Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Sci Rep ; 11(1): 644, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33436772

ABSTRACT

Ephrin type-A receptor 10 (EPHA10) has been implicated as a potential target for breast and prostate cancer therapy. However, its involvement in oral squamous cell carcinoma (OSCC) remains unclear. We demonstrated that EPHA10 supports in vivo tumor growth and lymphatic metastasis of OSCC cells. OSCC cell migration, epithelial mesenchymal transition (EMT), and sphere formation were found to be regulated by EPHA10, and EPHA10 was found to drive expression of some EMT- and stemness-associated transcription factors. Among EPHA10 ligands, exogenous ephrin A4 (EFNA4) induced the most OSCC cell migration and sphere formation, as well as up-regulation of SNAIL, NANOG, and OCT4. These effects were abolished by extracellular signal-regulated kinase (ERK) inhibition and NANOG knockdown. Also, EPHA10 was required for EFNA4-induced cell migration, sphere formation, and expression of NANOG and OCT4 mRNA. Our microarray dataset revealed that EFNA4 mRNA expression was associated with expression of NANOG and OCT4 mRNA, and OSCC patients showing high co-expression of EFNA4 with NANOG or OCT4 mRNA demonstrated poor recurrence-free survival rates. Targeting forward signaling of the EFNA4-EPHA10 axis may be a promising therapeutic approach for oral malignancies, and the combination of EFNA4 mRNA and downstream gene expression may be a useful prognostic biomarker for OSCC.


Subject(s)
Cell Movement , Ephrin-A4/metabolism , Gene Expression Regulation, Neoplastic , Mouth Neoplasms/pathology , Nanog Homeobox Protein/metabolism , Receptors, Eph Family/metabolism , Spheroids, Cellular/pathology , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Proliferation , Ephrin-A4/genetics , Epithelial-Mesenchymal Transition , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Nanog Homeobox Protein/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Prognosis , Receptors, Eph Family/genetics , Spheroids, Cellular/metabolism , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
Cell Tissue Res ; 380(1): 155-172, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31820147

ABSTRACT

Interleukin (IL)-6 is a proinflammatory cytokine released in injured and contracting skeletal muscles. In this study, we examined cellular expression of proteins associated with cytoskeleton organization and cell migration, chosen on the basis of microRNA profiling, in rat primary skeletal muscle cells (RSkMC) treated with IL-6 (1 ng/ml) for 11 days. MiRNA microarray analysis and qRT-PCR revealed increased expression of miR-154-3p and miR-338-3p in muscle cells treated with IL-6. Pacsin3 was downregulated post-transcriptionally by IL-6, but not by IGF-I. Ephrin4A protein was increased both in IL-6- and IGF-I-treated myocytes. IL-6, but not IGF-I, stimulated migratory ability of RSkMC, examined in wound healing assay. Alpha-actinin protein was slightly augmented in RSKMC treated with IL-6, similarly to IGF-I. IL-6, but not IGF-I, upregulated desmin in differentiating RSkMC. IL-6 supplementation caused accumulation of alpha-actinin and desmin in near-nuclear area of muscle cells, which was manifested by increased ratio: mean near-nuclear fluorescence/mean peripheral cytoplasm fluorescence of these proteins. We concluded that IL-6, a known proinflammatory cytokine and a physical activity-associated myokine, acting during differentiation of primary skeletal muscle cells, alters expression of nonmuscle-specific miRNAs. This cytokine causes differential effects on pacsin-3 and ephrinA4, through post-transcriptional inhibition and stimulation, respectively. IL-6-exerted modifications of cytoskeletal proteins in muscle cells include both transcriptional (desmin and dynein heavy chain 5) and post-transcriptional activation (alpha-actinin). Moreover, IL-6 augments near-nuclear distribution of cytoskeletal proteins, alpha-actinin and desmin and promotes migration of myocytes. Such effects suggest that IL-6 plays a role during skeletal muscle regeneration, acting through mechanisms independent of regulation of myogenic program.


Subject(s)
Adaptor Proteins, Signal Transducing/biosynthesis , Ephrin-A4/biosynthesis , Interleukin-6/pharmacology , Myoblasts, Skeletal/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Differentiation/drug effects , Cytoskeletal Proteins/biosynthesis , Cytoskeletal Proteins/genetics , Disease Models, Animal , Ephrin-A4/genetics , Insulin-Like Growth Factor I/pharmacology , MicroRNAs/biosynthesis , MicroRNAs/genetics , MicroRNAs/metabolism , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/drug effects , RNA Processing, Post-Transcriptional , Rats , Recombinant Proteins/pharmacology , Transcription, Genetic
3.
Cleft Palate Craniofac J ; 55(7): 1020-1025, 2018 08.
Article in English | MEDLINE | ID: mdl-28135115

