Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 154
Filter
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.
Biochem Pharmacol ; 220: 116004, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38142837

ABSTRACT

Ephrin B3, a member of Eph/ephrin family, contributes to embryogenesis and carcinogenesis, but few studies have suggested whether this ligand has regulatory effect on colitis. This study was to determine whether ephrin B3 played a role in colitis and colonic carcinogenesis. Dextran sodium sulfate (DSS)-induced colitis and azoxymethane (AOM)/DSS-induced colitis-associated carcinogenesis model was established in Efnb3-deficient (Efnb3-/-) mice. Label-free quantitative proteomics were performed to identify the Efnb3-regulated proteins. Our results showed that Efnb3 knock out reduced the symptoms of DSS-induced colitis, such as disease activity index (DAI), inflammatory factors release, and dysfunction of the intestinal barrier. Quantitative proteomics revealed that Efnb3 regulated 95 proteins which clustered in the platelet degranulation, response to elevated platelet cytosolic Ca2+, MAPK signaling for integrins such as ITGB4. Furthermore, ephrin B3 inactived ITGB4/AKT signal pathway and then promoted epithelial barrier dysfunction. Simultaneously, ephrin B3 promoted Gremlin-1/NF-κB signal pathway and thereby increased inflammatory factors release. In addition, the higher level of Efnb3 in colon cancer patients is correlated with worse survival. Efnb3-/- mice exhibited susceptibility to AOM/DSS-induced colorectal cancer. Our finding discovered that Efnb3 played an important role in the development of colitis and colitis-associated colorectal cancer. Efnb3 deficiency improved the intestinal barrier by ITGB4 and suppressed inflammation via Gremlin-1/NF-κB signal pathway, which may provide a novel therapeutic strategy for the treatment of colitis and colitis-associated colorectal cancer.


Subject(s)
Colitis-Associated Neoplasms , Colitis , Colorectal Neoplasms , Humans , Animals , Mice , Ephrin-B3 , NF-kappa B/metabolism , Colitis/chemically induced , Colitis/complications , Colitis/metabolism , Carcinogenesis , Azoxymethane/toxicity , Dextran Sulfate/toxicity , Disease Models, Animal , Mice, Inbred C57BL , Colorectal Neoplasms/metabolism
3.
Infect Genet Evol ; 116: 105516, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37924857

ABSTRACT

Nipah virus (NiV) is a lethal bat-borne zoonotic virus that causes mild to acute respiratory distress and neurological manifestations in humans with a high mortality rate. NiV transmission to humans occurs via consumption of bat-contaminated fruit and date palm sap (DPS), or through direct contact with infected individuals and livestock. Since NiV outbreaks were first reported in pigs from Malaysia and Singapore, non-neutralizing antibodies against NiV attachment Glycoprotein (G) have also been detected in a few domestic mammals. NiV infection is initiated after NiV G binds to the host cell receptors Ephrin-B2 and Ephrin-B3. In this study, we assessed the degree of NiV host tropism in domestic and peridomestic mammals commonly found in Bangladesh that may be crucial in the transmission of NiV by serving as intermediate hosts. We carried out a protein-protein docking analysis of NiV G complexes (n = 52) with Ephrin-B2 and B3 of 13 domestic and peridomestic species using bioinformatics tools. Protein models were generated by homology modelling and the structures were validated for model quality. The different protein-protein complexes in this study were stable, and their binding affinity (ΔG) scores ranged between -8.0 to -19.1 kcal/mol. NiV Bangladesh (NiV-B) strain displayed stronger binding to Ephrin receptors, especially with Ephrin-B3 than the NiV Malaysia (NiV-M) strain, correlating with the observed higher pathogenicity of NiV-B strains. From the docking result, we found that Ephrin receptors of domestic rat (R. norvegicus) had a higher binding affinity for NiV G, suggesting greater susceptibility to NiV infections compared to other study species. Investigations for NiV exposure to domestic/peridomestic animals will help us knowing more the possible role of rats and other animals as intermediate hosts of NiV and would improve future NiV outbreak control and prevention in humans and domestic animals.


