ets/metabolism , Transcription Factors/metabolism , Axons , Cell Line, Tumor , Cell Movement/genetics , Ephrin-B3/genetics , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Gene Expression Profiling , Hippocampus/cytology , Humans , Hypothalamus/cytology , Molecular Dynamics Simulation , Nerve Tissue Proteins/genetics , Neurons/cytology , Neurons/metabolism , Oligonucleotide Array Sequence Analysis , Protein Domains , Real-Time Polymerase Chain Reaction , Semaphorins/genetics , Transcriptional Activation
4.
Nat Commun ; 10(1): 5220, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31745086

ABSTRACT

The hepatopancreatic ductal (HPD) system connects the intrahepatic and intrapancreatic ducts to the intestine and ensures the afferent transport of the bile and pancreatic enzymes. Yet the molecular and cellular mechanisms controlling their differentiation and morphogenesis into a functional ductal system are poorly understood. Here, we characterize HPD system morphogenesis by high-resolution microscopy in zebrafish. The HPD system differentiates from a rod of unpolarized cells into mature ducts by de novo lumen formation in a dynamic multi-step process. The remodeling step from multiple nascent lumina into a single lumen requires active cell intercalation and myosin contractility. We identify key functions for EphB/EphrinB signaling in this dynamic remodeling step. Two EphrinB ligands, EphrinB1 and EphrinB2a, and two EphB receptors, EphB3b and EphB4a, control HPD morphogenesis by remodeling individual ductal compartments, and thereby coordinate the morphogenesis of this multi-compartment ductal system.


Subject(s)
Bile Ducts/metabolism , Ephrin-B1/metabolism , Hepatopancreas/metabolism , Receptors, Eph Family/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Animals, Genetically Modified , Bile Ducts/embryology , Cell Differentiation/genetics , Ephrin-B1/genetics , Ephrin-B3/genetics , Ephrin-B3/metabolism , Gene Expression Profiling , Hepatopancreas/embryology , Ligands , Morphogenesis/genetics , Mutation , Protein Binding , Receptors, Eph Family/genetics , Signal Transduction/genetics , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics
5.
Proc Natl Acad Sci U S A ; 116(41): 20707-20715, 2019 10 08.
Article in English | MEDLINE | ID: mdl-31548390

ABSTRACT

Cedar virus (CedV) is a bat-borne henipavirus related to Nipah virus (NiV) and Hendra virus (HeV), zoonotic agents of fatal human disease. CedV receptor-binding protein (G) shares only ∼30% sequence identity with those of NiV and HeV, although they can all use ephrin-B2 as an entry receptor. We demonstrate that CedV also enters cells through additional B- and A-class ephrins (ephrin-B1, ephrin-A2, and ephrin-A5) and report the crystal structure of the CedV G ectodomain alone and in complex with ephrin-B1 or ephrin-B2. The CedV G receptor-binding site is structurally distinct from other henipaviruses, underlying its capability to accommodate additional ephrin receptors. We also show that CedV can enter cells through mouse ephrin-A1 but not human ephrin-A1, which differ by 1 residue in the key contact region. This is evidence of species specific ephrin receptor usage by a henipavirus, and implicates additional ephrin receptors in potential zoonotic transmission.


Subject(s)
Ephrin-B1/metabolism , Ephrin-B2/metabolism , Ephrin-B3/metabolism , Henipavirus Infections/virology , Henipavirus/physiology , Receptors, Virus/metabolism , Viral Envelope Proteins/chemistry , Animals , Cell Fusion , Ephrin-B1/genetics , Ephrin-B2/genetics , Ephrin-B3/genetics , Henipavirus Infections/genetics , Henipavirus Infections/metabolism , Humans , Mice , Mutation , Protein Binding , Protein Conformation , Receptors, Virus/genetics , Species Specificity , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Virus Internalization
6.
Cereb Cortex ; 29(4): 1644-1658, 2019 04 01.
Article in English | MEDLINE | ID: mdl-29912395