ABSTRACT

Craniosynostosis (CS) has a prevalence of approximately 1 in every 2000 live births and is characterized by the premature fusion of one or more cranial sutures. Failure to maintain the cell lineage boundary at the coronal suture is thought to be involved in the pathology of some forms of CS. The Ephrin family of receptor tyrosine kinases consists of membrane-bound receptors and ligands that control cell patterning and the formation of developmental boundaries. Mutations in the ephrin A4 (EFNA4) and ephrin B1 (EFNB1) ligands have been linked to nonsyndromic CS and craniofrontonasal syndrome, respectively, in patient samples. We have previously described a colony of rabbits with a heritable pattern of coronal suture synostosis, although the genetic basis for synostosis within this model remains unknown. The present study was performed to determine if EFNA4 or EFNB1 could be the loci of the causal mutation in this unique animal model. Sequencing of EFNA4 and EFNB1 was performed using templates obtained from wild-type (n = 4) and craniosynostotic (n = 4) rabbits. No structural coding errors were identified in either gene. A single-nucleotide transversion was identified in one wild-type rabbit within the third intron of EFNA4. These data indicate that the causal locus for heritable CS in this rabbit model is not located within the structural coding regions of either EFNA4 or EFNB1.


Subject(s)
Craniosynostoses/genetics , Ephrin-A4/genetics , Ephrin-B1/genetics , Animals , Disease Models, Animal , Introns , Mutation , Phenotype , Polymerase Chain Reaction , Rabbits
4.
Oncotarget ; 7(30): 48481-48500, 2016 Jul 26.
Article in English | MEDLINE | ID: mdl-27374180

ABSTRACT

A role of endothelial cells in the survival of CLL cells during extravasation is presently unknown. Herein we show that CLL cells but not normal B cells can receive apoptotic signals through physical contact with TNF-α activated endothelium impairing survival in transendothelial migration (TEM) assays. In addition, the CLL cells of patients having lymphadenopathy (LApos) show a survival advantage during TEM that can be linked to increased expression of α4 and αL integrin chains. Within this context, ephrinA4 expressed on the surface of CLL cells sequestrates integrins and inactivates them resulting in reduced adhesion and inhibition of apoptotic/survival signals through them. In agreement, ephrinA4 silencing resulted in increased survival of CLL cells of LApos patients but not LA neg patients. Similarly was observed when a soluble ephrinA4 isoform was added to TEM assays strongly suggesting that accumulation of this isoform in the serum of LApos patients could contribute to CLL cells dissemination and survival in vivo. In supporting, CLL lymphadenopathies showed a preferential accumulation of apoptotic CLL cells around high endothelial venules lacking ephrinA4. Moreover, soluble ephrinA4 isolated from sera of patients increased the number and viability of CLL cells recovered from the lymph nodes of adoptively transferred mice. Finally, we present evidence suggesting that soluble ephrinA4 mediated survival during TEM could enhance a transcellular TEM route of the CLL cells. Together these findings point to an important role of ephrinA4 in the nodal dissemination of CLL cells governing extravasation and survival.


Subject(s)
Apoptosis , CD11a Antigen/metabolism , Cell Survival , Ephrin-A4/metabolism , Integrin alpha4/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Aged , Aged, 80 and over , Animals , B-Lymphocytes/physiology , Cells, Cultured , Coculture Techniques , Endothelium/metabolism , Ephrin-A4/blood , Ephrin-A4/genetics , Female , Human Umbilical Vein Endothelial Cells , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/blood , Lymph Nodes/pathology , Lymphatic Metastasis , Male , Mice , Mice, Inbred BALB C , Middle Aged , Protein Isoforms/blood , Protein Isoforms/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Specific Pathogen-Free Organisms , Transendothelial and Transepithelial Migration , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/metabolism , Xenograft Model Antitumor Assays
5.
Transl Psychiatry ; 4: e450, 2014 Sep 30.
Article in English | MEDLINE | ID: mdl-25268254

ABSTRACT

Fear conditioning leads to long-term fear memory formation and is a model for studying fear-related psychopathologies conditions such as phobias and posttraumatic stress disorder. Long-term fear memory formation is believed to involve alterations of synaptic efficacy mediated by changes in synaptic transmission and morphology in lateral amygdala (LA). EphrinA4 and its cognate Eph receptors are intimately involved in regulating neuronal morphogenesis, synaptic transmission and plasticity. To assess possible roles of ephrinA4 in fear memory formation we designed and used a specific inhibitory ephrinA4 mimetic peptide (pep-ephrinA4) targeted to EphA binding site. We show that this peptide, composed of the ephrinA4 binding domain, interacts with EphA4 and inhibits ephrinA4-induced phosphorylation of EphA4. Microinjection of the pep-ephrinA4 into rat LA 30 min before training impaired long- but not short-term fear conditioning memory. Microinjection of a control peptide derived from a nonbinding E helix site of ephrinA4, that does not interact with EphA, had no effect on fear memory formation. Microinjection of pep-ephrinA4 into areas adjacent to the amygdala had no effect on fear memory. Acute systemic administration of pep-ephrinA4 1 h after training also impaired long-term fear conditioning memory formation. These results demonstrate that ephrinA4 binding sites in LA are essential for long-term fear memory formation. Moreover, our research shows that ephrinA4 binding sites may serve as a target for pharmacological treatment of fear and anxiety disorders.