Subject(s)
Chiroptera , Henipavirus Infections , Nipah Virus , Animals , Rats , Ephrin-B2/genetics , Ephrin-B2/chemistry , Ephrin-B2/metabolism , Ephrin-B3/chemistry , Ephrin-B3/metabolism , Glycoproteins/metabolism , Receptors, Cell Surface/metabolism , Receptors, Eph Family/metabolism , Swine , Virus Attachment
4.
Altern Ther Health Med ; 29(3): 160-165, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36735714

ABSTRACT

Context: Gastric cancer (GC) remains one of the most prevalent malignancies worldwide, and no effective cure exists for advanced GC. Clinicians believe that molecularly targeted therapy through PCGs may replace surgery, radiotherapy, and other treatments as a breakthrough in curing malignancies. Objective: The study intended to examine the impact of aberrant expression of the protein-coding genes (PCGs) associated with regulatory T cells on the prognosis of patients with gastric cancer (GC). Design: The research team performed a genetic study through research of genetic data in online databases. Setting: The study took place at Zhongda Hospital. Outcome Measures: The research team selected a publicly available dataset, genetic suppressor element 109476 (GSE109476), from the Gene Expression Omnibus (GEO) database for differential gene analysis, gene ontology (GO) analysis, and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis to screen for PCGs associated with regulatory T cells as well as the Gene Expression Profiling Interactive Analysis (GEPIA) database with the Kaplan-Meier Plotter database to analyze the expression of the above PCGs in GC and the prognostic impact on GC. Results: The GEO2R analysis found 315 differentially expressed PCGs in GSE109476, among which nine PCGs were associated with regulatory T cells: (1) chemokine (C-C motif) ligand 19 (CCL19), (2) CCL21, (3) C-C chemokine receptor type 7 (CCR7), (4) cluster of differentiation 70 (CD70), (5) ephrin B3 (EFNB3), (6) early growth response 3 (EGR3), (7) interleukin-7 receptor (IL7R), (8) galectin-1 (LGALS1), and (9) tumor necrosis factor (TNF) receptor superfamily member 13C (TNFRSF13C). The GEPIA database indicated that no significant differences existed between the expression of CCL19, CCL21, CD70, EFNB3, EGR3, IL7R, and TNFRSF13C in stomach adenocarcinoma (STAD) tissues and that in normal tissues (P > .05), while expressions of CCR7 and LGALS1 were significantly elevated in STAD tissues compared to the normal tissues (P < .05). The Kaplan-Meier Plotter database analysis, on the other hand, showed a significant relationship between all of the above-mentioned PCGs, except CCL19, and the prognosis of GC. Conclusions: CCL19, CCL21, CCR7, CD70, EFNB3, EGR3, IL7R, LGALS1, and TNFRSF13C are PCGs are differentially expressed in GC and closely associated with regulatory T cells. They may affect the occurrence and development of GC through a variety of pathways, including regulation of immune infiltration and inflammation, and are of great potential research value.


Subject(s)
Adenocarcinoma , Stomach Neoplasms , Humans , Prognosis , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Galectin 1 , Receptors, CCR7 , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology , Ephrin-B3
5.
Genes (Basel) ; 13(10)2022 10 13.
Article in English | MEDLINE | ID: mdl-36292733