ABSTRACT

The delicate balance of excitation and inhibition is crucial for proper function of the cerebral cortex, relying on the accurate number and subtype composition of inhibitory gamma-aminobutyric (GABA)-expressing interneurons. Various intrinsic and extrinsic factors precisely orchestrate their multifaceted development including the long-range migration from the basal telencephalon to cortical targets as well as interneuron survival throughout the developmental period. Particularly expressed guidance receptors were described to channel the migration of cortical interneurons deriving from the medial ganglionic eminence (MGE) and the preoptic area (POA) along distinct routes. Hence, unveiling the regulatory genetic networks controlling subtype-specific gene expression profiles is key to understand interneuron-specific developmental programs and to reveal causes for associated disorders. In contrast to MGE-derived interneurons, little is known about the transcriptional networks in interneurons born in the POA. Here, we provide first evidence for the LIM-homeobox transcription factor LHX1 as a crucial key player in the post-mitotic development of POA-derived cortical interneurons. By transcriptional regulation of related genes, LHX1 modulates their survival as well as the subtype-specific expression of guidance receptors of the Eph/ephrin family, thereby affecting directional migration and layer distribution in the adult cortex.


Subject(s)
Cerebral Cortex/growth & development , Interneurons/physiology , LIM-Homeodomain Proteins/physiology , Preoptic Area/growth & development , Transcription Factors/physiology , Animals , Cell Movement , Cell Survival , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Ephrin-B3/genetics , Ephrin-B3/physiology , Gene Expression Regulation, Developmental , Interneurons/cytology , Interneurons/metabolism , LIM-Homeodomain Proteins/genetics , Mice, Inbred C57BL , Mice, Transgenic , Preoptic Area/cytology , Preoptic Area/metabolism , Receptor, EphA4/genetics , Receptor, EphA4/physiology , Transcription Factors/genetics
7.
Sci Rep ; 8(1): 14497, 2018 09 27.
Article in English | MEDLINE | ID: mdl-30262919

ABSTRACT

Several members of the EPH kinase family and their ligands are involved in blood pressure regulation, and such regulation is often sex- or sex hormone-dependent, based on animal and human genetic studies. EPHB6 gene knockout (KO) in mice leads to hypertension in castrated males but not in un-manipulated KO males or females. To assess whether this finding in mice is relevant to human hypertension, we conducted a human genetic study for the association of EPHB6 and its two ligands, EFNB1 and EFNB3, with hypertension in hypogonadic patients. Seven hundred and fifty hypertensive and 750 normotensive Han Chinese patients, all of whom were hypogonadic, were genotyped for single nucleotide polymorphisms (SNPs) within the regions of the genes, plus an additional 50 kb 5' of the genes for EPHB6, EFNB1 and EFNB3. An imputed insertion/deletion polymorphism, rs35530071, was found to be associated with hypertension at p-values below the Bonferroni-corrected significance level of 0.0024. This marker is located 5' upstream of the EFNB3 gene start site. Previous animal studies showed that while male EFNB3 gene knockout mice were normotensive, castration of these mice resulted in hypertension, corroborating the results of the human genetic study. Considering the significant associations of EFNB3 SNPs with hypertension in hypogonadic males and supporting evidence from castrated EFNB3 KO mice, we conclude that loss-of-function variants of molecules in the EPHB6 signaling pathway in the presence of testosterone are protective against hypertension in humans.


Subject(s)
Ephrin-B1/genetics , Ephrin-B3/genetics , Hypertension/genetics , Hypogonadism/genetics , Polymorphism, Single Nucleotide , Receptors, Eph Family/genetics , Adult , Animals , Asian People , China , Humans , Hypertension/pathology , Hypertension/physiopathology , Hypogonadism/pathology , Hypogonadism/physiopathology , Male , Mice , Mice, Knockout , Middle Aged
8.
Cell Death Dis ; 9(1): 7, 2018 01 08.
Article in English | MEDLINE | ID: mdl-29311672