Subject(s)
Amygdala/physiology , Ephrin-A4/administration & dosage , Fear/physiology , Memory, Long-Term/physiology , Animals , Binding Sites/genetics , Conditioning, Classical/physiology , Ephrin-A4/genetics , Neuronal Plasticity/genetics , Neuronal Plasticity/physiology , Rats , Rats, Sprague-Dawley , Synaptic Transmission/genetics , Synaptic Transmission/physiology
6.
Eur J Neurosci ; 38(11): 3567-79, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24103058

ABSTRACT

Adult central nervous system axons show restricted growth and regeneration properties after injury. One of the underlying mechanisms is the activation of the Nogo-A/Nogo receptor (NgR1) signaling pathway. Nogo-A knockout (KO) mice show enhanced regenerative growth in vivo, even though it is less pronounced than after acute antibody-mediated neutralization of Nogo-A. Residual inhibition may involve a compensatory component. By mRNA expression profiling and immunoblots we show increased expression of several members of the Ephrin/Eph and Semaphorin/Plexin families of axon guidance molecules, e.g. EphrinA3 and EphA4, in the intact spinal cord of adult Nogo-A KO vs. wild-type (WT) mice. EphrinA3 inhibits neurite outgrowth of EphA4-positive neurons in vitro. In addition, EphrinA3 KO myelin extracts are less growth-inhibitory than WT but more than Nogo-A KO myelin extracts. EphA4 KO cortical neurons show decreased growth inhibition on Nogo-A KO myelin as compared with WT neurons, supporting increased EphA4-mediated growth inhibition in Nogo-A KO mice. Consistently, in vivo, Nogo-A/EphA4 double KO mice show increased axonal sprouting and regeneration after spinal cord injury as compared with EphA4 KO mice. Our results reveal the upregulation of developmental axon guidance cues following constitutive Nogo-A deletion, e.g. the EphrinA3/EphA4 ligand/receptor pair, and support their role in restricting neurite outgrowth in the absence of Nogo-A.


Subject(s)
Axons/physiology , Cerebral Cortex/metabolism , Ganglia, Spinal/metabolism , Myelin Proteins/metabolism , Spinal Cord Regeneration , Up-Regulation , Animals , Axons/metabolism , Cells, Cultured , Cerebral Cortex/pathology , Cerebral Cortex/physiology , Ephrin-A3/genetics , Ephrin-A3/metabolism , Ephrin-A4/genetics , Ephrin-A4/metabolism , Ganglia, Spinal/pathology , Ganglia, Spinal/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin Proteins/genetics , Myelin Sheath/genetics , Myelin Sheath/metabolism , Nogo Proteins , Pyramidal Tracts/metabolism , Pyramidal Tracts/pathology , Pyramidal Tracts/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spinal Cord Injuries/metabolism
7.
Biochim Biophys Acta ; 1833(10): 2201-11, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23707953

ABSTRACT

The Eph receptors represent the largest family of receptor tyrosine kinases. Both Eph receptors and their ephrin ligands are cell-surface proteins, and they typically mediate cell-to-cell communication by interacting at sites of intercellular contact. The major aim of the present study was to investigate the involvement of EphA4-ephrin-A1 interaction in monocyte adhesion to endothelial cells, as this process is a crucial step during the initiation and progression of the atherosclerotic plaque. Immunohistochemical analysis of human atherosclerotic plaques revealed expression of EphA4 receptor and ephrin-A1 ligand in major cell types within the plaque. Short-time stimulation of endothelial cells with the soluble ligand ephrin-A1 leads to a fourfold increase in adhesion of human monocytes to endothelial cells. In addition, ephrin-A1 further increases monocyte adhesion to already inflamed endothelial cells. EphrinA1 mediates its effect on monocyte adhesion via the activated receptor EphA4. This ephrinA1/EphA4 induced process involves the activation of the Rho signaling pathway and does not require active transcription. Rho activation downstream of EphA4 leads to increased polymerization of actin filaments in endothelial cells. This process was shown to be crucial for the proadhesive effect of ephrin-A1. The results of the present study show that ephrin-A1-induced EphA4 forward signaling promotes monocyte adhesion to endothelial cells via activation of RhoA and subsequent stress-fiber formation by a non-transcriptional mechanism.