ABSTRACT

Hu sheep, a famous breed in the Taihu Basin, has the advantages of non-seasonal estrus, multiple fetuses, coarse feeding tolerance, and suitability for house feeding. Runs of homozygosity (ROHs) were found to be an effective tool to detect the animal population structure and economic traits. The detection of ROHs is beneficial for reducing the incidence of inbreeding as well as identifying harmful variants in the genome. However, there is a lack of systemic reports on ruminants in previous studies of ROHs. Here, we sequenced 108 Hu sheep, detected ROHs in Hu sheep to calculate their inbreeding coefficient, and selected genes of Hu sheep breeds within the ROH islands which are relevant to agricultural economic characteristics. Then, we compared the characteristics of the occurrences of SNPs between Hu sheep and other sheep breeds, and also investigated the distribution of the frequencies of SNPs within specific gene regions of Hu sheep breeds to select their breed-specific genes. Furthermore, we performed a comparative genome and transcriptome analysis in human and sheep breeds to identify important reproduction-related genes. In this way, we found some significant SNPs, and mapped these with a set of interesting candidate genes which are related to the productive value of livestock (FGF9, BMPR1B, EFNB3, MICU2, GFRA3), healthy characteristics (LGSN, EPHA5, ALOX15B), and breed specificity (FGF9, SAP18, MICU2). These results in our study describe various production traits of Hu sheep from a genetic perspective, and provide insights into the genetic management and complementary understanding of Hu sheep.


Subject(s)
Ephrin-B3 , Inbreeding , Female , Humans , Sheep/genetics , Animals , Homozygote , Reproduction/genetics , Livestock
6.
Fundam Clin Pharmacol ; 36(2): 262-276, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34904278

ABSTRACT

Ephrin B/EphB signaling pathway is involved in the regulation of pain caused by spinal cord injury. However, the role of ephrin-B3/EphBs signaling in regulation of nociceptive information is poorly understood. In the present study, formalin-induced inflammatory pain, mechanical allodynia and thermal hyperalgesia, was measured using Efnb3 mutant mice (Efnb3-/- ) and wild-type (Efnb3+/+ ) mice. The spinal cord (L4-6) was selected for molecular and cellular identification by western blotting and immunofluorescence. Efnb3 mutant mice showed a significant increased the thermal and mechanical threshold, followed by aberrant thin myelin sheath. Furthermore, expression of proteolipid protein (PLP) was significantly lower in L4-6 spinal cord of Efnb3-/- mice. These morphological and behavioral abnormalities in mutant mice were rescued by conditional knock-in of wild-type ephrin-B3. Intrathecal administration of specific PLP siRNA significantly increased the thermal and mechanical threshold hyperalgesia in wild-type mice. However, overexpressing PLP protein by AAV9-PLP could decrease the sensitivity of mice to thermal and mechanical stimuli in Efnb3-/- mice, compared with scrabble Efnb3-/- mice. Further, Efnb3lacz mice, which have activities to initiate forward signaling, but transduce reverse signals by ephrin-B3, shows normal acute pain behavior, compared with wild type mice. These findings indicate that a key molecule Efnb3 act as a prominent contributor to hyperalgesia and essential roles of ephrin-B3/EphBs in nociception through a myelin-mediated mechanism.


Subject(s)
Ephrin-B3 , Hyperalgesia , Animals , Ephrin-B3/metabolism , Hyperalgesia/metabolism , Mice , Proteolipids/adverse effects , Proteolipids/metabolism , Receptors, Eph Family/metabolism , Signal Transduction/physiology , Spinal Cord/metabolism
7.
Cell Rep ; 36(9): 109628, 2021 08 31.
Article in English | MEDLINE | ID: mdl-34469726