ABSTRACT

Damage to the cerebrovascular network is a major contributor to dysfunction in patients suffering from traumatic brain injury (TBI). Vessels are composed of lumen-forming endothelial cells that associate closely with both glial and neuronal units to establish a functional blood-brain barrier (BBB). Under normal physiological conditions, these vascular units play important roles in central nervous system (CNS) homeostasis by delivering oxygen and nutrients while filtering out molecules and cells that could be harmful; however, after TBI this system is disrupted. Here, we describe a novel role for a class of receptors, called dependence receptors, in regulating vessel stability and BBB integrity after CCI injury in mice. Specifically, we identified that EphB3 receptors function as a pro-apoptotic dependence receptor in endothelial cells (ECs) that contributes to increased BBB damage after CCI injury. In the absence of EphB3, we observed increased endothelial cell survival, reduced BBB permeability and enhanced interactions of astrocyte-EC membranes. Interestingly, the brain's response to CCI injury is to reduce EphB3 levels and its ligand ephrinB3; however, the degree and timing of those reductions limit the protective response of the CNS. We conclude that EphB3 is a negative regulator of cell survival and BBB integrity that undermine tissue repair, and represents a protective therapeutic target for TBI patients.


Subject(s)
Blood-Brain Barrier/metabolism , Brain Injuries, Traumatic/pathology , Receptor, EphB3/metabolism , Animals , Brain Injuries, Traumatic/metabolism , CD11b Antigen/metabolism , Cell Death/drug effects , Disease Models, Animal , Endothelial Cells/cytology , Endothelial Cells/metabolism , Ephrin-B3/genetics , Ephrin-B3/metabolism , Gene Expression/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Leukocyte Common Antigens/metabolism , Male , Mice , Mice, Inbred C57BL , Receptor, EphB3/genetics , Signal Transduction
9.
Sci Rep ; 7: 44114, 2017 03 08.
Article in English | MEDLINE | ID: mdl-28272517

ABSTRACT

EPH kinases and their ligands, ephrins (EFNs), have vital and diverse biological functions. We recently reported that Efnb3 gene deletion results in hypertension in female but not male mice. These data suggest that EFNB3 regulates blood pressure in a sex- and sex hormone-dependent way. In the present study, we conducted a human genetic study to assess the association of EFNB3 single nucleotide polymorphisms with human hypertension risks, using 3,448 patients with type 2 diabetes from the ADVANCE study (Action in Diabetes and Vascular Disease: Peterax and Diamicron MR Controlled Evaluation). We have observed significant association between 2 SNPs in the 3' untranslated region or within the adjacent region just 3' of the EFNB3 gene with hypertension, corroborating our findings from the mouse model. Thus, our investigation has shown that EFNB3 is a hypertension risk gene in certain individuals.


Subject(s)
Diabetes Mellitus, Type 2/complications , Ephrin-B3/genetics , Genetic Predisposition to Disease , Hypertension/genetics , Polymorphism, Single Nucleotide , Aged , Female , Genetic Association Studies , Genotype , Humans , Hypertension/etiology , Male , Risk Factors
10.
Sci Rep ; 6: 38976, 2016 12 12.
Article in English | MEDLINE | ID: mdl-27941904

ABSTRACT

Several erythropoietin-producing hepatocellular receptor B family (EPHB) and their ligands, ephrinBs (EFNBs), are involved in blood pressure regulation in animal models. We selected 528 single nucleotide polymorphisms (SNPs) within the genes of EPHB6, EFNB2, EFNB3 and GRIP1 in the EPH/EFN signalling system to query the International Blood Pressure Consortium dataset. A SNP within the glutamate receptor interacting protein 1 (GRIP1) gene presented a p-value of 0.000389, approaching the critical p-value of 0.000302, for association with diastolic blood pressure of 60,396 individuals. According to echocardiography, we found that Efnb3 gene knockout mice showed enhanced constriction in the carotid arteries. In vitro studies revealed that in mouse vascular smooth muscle cells, siRNA knockdown of GRIP1, which is in the EFNB3 reverse signalling pathway, resulted in increased contractility of these cells. These data suggest that molecules in the EPHB/EFNB signalling pathways, specifically EFNB3 and GRIP1, are involved blood pressure regulation.