Subject(s)
Atherosclerosis/metabolism , Cell Adhesion , Endothelium, Vascular/metabolism , Ephrin-A1/metabolism , Ephrin-A4/metabolism , Monocytes/metabolism , Atherosclerosis/genetics , Atherosclerosis/pathology , Blotting, Western , Cell Proliferation , Cells, Cultured , Endothelium, Vascular/cytology , Ephrin-A1/antagonists & inhibitors , Ephrin-A1/genetics , Ephrin-A4/antagonists & inhibitors , Ephrin-A4/genetics , Flow Cytometry , Humans , Immunoenzyme Techniques , Immunoprecipitation , Lipoproteins, LDL/genetics , Lipoproteins, LDL/metabolism , Macrophages/cytology , Macrophages/metabolism , Monocytes/cytology , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
8.
Invest Ophthalmol Vis Sci ; 53(4): 1990-8, 2012 Apr 18.
Article in English | MEDLINE | ID: mdl-22408005

ABSTRACT

PURPOSE: Retinal neovascularization (NV) is a major cause of blindness. Recent research suggests that factors other than VEGF participate in this process. This study aimed to determine the role of ephrin-A4 in retinal NV. METHODS: The expression and effect of ephrin-A4 was investigated in a mouse model of oxygen-induced retinopathy (OIR) and the RF/6A retina endothelial cell line. Ephrin-A4 expression and VEGF signaling pathway phosphorylation were determined by PCR, immunohistochemistry, and western blot analyses. ShRNA was used to silence ephrin-A4 in vitro and in vivo. Retinal flat mounts and tube formation assays were performed to evaluate ephrin-A4 function in the NV process in vivo and in vitro. RESULTS: Ephrin-A4 was overexpressed in the retina of OIR mice and in RF/6A and RPE cells after CoCl2 stimulation. In vitro, Ephrin-A4/Fc treatment significantly increased the tube number of RF/6A cells on a membrane preparation and the phosphorylation levels of VEGR2, Akt1, and ERK1/2 in RF/6A cells. Moreover, ephrin-A4 knockout markedly suppressed pathologic neovascularization in vivo and inhibited the proliferation and tube formation capacity of RF/6A cells in vitro. Furthermore, in the absence of ephrin-A4, the phosphorylation of VEGFR2, Akt1, and ERK1/2 was defective under VEGF165 stimulation, and the proangiogenic function of VEGF165 was also compromised. CONCLUSIONS: This study suggests that ephrin-A4 plays an important role in retinal NV and is a potential target against retinal NV. The proangiogenic function of ephrin-A4 may be linked to its crucial role in the VEGF signaling pathway.


Subject(s)
Ephrin-A4/genetics , Gene Expression Regulation, Developmental , RNA/genetics , Retinal Neovascularization/metabolism , Retinal Pigment Epithelium/pathology , Vascular Endothelial Growth Factor A/genetics , Animals , Animals, Newborn , Blotting, Western , Cells, Cultured , Disease Models, Animal , Ephrin-A4/biosynthesis , Follow-Up Studies , Immunohistochemistry , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction , Retinal Neovascularization/pathology , Retinal Pigment Epithelium/metabolism , Signal Transduction/genetics , Vascular Endothelial Growth Factor A/biosynthesis
9.
PLoS Pathog ; 7(10): e1002309, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22028653

ABSTRACT

The IAPE (Intracisternal A-type Particles elements with an Envelope) family of murine endogenous retroelements is present at more than 200 copies in the mouse genome. We had previously identified a single copy that proved to be fully functional, i.e. which can generate viral particles budding out of the cell and infectious on a series of cells, including human cells. We also showed that IAPE are the progenitors of the highly reiterated IAP elements. The latter are now strictly intracellular retrotransposons, due to the loss of the envelope gene and re-localisation of the associated particles in the course of evolution. In the present study we searched for the cellular receptor of the IAPE elements, by using a lentiviral human cDNA library and a pseudotype assay on transduced cells. We identified Ephrin A4, a GPI-anchored molecule involved in several developmental processes, as a receptor for the IAPE pseudotypes. We also found that the other 4 members of the Ephrin A family -but not those of the closely related Ephrin B family- were also able to mediate IAPE cell entry, thus significantly increasing the amount of possible cell types susceptible to IAPE infection. We show that these include mouse germline cells, as illustrated by immunohistochemistry experiments, consistent with IAPE genomic amplification by successive re-infection. We propose that the uncovered properties of the identified receptors played a role in the accumulation of IAPE elements in the mouse genome, and in the survival of a functional copy.