ABSTRACT

Hendra virus and Nipah virus (NiV), members of the Henipavirus (HNV) genus, are zoonotic paramyxoviruses known to cause severe disease across six mammalian orders, including humans. We isolated a panel of human monoclonal antibodies (mAbs) from the B cells of an individual with prior exposure to equine Hendra virus (HeV) vaccine, targeting distinct antigenic sites. The most potent class of cross-reactive antibodies achieves neutralization by blocking viral attachment to the host cell receptors ephrin-B2 and ephrin-B3, with a second class being enhanced by receptor binding. mAbs from both classes display synergistic activity in vitro. In a stringent hamster model of NiV Bangladesh (NiVB) infection, antibodies from both classes reduce morbidity and mortality and achieve synergistic protection in combination. These candidate mAbs might be suitable for use in a cocktail therapeutic approach to achieve synergistic potency and reduce the risk of virus escape.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Antiviral Agents/pharmacology , Ephrin-B2/antagonists & inhibitors , Ephrin-B3/antagonists & inhibitors , Henipavirus Infections/prevention & control , Henipavirus/pathogenicity , Receptors, Virus/antagonists & inhibitors , Animals , Antibody Specificity , Chlorocebus aethiops , Cross Reactions , Disease Models, Animal , Drug Therapy, Combination , Ephrin-B2/immunology , Ephrin-B2/metabolism , Ephrin-B3/immunology , Ephrin-B3/metabolism , Female , Henipavirus Infections/immunology , Henipavirus Infections/metabolism , Henipavirus Infections/virology , Host-Pathogen Interactions , Humans , Mesocricetus , Receptors, Virus/immunology , Receptors, Virus/metabolism , Vero Cells
8.
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
9.
Science ; 372(6540)2021 04 23.
Article in English | MEDLINE | ID: mdl-33888612

ABSTRACT

Cell-cell interactions control the physiology and pathology of the central nervous system (CNS). To study astrocyte cell interactions in vivo, we developed rabies barcode interaction detection followed by sequencing (RABID-seq), which combines barcoded viral tracing and single-cell RNA sequencing (scRNA-seq). Using RABID-seq, we identified axon guidance molecules as candidate mediators of microglia-astrocyte interactions that promote CNS pathology in experimental autoimmune encephalomyelitis (EAE) and, potentially, multiple sclerosis (MS). In vivo cell-specific genetic perturbation EAE studies, in vitro systems, and the analysis of MS scRNA-seq datasets and CNS tissue established that Sema4D and Ephrin-B3 expressed in microglia control astrocyte responses via PlexinB2 and EphB3, respectively. Furthermore, a CNS-penetrant EphB3 inhibitor suppressed astrocyte and microglia proinflammatory responses and ameliorated EAE. In summary, RABID-seq identified microglia-astrocyte interactions and candidate therapeutic targets.


Subject(s)
Astrocytes/physiology , Cell Communication , Central Nervous System/pathology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Microglia/physiology , Multiple Sclerosis/physiopathology , Single-Cell Analysis , Animals , Antigens, CD/metabolism , Brain/pathology , Brain/physiopathology , Central Nervous System/physiopathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Ephrin-B3/metabolism , Herpesvirus 1, Suid/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Multiple Sclerosis/pathology , NF-kappa B/metabolism , Nerve Tissue Proteins/metabolism , RNA-Seq , Reactive Oxygen Species/metabolism , Receptor, EphB3/antagonists & inhibitors , Receptor, EphB3/metabolism , Receptors, Cell Surface/metabolism , Semaphorins/metabolism , Signal Transduction , T-Lymphocytes/physiology , TOR Serine-Threonine Kinases/metabolism
10.
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
11.
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
12.
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
13.
Acta Neuropathol ; 138(3): 457-476, 2019 09.
Article in English | MEDLINE | ID: mdl-31011859

ABSTRACT

Schwann cells (SC) enter the central nervous system (CNS) in pathophysiological conditions. However, how SC invade the CNS to remyelinate central axons remains undetermined. We studied SC migratory behavior ex vivo and in vivo after exogenous transplantation in the demyelinated spinal cord. The data highlight for the first time that SC migrate preferentially along blood vessels in perivascular extracellular matrix (ECM), avoiding CNS myelin. We demonstrate in vitro and in vivo that this migration route occurs by virtue of a dual mode of action of Eph/ephrin signaling. Indeed, EphrinB3, enriched in myelin, interacts with SC Eph receptors, to drive SC away from CNS myelin, and triggers their preferential adhesion to ECM components, such as fibronectin via integrinß1 interactions. This complex interplay enhances SC migration along the blood vessel network and together with lesion-induced vascular remodeling facilitates their timely invasion of the lesion site. These novel findings elucidate the mechanism by which SC invade and contribute to spinal cord repair.