Subject(s)
Blood Pressure , Carrier Proteins/metabolism , Ephrin-B3/metabolism , Muscle Contraction , Muscle, Smooth, Vascular/metabolism , Nerve Tissue Proteins/metabolism , Animals , Carrier Proteins/genetics , Ephrin-B3/genetics , Humans , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Polymorphism, Single Nucleotide , Signal Transduction
11.
Oncotarget ; 7(37): 60332-60347, 2016 Sep 13.
Article in English | MEDLINE | ID: mdl-27533087

ABSTRACT

Ephrin receptors (Ephs) are reported to control metastatic signaling of non-small cell lung cancer (NSCLC) and other tumors. Here we show for the first time that blocking expression of the Eph ligand Ephrin B3 inhibits NSCLC cell migration and invasion. We demonstrate that Ephrin B3 directly binds the EphAs EphA2, EphA3, EphA4, and EphA5. EphA2 Ser897 was previously shown to drive migration propensity of tumor cells and our study reveals that EphA2 stays phosphorylated on Ser897 in the Ephrin B3/EphA2 complex in NSCLC cells of different histology. Moreover, we report that within such Ephrin B3/EphA2 complex both Akt Ser 129 and p38MAPK are found indicating a potential to drive migration/proliferation. We also found the EMT marker E-cadherin expression to be maintained or increased upon Ephrin B3 blockade in NSCLC cells. Expression of Ephrin B3 was furthermore analyzed in a cohort of NSCLC stage IA-IB cases (n=200) alongside EphA2 and Ephrin A1. We found that Ephrin B3 was concomitantly expressed with EphA2 and Ephrin A1 with higher Ephrin B3 levels found in non-squamous than in squamous tumors, whereas EphA2 was higher expressed in well-differentiated than in low-differentiated tumors. In the entire NSCLC cohort, Ephrin B3 expression was not linked to patient survival, whereas a high EphA2 expression was associated with improved survival (p=0.03). In conclusion, we show that blocking Ephrin B3 expression inhibits NSCLC proliferation-, migration- and invasion capacity which calls for further studies on interference with Ephrin B3 as a possible therapeutic avenue in this tumor malignancy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Cell Movement/genetics , Ephrin-B3/genetics , Lung Neoplasms/genetics , Receptors, Eph Family/genetics , A549 Cells , Aged , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Ephrin-A1/genetics , Ephrin-A1/metabolism , Ephrin-B3/metabolism , Female , Humans , Kaplan-Meier Estimate , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Invasiveness , Protein Binding , RNA Interference , Receptor, EphA2/genetics , Receptor, EphA2/metabolism , Receptors, Eph Family/metabolism
12.
Neurosci Lett ; 622: 61-6, 2016 05 27.
Article in English | MEDLINE | ID: mdl-27102143

ABSTRACT

Axon regeneration as a critical step in nerve repairing and remodeling after peripheral nerve injury relies on regulation of gene expression. MicroRNAs are emerging to be important epigenetic regulators of gene expression to control axon regeneration. Here we used a novel in vivo electroporation approach to transfect microRNA-210 (miR-210) or siRNAs to adult mice dorsal root ganglion (DRG) neurons, measured the axon length 3days after sciatic nerve crush or dissociated DRG cultures in vitro to detect the effect of miR-210 in sensory axon regeneration. Importantly, we found that miR-210 overexpression could promote sensory axon regeneration and inhibit apoptsosis by ephrin-A3 (EFNA3). In addition, inhibition of endogenous miR-210 in DRG neurons impaired axon regeneration in vitro and in vivo, the regulatory effect of miR-210 was mediated by increased expression of EFNA3 because downregulation of EFNA3 fully rescued axon regeneration. We thus demonstrate that miR-210 is a new physiological regulator of sensory axon regeneration, and EFNA3 may be the functional target of miR-210. We conclude that miR-210 may play an important role in sensory axon regeneration.