Subject(s)
Endogenous Retroviruses/pathogenicity , Ephrins/metabolism , Host-Pathogen Interactions/physiology , Retroviridae Infections/virology , Animals , Chlorocebus aethiops , Endogenous Retroviruses/genetics , Ephrin-A4/genetics , Ephrin-A4/metabolism , Ephrins/genetics , Female , Gene Expression Regulation, Viral , Gene Library , Genes, Intracisternal A-Particle/genetics , Genes, Viral , HEK293 Cells , Humans , Mice , Ovary/metabolism , Retroviridae Infections/metabolism , Vero Cells , Virus Replication
10.
Leuk Res ; 33(3): 395-406, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18819711

ABSTRACT

Increasing information relates some Eph receptors and their ligands, ephrins (EFN), with the immune system. Herein, we found that normal B-cells from peripheral blood (PB) and lymph nodes (LN) showed a differential expression of certain Eph/EFN members, some of them being modulated upon in vitro stimulation including EFNA1, EFNA4, EphB6 and EphA10. In contrast, PB CLL B-cells showed a more heterogeneous Eph/EFN profile than their normal PB B-cell counterparts, expressing Eph/EFN members frequently found within the LN and activated B-cells, specially EFNA4, EphB6 and EphA10. Two of them, EphB6 and EFNA4 were further related with the clinical course of CLL patients. EphB6 expression correlated with a high content of ZAP-70 mRNA and a poor prognosis. High serum levels of a soluble EFNA4 isoform positively correlated with increasing peripheral blood lymphocyte counts and lymphadenopathy. These findings suggest that Eph/EFN might be relevant in normal B-cell biology and could represent new potential prognostic markers and therapeutic targets for CLL.


Subject(s)
B-Lymphocytes/pathology , Ephrins/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Receptors, Eph Family/genetics , Biomarkers, Tumor/analysis , Case-Control Studies , Ephrin-A4/genetics , Gene Expression Profiling , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Prognosis , Receptor Protein-Tyrosine Kinases/genetics , ZAP-70 Protein-Tyrosine Kinase/genetics
11.
Hear Res ; 235(1-2): 39-46, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17967521

ABSTRACT

The Eph receptor tyrosine kinases and their membrane-anchored ligands, ephrins, are signaling proteins that act as axon guidance molecules during chick auditory brainstem development. We recently showed that Eph proteins also affect patterns of neural activation in the mammalian brainstem. However, functional deficits in the brainstems of mutant mice have not been assessed physiologically. The present study characterizes neural activation in Eph protein deficient mice in the auditory brainstem response (ABR). We recorded the ABR of EphA4 and ephrin-B2 mutant mice, aged postnatal day 18-20, and compared them to wild type controls. The peripheral hearing threshold of EphA4(-/-) mice was 75% higher than that of controls. Waveform amplitudes of peak 1 (P1) were 54% lower in EphA4(-/-) mice than in controls. The peripheral hearing thresholds in ephrin-B2(lacZ/)(+) mice were also elevated, with a mean value 20% higher than that of controls. These ephrin-B2(lacZ/)(+) mice showed a 38% smaller P1 amplitude. Significant differences in latency to waveform peaks were also observed. These elevated thresholds and reduced peak amplitudes provide evidence for hearing deficits in both of these mutant mouse lines, and further emphasize an important role for Eph family proteins in the formation of functional auditory circuitry.


Subject(s)
Auditory Pathways/metabolism , Auditory Threshold , Ephrin-A4/metabolism , Ephrin-B2/metabolism , Evoked Potentials, Auditory, Brain Stem , Hearing Disorders/metabolism , Acoustic Stimulation , Animals , Auditory Pathways/physiopathology , Ephrin-A4/deficiency , Ephrin-A4/genetics , Ephrin-B2/deficiency , Ephrin-B2/genetics , Genotype , Hearing Disorders/genetics , Hearing Disorders/physiopathology , Mice , Mice, Knockout , Phenotype , Reaction Time , Time Factors
12.
BMC Mol Biol ; 8: 90, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17937806

ABSTRACT

BACKGROUND: ADAM15 is a metalloprotease-disintegrin implicated in ectodomain shedding and cell adhesion. Aberrant ADAM15 expression has been associated with human cancer and other disorders. We have previously shown that the alternative splicing of ADAM15 transcripts is mis-regulated in cancer cells. To gain a better understanding of ADAM15 regulation, its genomic organization and regulatory elements as well as the alternative exon use in human tissues were characterized. RESULTS: Human ADAM15, flanked by the FLJ32785/DCST1 and ephrin-A4 genes, spans 11.4 kb from the translation initiation codon to the polyadenylation signal, being the shortest multiple-exon ADAM gene. The gene contains 23 exons varying from 63 to 316 bp and 22 introns from 79 to 1283 bp. The gene appeared to have several transcription start sites and their location suggested the promoter location within a CpG island proximal to the translation start. Reporter expression experiments confirmed the location of functional GC-rich, TATAless and CAATless promoter, with the most critical transcription-supporting elements located -266 to -23 bp relative to the translation start. Normal human tissues showed different complex patterns of at least 13 different ADAM15 splice variants arising from the alternative use of the cytosolic-encoding exons 19, 20a/b, and 21a/b. The deduced ADAM15 protein isoforms have different combinations of cytosolic regulatory protein interaction motifs. CONCLUSION: Characterization of human ADAM15 gene and identification of elements involved in the regulation of transcription and alternative splicing provide important clues for elucidation of physiological and pathological roles of ADAM15. The present results also show that the alternative exon use is a physiological post-transcriptional mechanism regulating ADAM15 expression in human tissues.


Subject(s)
ADAM Proteins/genetics , Alternative Splicing/physiology , Exons/physiology , Gene Expression Regulation, Enzymologic/physiology , Membrane Proteins/genetics , ADAM Proteins/biosynthesis , Amino Acid Motifs/genetics , Cell Adhesion/physiology , Cell Line , Codon, Initiator/physiology , CpG Islands/physiology , Ephrin-A4/genetics , Ephrin-A4/metabolism , Humans , Membrane Proteins/biosynthesis , Neoplasms/enzymology , Neoplasms/genetics , Organ Specificity/physiology , Promoter Regions, Genetic/physiology , Protein Isoforms/biosynthesis , Protein Isoforms/genetics
13.
Hum Mol Genet ; 15(8): 1319-28, 2006 Apr 15.
Article in English | MEDLINE | ID: mdl-16540516

ABSTRACT

Boundaries between cellular compartments often serve as signaling interfaces during embryogenesis. The coronal suture is a major growth center of the skull vault and develops at a boundary between cells derived from neural crest and mesodermal origin, forming the frontal and parietal bones, respectively. Premature fusion of these bones, termed coronal synostosis, is a common human developmental anomaly. Known causes of coronal synostosis include haploinsufficiency of TWIST1 and a gain of function mutation in MSX2. In Twist1(+/-) mice with coronal synostosis, we found that the frontal-parietal boundary is defective. Specifically, neural crest cells invade the undifferentiated mesoderm of the Twist1(+/-) mutant coronal suture. This boundary defect is accompanied by an expansion in Msx2 expression and reduction in ephrin-A4 distribution. Reduced dosage of Msx2 in the Twist1 mutant background restores the expression of ephrin-A4, rescues the suture boundary and inhibits craniosynostosis. Underlining the importance of ephrin-A4, we identified heterozygous mutations in the human orthologue, EFNA4, in three of 81 patients with non-syndromic coronal synostosis. This provides genetic evidence that Twist1, Msx2 and Efna4 function together in boundary formation and the pathogenesis of coronal synostosis.


Subject(s)
Cranial Sutures/metabolism , Craniosynostoses/metabolism , Craniosynostoses/pathology , Ephrins/metabolism , Mesoderm/metabolism , Neural Crest/metabolism , Receptors, Eph Family/metabolism , Animals , Base Sequence , COS Cells , Cells, Cultured , Chlorocebus aethiops , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Embryo, Mammalian , Ephrin-A2/genetics , Ephrin-A2/metabolism , Ephrin-A4/genetics , Ephrin-A4/metabolism , Gene Expression Regulation, Developmental , Heterozygote , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Immunohistochemistry , Mesoderm/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Mutation , Neural Crest/cytology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phenotype , Signal Transduction , Tumor Cells, Cultured , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism
14.
Dev Cell ; 6(3): 383-95, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15030761

ABSTRACT

Semaphorins and ephrins are axon guidance cues. In C. elegans, semaphorin-2a/mab-20 and ephrin-4/efn-4/mab-26 also regulate cell sorting to form distinct rays in the male tail. Several erf (enhancer of ray fusion) mutations were identified in a mab-20 enhancer screen. Mutants of plexin-2 (plx-2) and unc-129, which encodes an axon guiding TGF-beta, were also found to be erfs. Genetic analyses show that plx-2 and mab-20 function in the same pathway, as expected if PLX-2 is a receptor for MAB-20. Surprisingly, MAB-20 also signals in a parallel pathway that requires efn-4. This signal utilizes a non-plexin receptor. The expression of plx-2, efn-4, and unc-129 in subsets of 3-cell sensory ray clusters likely mediates the ray-specific cell sorting functions of the ubiquitously expressed mab-20. We present a model for the integrated control of TGF-beta, semaphorin, and ephrin signaling in the sorting of cell clusters into distinct rays in the developing male tail.


Subject(s)
Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/metabolism , Ephrin-A4/physiology , Membrane Proteins/physiology , Nerve Tissue Proteins/physiology , Sense Organs/physiology , Signal Transduction/physiology , Transforming Growth Factor beta/physiology , Animals , Animals, Genetically Modified , Caenorhabditis elegans/embryology , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chi-Square Distribution , Chromosome Mapping/methods , Cloning, Molecular , DNA Mutational Analysis , Enhancer Elements, Genetic , Ephrin-A4/genetics , Ephrins/physiology , Gene Expression Regulation, Developmental , Green Fluorescent Proteins , Immunohistochemistry/methods , Luminescent Proteins/metabolism , Male , Membrane Proteins/genetics , Models, Molecular , Mutagenesis, Insertional/methods , Mutation , Nerve Tissue Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Sequence Analysis, DNA , Transforming Growth Factor beta/genetics , Xenopus Proteins/genetics , Xenopus Proteins/metabolism
15.
J Neurosci ; 24(5): 1070-8, 2004 Feb 04.
Article in English | MEDLINE | ID: mdl-14762125

ABSTRACT

Eph receptor tyrosine kinases and ephrins are required for axon patterning and plasticity in the developing nervous system. Typically, Eph-ephrin interactions promote inhibitory events; for example, prohibiting the entry of neural cells into certain embryonic territories. Here, we show that distinct subsets of motor neurons that express EphA4 respond differently to ephrin-A5. EphA4-positive LMC(l) axons avoid entering ephrin-A5-positive hindlimb mesoderm. In contrast, EphA4-positive MMC(m) axons extend through ephrin-A5-positive rostral half-sclerotome. Blocking EphA4 activation in MMC(m) neurons or expanding the domain of ephrin-A5 expression in the somite results in the aberrant growth of MMC(m) axons into the caudal half-sclerotome. Moreover, premature expression of EphA4 in MMC(m) neurons leads to a portion of their axons growing into novel ephrin-A5-positive territories. Together, these results indicate that EphA4-ephrin-A5 signaling acts in a positive manner to constrain MMC(m) axons to the rostral half-sclerotome. Furthermore, we show that Eph activation localizes to distinct subcellular compartments of LMC(l) and MMC(m) neurons, consistent with distinct EphA4 signaling cascades in these neuronal subpopulations.


Subject(s)
Ephrin-A4/biosynthesis , Ephrin-A5/physiology , Motor Neurons/metabolism , Neural Inhibition/physiology , Animals , Axons/drug effects , Axons/physiology , Cell Differentiation/physiology , Cells, Cultured , Chick Embryo , Electroporation , Ephrin-A4/genetics , Ephrin-A5/genetics , Ephrin-A5/pharmacology , Hindlimb/embryology , Hindlimb/innervation , Ligands , Mesoderm/metabolism , Motor Neurons/drug effects , Neural Inhibition/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology
16.
Blood ; 103(4): 1348-55, 2004 Feb 15.
Article in English | MEDLINE | ID: mdl-14576067

ABSTRACT

We have previously shown that platelets express 2 receptor tyrosine kinases, EphA4 and EphB1, and the Eph kinase ligand, ephrinB1, and proposed that transcellular Eph/ephrin interactions made possible by the onset of platelet aggregation promote the further growth and stability of the hemostatic plug. The present study examines how this might occur. The results show that clustering of either ephrinB1 or EphA4 causes platelets to adhere to immobilized fibrinogen via alpha(IIb)beta(3). Adhesion occurs more slowly than with adenosine diphosphate (ADP) and requires phosphatidylinositol 3 (PI3)-kinase and protein kinase C activity but not ephrinB1 phosphorylation. By itself, Eph and ephrin signaling is insufficient to cause aggregation or the binding of soluble fibrinogen, but it can potentiate aggregation initiated by a Ca(++) ionophore or by agonists for thrombin and thromboxane receptors. It also enhances Rap1 activation without requiring ADP secretion, ephrinB1 phosphorylation, or the activation of PI3-kinase and Src. From this we conclude that (1) Eph/ephrin signaling enhances the ability of platelet agonists to cause aggregation provided that those agonists can increase cytosolic Ca(++); (2) this is accomplished in part by activating Rap1; and (3) these effects require oligomerization of ephrinB1 but not phosphotyrosine-based interactions with the ephrinB1 cytoplasmic domain.


Subject(s)
Blood Platelets/metabolism , Ephrin-B1/metabolism , Platelet Aggregation/physiology , Signal Transduction/physiology , rap1 GTP-Binding Proteins/metabolism , Ephrin-A4/genetics , Ephrin-A4/metabolism , Humans , Phosphorylation , Platelet Adhesiveness/physiology , Protein Kinases/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
17.
Blood ; 102(13): 4431-40, 2003 Dec 15.
Article in English | MEDLINE | ID: mdl-12907451

ABSTRACT

Eph receptor tyrosine kinases and their ligands, the ephrins, have been primarily described in the nervous system for their roles in axon guidance, development, and cell intermingling. Here we address whether Eph receptors may also regulate dendritic cell (DC) trafficking. Reverse transcription-polymerase chain reaction (RT-PCR) analysis showed that DCs derived from CD34+ progenitors, but not from monocytes, expressed several receptors, in particular EphA2, EphA4, EphA7, EphB1, and EphB3 mRNA. EphB3 was specifically expressed by Langerhans cells, and EphA2 and EphA7 were expressed by both Langerhans- and interstitial-type DCs. EphA and EphB protein expression on DCs generated in vitro was confirmed by staining with ephrin-A3-Fc and ephrin-B3-Fc fusion proteins that bind to different Eph members, in particular EphA2 and EphB3. Immunostaining with anti-EphA2 antibodies demonstrated the expression of EphA2 by immature DCs and by skin Langerhans cells isolated ex vivo. Interestingly, ephrin expression was detected in epidermal keratinocytes and also in DCs. Adhesion of CD34+-derived DCs to fibronectin, but not to poly-l-lysine, was increased in the presence of ephrin-A3-Fc, a ligand of EphA2, through a beta1 integrin activation pathway. As such, EphA2/ephrin-A3 interactions may play a role in the localization and network of Langerhans cells in the epithelium and in the regulation of their trafficking.


Subject(s)
Dendritic Cells/enzymology , Ephrin-A2/physiology , Fibronectins/chemistry , Receptors, Eph Family/physiology , Antigens, CD34/analysis , Cell Adhesion/physiology , Cell Differentiation/drug effects , Cell Lineage , Cell Movement , Dendritic Cells/cytology , Ephrin-A2/biosynthesis , Ephrin-A2/genetics , Ephrin-A4/biosynthesis , Ephrin-A4/genetics , Ephrin-B1/biosynthesis , Ephrin-B1/genetics , Ephrin-B3/biosynthesis , Ephrin-B3/genetics , Epidermal Cells , Epidermis/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Humans , Integrin beta1/physiology , Keratinocytes/enzymology , Langerhans Cells/cytology , Langerhans Cells/enzymology , Polylysine/chemistry , Recombinant Fusion Proteins/immunology , Tumor Necrosis Factor-alpha/pharmacology
18.
Neuron ; 39(3): 453-65, 2003 Jul 31.
Article in English | MEDLINE | ID: mdl-12895420

ABSTRACT

The mechanisms generating precise connections between specific thalamic nuclei and cortical areas remain poorly understood. Using axon tracing analysis of ephrin/Eph mutant mice, we provide in vivo evidence that Eph receptors in the thalamus and ephrins in the cortex control intra-areal topographic mapping of thalamocortical (TC) axons. In addition, we show that the same ephrin/Eph genes unexpectedly control the inter-areal specificity of TC projections through the early topographic sorting of TC axons in an intermediate target, the ventral telencephalon. Our results constitute the first identification of guidance cues involved in inter-areal specificity of TC projections and demonstrate that the same set of mapping labels is used differentially for the generation of topographic specificity of TC projections between and within individual cortical areas.


Subject(s)
Cerebral Cortex/metabolism , Ephrin-A4/genetics , Ephrin-A5/genetics , Receptor, EphA4/genetics , Receptor, EphA5/genetics , Thalamus/metabolism , Animals , Brain Mapping/methods , Cerebral Cortex/embryology , Cerebral Cortex/enzymology , Ephrin-A4/biosynthesis , Ephrin-A4/physiology , Ephrin-A5/biosynthesis , Ephrin-A5/physiology , Female , Ligands , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Pathways/embryology , Neural Pathways/enzymology , Neural Pathways/metabolism , Neural Pathways/physiology , Receptor, EphA4/biosynthesis , Receptor, EphA4/physiology , Receptor, EphA5/biosynthesis , Receptor, EphA5/physiology , Thalamus/embryology , Thalamus/enzymology
19.
Neuron ; 38(4): 581-96, 2003 May 22.
Article in English | MEDLINE | ID: mdl-12765610

ABSTRACT

The formation of topographic neural maps relies on the coordinate assignment of neuronal cell body position and axonal trajectory. The projection of motor neurons of the lateral motor column (LMC) along the dorsoventral axis of the limb mesenchyme constitutes a simple topographic map that is organized in a binary manner. We show that LIM homeodomain proteins establish motor neuron topography by coordinating the mediolateral settling position of motor neurons within the LMC with the dorsoventral selection of axon pathways in the limb. These topographic projections are established, in part, through LIM homeodomain protein control of EphA receptors and ephrin-A ligands in motor neurons and limb mesenchymal cells.


Subject(s)
Ephrin-A4/metabolism , Homeodomain Proteins/metabolism , Motor Neurons/physiology , Nerve Tissue Proteins , Receptors, Eph Family/metabolism , Spinal Cord/physiology , Animals , Axons/physiology , Chick Embryo , Chimera , Ephrin-A4/genetics , Extremities/embryology , Extremities/innervation , Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/genetics , LIM-Homeodomain Proteins , Mesoderm/cytology , Mice , Motor Cortex/cytology , Motor Cortex/embryology , Receptors, Eph Family/genetics , Spinal Cord/cytology , Spinal Cord/embryology , Transcription Factors
SELECTION OF CITATIONS
SEARCH DETAIL