Subject(s)
Blood Vessels , Cell Movement/physiology , Ephrin-B3/metabolism , Remyelination/physiology , Schwann Cells/physiology , Spinal Cord/metabolism , Animals , Demyelinating Diseases/pathology , Female , Fibronectins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction/physiology , Spinal Cord/pathology
14.
Elife ; 82019 02 21.
Article in English | MEDLINE | ID: mdl-30789343

ABSTRACT

Cortical networks are characterized by sparse connectivity, with synapses found at only a subset of axo-dendritic contacts. Yet within these networks, neurons can exhibit high connection probabilities, suggesting that cell-intrinsic factors, not proximity, determine connectivity. Here, we identify ephrin-B3 (eB3) as a factor that determines synapse density by mediating a cell-cell competition that requires ephrin-B-EphB signaling. In a microisland culture system designed to isolate cell-cell competition, we find that eB3 determines winning and losing neurons in a contest for synapses. In a Mosaic Analysis with Double Markers (MADM) genetic mouse model system in vivo the relative levels of eB3 control spine density in layer 5 and 6 neurons. MADM cortical neurons in vitro reveal that eB3 controls synapse density independently of action potential-driven activity. Our findings illustrate a new class of competitive mechanism mediated by trans-synaptic organizing proteins which control the number of synapses neurons receive relative to neighboring neurons.


Subject(s)
Cell Communication , Cerebral Cortex/cytology , Ephrin-B3/metabolism , Nerve Net/physiology , Neurons/metabolism , Animals , Mice
15.
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
16.
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
17.
Biochem Biophys Res Commun ; 503(4): 2212-2217, 2018 09 18.
Article in English | MEDLINE | ID: mdl-29953858

ABSTRACT

The ephrin family of membrane proteins binds Eph tyrosine kinase receptors. We have previously shown that ephrin-B3 also binds to heparan sulfate proteoglycans (HSPGs). We now show that ephrin-B3 can bind both secretory and cell associated PGs, such as agrin, collagen XVIII, Perlecan, and CD44, and indicate that such interaction with cell associated PGs involves a complex including 20 and 45 kDa proteins. Ephrin-B3 binding to HEK-293T cells is blocked by a secretory variant of CD44 (v3-v10), while over-expression of membrane associated CD44 increased ephrin-B3 binding. In addition, ephrin-B3 precipitated CD44 expressed by the oral squamous carcinoma cell line H376. Moreover, ephrin-B3 binding affinities to heparin and CD44 in solution was strong. In conclusion, we have identified secretory and cell associated PGs with high ability to bind ephrin-B3 and suggest that ephrin-B3 can bind to a protein complex organized by a membrane associated PG.


Subject(s)
Ephrin-B3/metabolism , Proteoglycans/metabolism , Cell Line, Tumor , HEK293 Cells , Heparin/metabolism , Humans , Hyaluronan Receptors/metabolism , Multiprotein Complexes/metabolism , Protein Binding
18.
Mol Med Rep ; 18(2): 2171-2181, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29901151

ABSTRACT

A growing body of evidence has demonstrated that Eph/ephrin signalling may serve a central role in intestinal diseases. However, whether erythropoietin­producing hepatocellular (Eph)/ephrin signalling is associated with the development of post­infectious irritable bowel syndrome (PI­IBS) is still unknown. In the present study, the role of Eph/Ephrin signalling in lipopolysaccharide (LPS)­induced intestinal injury was evaluated in vivo and in vitro. LPS treatment significantly increased the levels of proinflammatory mediators [monocyte chemoattractant protein­1, tumour necrosis factor α, interleukin (IL)­1ß, IL­6, intercellular adhesion molecule 1 and vascular cell adhesion molecule­1], activated the EphA2­Ephrin A1, protein kinase B (Akt)­nuclear factor (NF)­κB, Src­NF­κB and Wnt/ß­catenin signalling pathways, and inhibited EphB1­Ephrin B3 signalling in colon tissues, and primary cultured enteric neuronal and glial cells. Notably, EphA2 monoclonal antibody (mAb) treatment or Ephrin B3 overexpression could partially alleviate the LPS­induced upregulation of proinflammatory mediators, and Akt­NF­κB, Src­NF­κB and Wnt/ß­catenin signalling pathways. In addition, EphA2 mAb treatment could partially inhibit LPS­induced inactivation of EphB­Ephrin B3 signalling, while Ephrin B3 overexpression could abrogate LPS­induced activation of EphA2­Ephrin A1 signalling. EphB1/Ephrin B3 signalling may antagonise the EphA2/Ephrin A1­dependent pathway following LPS treatment. The results associated with the EphA2 signaling pathway, indicated that Eph/ephrin signalling may serve a bidirectional role in LPS­induced intestinal injury. Eph/ephrin signalling may be a novel therapeutic target for LPS­induced intestinal injury and potentially PI­IBS.