Subject(s)
Axons/physiology , MicroRNAs/metabolism , Sensory Receptor Cells/physiology , Animals , Apoptosis , Cells, Cultured , Electroporation , Ephrin-B3/genetics , Ephrin-B3/metabolism , Ganglia, Spinal/cytology , Mice , MicroRNAs/genetics , Nerve Regeneration , Primary Cell Culture , RNA, Small Interfering/genetics , Sciatic Nerve/injuries , Sensory Receptor Cells/cytology
13.
Nat Commun ; 7: 11096, 2016 Mar 24.
Article in English | MEDLINE | ID: mdl-27008987

ABSTRACT

Innate emotion response to environmental stimuli is a fundamental brain function that is controlled by specific neural circuits. Dysfunction of early emotional circuits may lead to neurodevelopmental disorders such as autism and schizophrenia. However, how the functional circuits are formed to prime initial emotional behaviours remain elusive. We reveal here using gene-targeted mutations an essential role for ephrin-B3 ligand-like activity in the development of innate fear in the neonatal brain. We further demonstrate that ephrin-B3 controls axon targeting and coordinates spinogenesis and neuronal activity within the amygdala. The morphological and behavioural abnormalities in ephrin-B3 mutant mice are rescued by conditional knock-in of wild-type ephrin-B3 during the critical period when axon targeting and fear responses are initiated. Our results thus define a key axonal molecule that participates in the wiring of amygdala circuits and helps bring about fear emotion during the important adolescence period.


Subject(s)
Amygdala/metabolism , Axons/metabolism , Ephrin-B3/metabolism , Fear/physiology , Instinct , Neurogenesis , Animals , Cell Nucleus/metabolism , Ephrin-B3/genetics , Mice , Mutation/genetics , Synapses/metabolism , Time Factors
14.
Acta Neuropathol ; 131(2): 281-298, 2016 02.
Article in English | MEDLINE | ID: mdl-26687980

ABSTRACT

Remyelination in multiple sclerosis (MS) lesions often remains incomplete despite the presence of oligodendrocyte progenitor cells (OPCs). Amongst other factors, successful remyelination depends on the phagocytic clearance of myelin debris. However, the proteins in myelin debris that act as potent and selective inhibitors on OPC differentiation and inhibit CNS remyelination remain unknown. Here, we identify the transmembrane signalling protein EphrinB3 as important mediator of this inhibition, using a protein analytical approach in combination with a primary rodent OPC assay. In the presence of EphrinB3, OPCs fail to differentiate. In a rat model of remyelination, infusion of EphrinB3 inhibits remyelination. In contrast, masking EphrinB3 epitopes using antibodies promotes remyelination. Finally, we identify EphrinB3 in MS lesions and demonstrate that MS lesion extracts inhibit OPC differentiation while antibody-mediated masking of EphrinB3 epitopes promotes it. Our findings suggest that EphrinB3 could be a target for therapies aiming at promoting remyelination in demyelinating disease.


Subject(s)
Ephrin-B3/metabolism , Multiple Sclerosis/metabolism , Myelin Sheath/metabolism , Neural Stem Cells/metabolism , Oligodendroglia/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Ephrin-B3/genetics , Epitopes/metabolism , Female , Humans , Macrophages/metabolism , Macrophages/pathology , Mice, Knockout , Multiple Sclerosis/pathology , Myelin Sheath/pathology , Nerve Regeneration/physiology , Neural Stem Cells/pathology , Neurogenesis/physiology , Oligodendroglia/pathology , Random Allocation , Rats, Sprague-Dawley , Receptor, EphA4/metabolism
15.
Nat Neurosci ; 18(11): 1594-605, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26479588

ABSTRACT

Organization of signaling complexes at excitatory synapses by membrane-associated guanylate kinase (MAGUK) proteins regulates synapse development, plasticity, senescence and disease. Post-translational modification of MAGUK family proteins can drive their membrane localization, yet it is unclear how these intracellular proteins are targeted to sites of synaptic contact. Here we show using super-resolution imaging, biochemical approaches and in vivo models that the trans-synaptic organizing protein ephrin-B3 controls the synaptic localization and stability of PSD-95 and links these events to changes in neuronal activity via negative regulation of a newly identified mitogen-associated protein kinase (MAPK)-dependent phosphorylation site on ephrin-B3, Ser332. Unphosphorylated ephrin-B3 was enriched at synapses, and interacted directly with and stabilized PSD-95 at synapses. Activity-induced phosphorylation of Ser332 dispersed ephrin-B3 from synapses, prevented the interaction with PSD-95 and enhanced the turnover of PSD-95. Thus, ephrin-B3 specifies the synaptic localization of PSD-95 and likely links the synaptic stability of PSD-95 to changes in neuronal activity.


Subject(s)
Ephrin-B3/metabolism , Guanylate Kinases/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Neurons/metabolism , Synapses/metabolism , Animals , Cats , Disks Large Homolog 4 Protein , Ephrin-B3/genetics , Female , Guanylate Kinases/genetics , Intracellular Signaling Peptides and Proteins/genetics , Male , Membrane Proteins/genetics , Pregnancy , Protein Processing, Post-Translational/genetics , Rats , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism
16.
Proc Natl Acad Sci U S A ; 112(17): E2156-65, 2015 Apr 28.
Article in English | MEDLINE | ID: mdl-25825759

ABSTRACT

The discovery of African henipaviruses (HNVs) related to pathogenic Hendra virus (HeV) and Nipah virus (NiV) from Southeast Asia and Australia presents an open-ended health risk. Cell receptor use by emerging African HNVs at the stage of host-cell entry is a key parameter when considering the potential for spillover and infection of human populations. The attachment glycoprotein from a Ghanaian bat isolate (GhV-G) exhibits <30% sequence identity with Asiatic NiV-G/HeV-G. Here, through functional and structural analysis of GhV-G, we show how this African HNV targets the same human cell-surface receptor (ephrinB2) as the Asiatic HNVs. We first characterized this virus-receptor interaction crystallographically. Compared with extant HNV-G-ephrinB2 structures, there was significant structural variation in the six-bladed ß-propeller scaffold of the GhV-G receptor-binding domain, but not the Greek key fold of the bound ephrinB2. Analysis revealed a surprisingly conserved mode of ephrinB2 interaction that reflects an ongoing evolutionary constraint among geographically distal and phylogenetically divergent HNVs to maintain the functionality of ephrinB2 recognition during virus-host entry. Interestingly, unlike NiV-G/HeV-G, we could not detect binding of GhV-G to ephrinB3. Comparative structure-function analysis further revealed several distinguishing features of HNV-G function: a secondary ephrinB2 interaction site that contributes to more efficient ephrinB2-mediated entry in NiV-G relative to GhV-G and cognate residues at the very C terminus of GhV-G (absent in Asiatic HNV-Gs) that are vital for efficient receptor-induced fusion, but not receptor binding per se. These data provide molecular-level details for evaluating the likelihood of African HNVs to spill over into human populations.


Subject(s)
Ephrin-B2 , Henipavirus Infections/metabolism , Henipavirus , Viral Proteins , Virus Internalization , Ephrin-B2/chemistry , Ephrin-B2/genetics , Ephrin-B2/metabolism , Ephrin-B3/chemistry , Ephrin-B3/genetics , Ephrin-B3/metabolism , HEK293 Cells , Henipavirus/chemistry , Henipavirus/physiology , Henipavirus Infections/genetics , Humans , Protein Structure, Quaternary , Protein Structure, Secondary , Protein Structure, Tertiary , Structure-Activity Relationship , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism
17.
PLoS Biol ; 12(9): e1001955, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25247423

ABSTRACT

Ephrins and Eph receptors are involved in the establishment of vertebrate tissue boundaries. The complexity of the system is puzzling, however in many instances, tissues express multiple ephrins and Ephs on both sides of the boundary, a situation that should in principle cause repulsion between cells within each tissue. Although co-expression of ephrins and Eph receptors is widespread in embryonic tissues, neurons, and cancer cells, it is still unresolved how the respective signals are integrated into a coherent output. We present a simple explanation for the confinement of repulsion to the tissue interface: Using the dorsal ectoderm-mesoderm boundary of the Xenopus embryo as a model, we identify selective functional interactions between ephrin-Eph pairs that are expressed in partial complementary patterns. The combined repulsive signals add up to be strongest across the boundary, where they reach sufficient intensity to trigger cell detachments. The process can be largely explained using a simple model based exclusively on relative ephrin and Eph concentrations and binding affinities. We generalize these findings for the ventral ectoderm-mesoderm boundary and the notochord boundary, both of which appear to function on the same principles. These results provide a paradigm for how developmental systems may integrate multiple cues to generate discrete local outcomes.


Subject(s)
Ectoderm/metabolism , Gene Expression Regulation, Developmental , Mesoderm/metabolism , Morphogenesis/genetics , Notochord/metabolism , Xenopus laevis/genetics , Animals , Ectoderm/growth & development , Embryo, Nonmammalian , Ephrin-B1/genetics , Ephrin-B1/metabolism , Ephrin-B2/genetics , Ephrin-B2/metabolism , Ephrin-B3/genetics , Ephrin-B3/metabolism , Mesoderm/growth & development , Mice , Notochord/growth & development , Receptor, EphA4/genetics , Receptor, EphA4/metabolism , Receptor, EphB2/genetics , Receptor, EphB2/metabolism , Receptor, EphB4/genetics , Receptor, EphB4/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Xenopus laevis/growth & development , Xenopus laevis/metabolism
18.
Physiol Behav ; 135: 232-6, 2014 08.
Article in English | MEDLINE | ID: mdl-24949848

ABSTRACT

Eph-ephrin signaling is known to be important in directing topographic projections in the afferent auditory pathway, including connections to various subdivisions of the inferior colliculus (IC). The acoustic startle-response (ASR) is a reliable reflexive behavioral response in mammals elicited by an unexpected intense acoustic startle-eliciting stimulus (ES). It is mediated by a sub-cortical pathway that includes the IC. The ASR amplitude can be measured with an accelerometer under the subject and can be decreased in amplitude by presenting a less intense, non-startling stimulus 5-300ms before the ES. This reflexive decrement in ASR is called pre-pulse inhibition (PPI) and indicates that the relatively soft pre-pulse was heard. PPI is a general trait among mammals. Mice have been used recently to study this response and to reveal how genetic mutations affect neural circuits and hence the ASR and PPI. In this experiment, we measured the effect of Eph-ephrin mutations using control mice (C57BL/6J), mice with compromised EphA4 signaling (EphA4(lacZ/+), EphA4(lacZ/lacZ)), and knockout ephrin-B3 mice (ephrin-B3 (+/-, -/-)). Control and EphA4(lacZ/+s)trains showed robust PPI (up to 75% decrement in ASR) to an offset of a 70dB SPL background noise at 50ms before the ES. Ephrin-B3 knockout mice and EphA4 homozygous mutants were only marginally significant in PPI (<25% decrement and <33% decrement, respectively) to the same conditions. This decrement in PPI highlights the importance of ephrin-B3 and EphA4 interactions in ordering auditory behavioral circuits. Thus, different mutations in certain members of the signaling family produce a full range of changes in PPI, from minimal to nearly maximal. This technique can be easily adapted to study other aspects of hearing in a wider range of mutations. Along with ongoing neuroanatomical studies, this allows careful quantification of how the auditory anatomical, physiological and now behavioral phenotype is affected by changes in Eph-ephrin expression and functionality.


Subject(s)
Ephrin-B3/genetics , Prepulse Inhibition/genetics , Receptor, EphA4/genetics , Reflex, Startle/genetics , Acoustic Stimulation , Animals , Ephrin-B3/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, EphA4/metabolism , Signal Transduction/genetics
19.
Cell Death Dis ; 5: e1207, 2014 May 08.
Article in English | MEDLINE | ID: mdl-24810043

ABSTRACT

Eph receptor tyrosine kinases and their membrane-bound ligands, ephrins, have a variety of roles in the developing and adult central nervous system that require direct cell-cell interactions; including regulating axon path finding, cell proliferation, migration and synaptic plasticity. Recently, we identified a novel pro-survival role for ephrins in the adult subventricular zone, where ephrinB3 blocks Eph-mediated cell death during adult neurogenesis. Here, we examined whether EphB3 mediates cell death in the adult forebrain following traumatic brain injury and whether ephrinB3 infusion could limit this effect. We show that EphB3 co-labels with microtubule-associated protein 2-positive neurons in the adult cortex and is closely associated with ephrinB3 ligand, which is reduced following controlled cortical impact (CCI) injury. In the complete absence of EphB3 (EphB3(-/-)), we observed reduced terminal deoxynucleotidyl transferase-dUTP nick end labeling (TUNEL), and functional improvements in motor deficits after CCI injury as compared with wild-type and ephrinB3(-/-) mice. We also demonstrated that EphB3 exhibits dependence receptor characteristics as it is cleaved by caspases and induces cell death, which is not observed in the presence of ephrinB3. Following trauma, infusion of pre-clustered ephrinB3-Fc molecules (eB3-Fc) into the contralateral ventricle reduced cortical infarct volume and TUNEL staining in the cortex, dentate gyrus and CA3 hippocampus of wild-type and ephrinB3(-/-) mice, but not EphB3(-/-) mice. Similarly, application of eB3-Fc improved motor functions after CCI injury. We conclude that EphB3 mediates cell death in the adult cortex through a novel dependence receptor-mediated cell death mechanism in the injured adult cortex and is attenuated following ephrinB3 stimulation.


Subject(s)
Brain Injuries/drug therapy , Brain/drug effects , Ephrin-B3/administration & dosage , Ephrin-B3/metabolism , Neurons/drug effects , Neuroprotective Agents/administration & dosage , Receptor, EphB3/metabolism , Animals , Apoptosis/drug effects , Brain/enzymology , Brain/pathology , Brain/physiopathology , Brain Injuries/enzymology , Brain Injuries/genetics , Brain Injuries/pathology , Brain Injuries/physiopathology , Cell Line, Tumor , Cytoprotection , Disease Models, Animal , Ephrin-B3/deficiency , Ephrin-B3/genetics , HEK293 Cells , Humans , Infusions, Intraventricular , Male , Mice , Mice, Knockout , Motor Activity/drug effects , Nerve Degeneration , Neurons/enzymology , Neurons/pathology , Receptor, EphB3/deficiency , Receptor, EphB3/genetics , Recovery of Function , Rotarod Performance Test , Time Factors , Transfection
20.
Mol Med Rep ; 9(5): 1761-6, 2014 May.
Article in English | MEDLINE | ID: mdl-24604122

ABSTRACT

EphrinB3 may restrict axonal regeneration and recovery following traumatic injury of the adult mammalian central nervous system (CNS). Therefore, inhibition of ephrinB3 expression may enhance the ability of the nervous system to regenerate following damage. In the present study, lentiviral expressing vectors, pGCSIL-GFP, expressing an active small interfering RNA (siRNA) targeting the EphB3 sequence, were used to determine the effect of inhibiting EphB3 on nerve functional recovery and regeneration. Basso-Beattie-Bresnahan (BBB) locomotor scores and growth-associated protein, 43 kDa (GAP43) expression levels were determined at 1, 2, 4 and 8 weeks following transplantation of the siRNA. It was identified that EphB3 mRNA and protein levels in the siRNA group were significantly reduced (P<0.01) in the spine compared with the control group, at four weeks following intraparenchymal administration of the siRNA into the right lumbar. BBB locomotor scores were significantly increased (P<0.05) in siRNA animals compared with the sham group, at 1, 2, 4 and 8 weeks following transplantation of the siRNA. In addition, a significant increase in GAP43 expression was detected in the siRNA group compared with the control group. This preclinical study demonstrates that ephrinB3 silencing contributes to axonal growth regeneration and improves recovery from spinal cord injury (SCI).


Subject(s)
Ephrin-B3/genetics , Gene Silencing , Recovery of Function/genetics , Spinal Cord Injuries/genetics , Spinal Cord Injuries/rehabilitation , Animals , Axons/metabolism , Disease Models, Animal , Down-Regulation/genetics , Female , Hindlimb/metabolism , Hindlimb/physiopathology , Motor Activity , RNA Interference , Rats , Spinal Cord Injuries/etiology
SELECTION OF CITATIONS
SEARCH DETAIL
...