Subject(s)
Ephrin-A1/metabolism , Ephrin-B3/metabolism , Intestinal Mucosa/metabolism , Irritable Bowel Syndrome/metabolism , Lipopolysaccharides/toxicity , Receptor, EphA2/metabolism , Receptor, EphB1/metabolism , Signal Transduction/drug effects , Animals , Intestines/injuries , Intestines/pathology , Irritable Bowel Syndrome/chemically induced , Irritable Bowel Syndrome/pathology , Male , Mice , Mice, Inbred BALB C
19.
Int J Mol Med ; 41(6): 3457-3467, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29512697

ABSTRACT

Ephrin­B3 is important in the regulation of cell proliferation, differentiation and migration via cell­cell contact, and can activate the reelin pathway during brain development. However, the effect of ephrin­B3 on hippocampal neurogenesis and the reelin pathway in epilepsy remains to be fully elucidated. In the present study, the expression of ephrin­B3 in pilocarpine­induced status epilepticus (SE) rats was investigated. SYBR Green­based reverse transcription­quantitative polymerase chain reaction analysis, immunohistochemical labeling and western blot analysis were used to detect the gene and protein expression levels of ephrin­B3 and reelin pathway proteins. Immunofluorescence staining of doublecortin (DCX) was utilized to analyze hippocampal neurogenesis. The data revealed that the mRNA and protein expression levels of ephrin­B3 in the hippocampus decreased during the spontaneous seizure period. Of note, the expression of reelin and its downstream phosphorylation disabled 1 (p­Dab1) were also notably decreased during the spontaneous seizure period, which showed similar dynamic changes as in the expression of ephrin­B3. In addition, it was found that the number of DCX­labeled neuronal progenitor cells was increased in the hippocampus following pilocarpine­induced SE. To further clarify the role of ephrin­B3 in neurogenesis and the reelin pathway in epilepsy, an exogenous ephrin­B3 clustering stimulator, EphB3­Fc, was infused into the bilateral hippocampus of the rats post­SE. Following EphB3­Fc injection, it was found that the expression levels of reelin and p­Dab1 were significantly increased in the epileptic rats following EphB3­Fc injection. The number of DCX­labeled neuronal progenitor cells was reduced in the hippocampus of the epileptic rats. Furthermore, the intensity and frequency of spontaneous recurrent seizures and electroencephalographic seizures were attenuated in the epileptic rats post­injection. These results demonstrated the critical role of ephrin­B3 in regulation of the reelin pathway and hippocampal neurogenesis in epilepsy, providing experimental evidence that ephrin­B3 functions as a potential protective factor in epilepsy, at least in animals.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Ephrin-B3/metabolism , Epilepsy/chemically induced , Extracellular Matrix Proteins/metabolism , Hippocampus/metabolism , Nerve Tissue Proteins/metabolism , Pilocarpine/toxicity , Serine Endopeptidases/metabolism , Animals , Doublecortin Protein , Epilepsy/metabolism , Male , Neurogenesis/drug effects , Neurogenesis/physiology , Rats , Rats, Sprague-Dawley , Reelin Protein , Signal Transduction/physiology
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL