Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Eur Rev Med Pharmacol Sci ; 24(12): 6735-6743, 2020 06.
Article in English | MEDLINE | ID: mdl-32633364

ABSTRACT

OBJECTIVE: The purpose of this study was to investigate the expression level of EphA3 in nasopharyngeal carcinoma (NPC) and its effect on the proliferative capacity of NPC. Meanwhile, the underlying mechanism by which EphA3 prompts NPC malignant progression was further explored. PATIENTS AND METHODS: In this study, the expression of EphA3 in 42 pairs of tumor tissue specimens and paracancerous ones collected from NPC patients was detected by quantitative Real Time-Polymerase Chain Reaction (qRT-PCR), and the interplay between EphA3 expression and clinical indicators, as well as prognosis of NPC patients was analyzed. Meanwhile, qRT-PCR was also applied to further verify EphA expression in NPC cell lines. In addition, EphA knockdown model was constructed in NPC cell lines, CNE2, and 6-10B, and the impacts of EphA on NPC cell functions was assessed through Cell Counting Kit-8 (CCK-8), cell colony formation, as well as 5-Ethynyl-2'- deoxyuridine (EdU) assays. Finally, a potential interplay between EphA3 and FOG2 was also investigated. RESULTS: In this study, qRT-PCR results revealed that EphA3 expression levels in tumor tissues of patients with NPC were markedly higher than those in adjacent tissues. Compared with patients with low expression of EphA3, those with highly expressed EphA3 had a more advanced pathological stage. In addition, in vitro experiments showed that knocking down EphA3 notably attenuated the proliferation capacity of NPC cells. Subsequently, it was found that the expression of FOG2 in NPC cells was remarkably decreased both in NPC cell lines and tissues, which had a negative correlation with EphA3. Finally, cell recovery experiment revealed a mutual regulation between EphA3 and FOG2, which then together affected the malignant progression of NPC. CONCLUSIONS: EphA3 is significantly relevant to pathological staging and poor prognosis of patients with NPC and may enhance the proliferation ability of NPC cells by modulating FOG2.


Subject(s)
Cell Proliferation/physiology , DNA-Binding Proteins/biosynthesis , Nasopharyngeal Carcinoma/metabolism , Nasopharyngeal Neoplasms/metabolism , Receptor, EphA3/biosynthesis , Transcription Factors/biosynthesis , Adult , Cell Line, Transformed , Cell Line, Tumor , Female , Humans , Male , Middle Aged , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Neoplasms/pathology
2.
Cancer Res ; 74(16): 4470-81, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25125683

ABSTRACT

Eph receptor tyrosine kinases are critical for cell-cell communication during normal and oncogenic tissue patterning and tumor growth. Somatic mutation profiles of several cancer genomes suggest EphA3 as a tumor suppressor, but its oncogenic expression pattern and role in tumorigenesis remain largely undefined. Here, we report unexpected EphA3 overexpression within the microenvironment of a range of human cancers and mouse tumor xenografts where its activation inhibits tumor growth. EphA3 is found on mouse bone marrow-derived cells with mesenchymal and myeloid phenotypes, and activation of EphA3(+)/CD90(+)/Sca1(+) mesenchymal/stromal cells with an EphA3 agonist leads to cell contraction, cell-cell segregation, and apoptosis. Treatment of mice with an agonistic α-EphA3 antibody inhibits tumor growth by severely disrupting the integrity and function of newly formed tumor stroma and microvasculature. Our data define EphA3 as a novel target for selective ablation of the tumor microenvironment and demonstrate the potential of EphA3 agonists for anticancer therapy.


Subject(s)
Antibodies, Monoclonal/pharmacology , Receptor Protein-Tyrosine Kinases/agonists , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptor, EphA3/agonists , Receptor, EphA3/biosynthesis , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Transformation, Neoplastic , Disease Models, Animal , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Mice , Mice, Nude , Molecular Targeted Therapy , Receptor Protein-Tyrosine Kinases/immunology , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, EphA3/immunology , Receptor, EphA3/metabolism , Signal Transduction , Stromal Cells/drug effects , Stromal Cells/pathology , Tumor Microenvironment/drug effects
3.
PLoS One ; 7(6): e38566, 2012.
Article in English | MEDLINE | ID: mdl-22685584

ABSTRACT

BACKGROUND: Retinotopic projection onto the tectum/colliculus constitutes the most studied model of topographic mapping and Eph receptors and their ligands, the ephrins, are the best characterized molecular system involved in this process. Ephrin-As, expressed in an increasing rostro-caudal gradient in the tectum/colliculus, repel temporal retinal ganglion cell (RGC) axons from the caudal tectum and inhibit their branching posterior to their termination zones. However, there are conflicting data regarding the nature of the second force that guides nasal axons to invade and branch only in the caudal tectum/colliculus. The predominant model postulates that this second force is produced by a decreasing rostro-caudal gradient of EphA7 which repels nasal optic fibers and prevents their branching in the rostral tectum/colliculus. However, as optic fibers invade the tectum/colliculus growing throughout this gradient, this model cannot explain how the axons grow throughout this repellent molecule. METHODOLOGY/PRINCIPAL FINDINGS: By using chicken retinal cultures we showed that EphA3 ectodomain stimulates nasal RGC axon growth in a concentration dependent way. Moreover, we showed that nasal axons choose growing on EphA3-expressing cells and that EphA3 diminishes the density of interstitial filopodia in nasal RGC axons. Accordingly, in vivo EphA3 ectodomain misexpression directs nasal optic fibers toward the caudal tectum preventing their branching in the rostral tectum. CONCLUSIONS: We demonstrated in vitro and in vivo that EphA3 ectodomain (which is expressed in a decreasing rostro-caudal gradient in the tectum) is necessary for topographic mapping by stimulating the nasal axon growth toward the caudal tectum and inhibiting their branching in the rostral tectum. Furthermore, the ability of EphA3 of stimulating axon growth allows understanding how optic fibers invade the tectum growing throughout this molecular gradient. Therefore, opposing tectal gradients of repellent ephrin-As and of axon growth stimulating EphA3 complement each other to map optic fibers along the rostro-caudal tectal axis.


Subject(s)
Axons/metabolism , Receptor, EphA3/biosynthesis , Retinal Ganglion Cells/metabolism , Tectum Mesencephali/metabolism , Animals , Axons/physiology , Blotting, Western , Cells, Cultured , Chick Embryo , Chickens , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Immunohistochemistry , Microscopy, Confocal , Phosphorylation , Receptor, EphA3/genetics , Receptor, EphA3/metabolism , Retina/embryology , Retina/growth & development , Retina/metabolism , Superior Colliculi/embryology , Superior Colliculi/growth & development , Superior Colliculi/metabolism , Tectum Mesencephali/embryology , Tectum Mesencephali/growth & development , Time Factors , Time-Lapse Imaging , Tissue Culture Techniques , Tyrosine/metabolism , Visual Pathways
4.
Biochem Biophys Res Commun ; 405(4): 521-6, 2011 Feb 25.
Article in English | MEDLINE | ID: mdl-21276420

ABSTRACT

Deficiency of Smad3, an intracellular mediator of TGF-ß, was shown to significantly accelerate re-epithelialization of the colonic mucosa. This study was performed to investigate the molecular mechanisms by which Smad3 controls colonic epithelial cell proliferation and crypt formation. Smad3(ex8/ex8) C57BL/6 mice were used in this study and wild-type littermates served as controls. The number of proliferating cells in the isolated colonic epithelium of Smad3(-/-) mice was significantly increased compared to that in wild-type littermates. Protein levels of the cell cycle inhibitors p21 and p27 were significantly decreased, while that of c-Myc was increased in the isolated colonic epithelium from Smad3(-/-) mice. In the colonic tissue of wild-type mice, cell proliferation was restricted to the bottom of the crypts in accordance with nuclear ß-catenin staining, whereas proliferating cells were located throughout the crypts in Smad3(-/-) mice in accordance with nuclear ß-catenin staining, suggesting that Smad3 is essential for locating proliferating cells at the bottom of the colonic crypts. Notably, in Smad3(-/-) mice, there was loss of EphB2 and EphB3 receptor protein expression, critical regulators of proliferating cell positioning, while EphB receptor protein expression was confirmed at the bottom of the colonic crypts in wild-type mice. These observations indicated that disturbance of the EphB/ephrin B system brings about mispositioning of proliferating cells in the colonic crypts of Smad3(-/-) mice. In conclusion, Smad3 is essential for controlling number and positioning of proliferating cells in the colonic crypts and contributes to formation of a "proliferative zone" at the bottom of colonic crypts in the normal colon.


Subject(s)
Colon/physiology , Intestinal Mucosa/physiology , Receptors, Eph Family/biosynthesis , Smad3 Protein/physiology , Animals , Cell Proliferation , Cells, Cultured , Colon/cytology , Intestinal Mucosa/cytology , Ki-67 Antigen/metabolism , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Protein Biosynthesis , Receptor, EphA2/biosynthesis , Receptor, EphA3/biosynthesis , Smad3 Protein/genetics , Transforming Growth Factor beta/physiology
5.
Neuroscience ; 135(1): 97-109, 2005.
Article in English | MEDLINE | ID: mdl-16054765

ABSTRACT

The ophthalmic, maxillary and mandibular axon branches of the trigeminal ganglion provide cutaneous sensory innervation to the vertebrate face. In the chick embryo, the trigeminal ganglion is bilobed, with ophthalmic axons projecting from the ophthalmic lobe, while maxillary and mandibular projections emerge from the maxillomandibular lobe. To date, target tissue specific guidance cues that discriminately guide the axon projections from the two trigeminal ganglion lobes are unknown. EphA receptor tyrosine kinases and ephrin-A ligands are excellent candidates for this process as they are known to mediate axon guidance in the developing nervous system. Accordingly, the expression of EphAs and ephrin-As was investigated at stages 13, 15, 20 of chick embryogenesis when peripheral axons from the trigeminal ganglion are pathfinding. EphA3 is expressed highly in the ophthalmic trigeminal ganglion lobe neurons in comparison to maxillomandibular trigeminal ganglion lobe neurons. Furthermore, from stages 13-20 ephrin-A2 and ephrin-A5 ligands are only localized to the mesenchyme of the first branchial arch (maxillary and mandibular processes), the target fields for maxillomandibular trigeminal ganglion axons. We found that ophthalmic and not maxillomandibular lobe axons were responsive to ephrin-A5-Fc utilizing a substratum choice assay. The implication of these results is that EphA3 forward signaling in ophthalmic sensory axons may be an important mechanism in vivo for lobe specific guidance of trigeminal ganglion ophthalmic projections.


Subject(s)
Axons/metabolism , Ephrin-A5/pharmacology , Neurons, Afferent/metabolism , Ophthalmic Nerve/metabolism , Receptor, EphA3/biosynthesis , Trigeminal Nerve/metabolism , Animals , Axons/drug effects , Chick Embryo , Growth Cones/physiology , In Situ Hybridization , Neurons, Afferent/drug effects , Ophthalmic Nerve/cytology , Ophthalmic Nerve/growth & development , RNA/biosynthesis , RNA/genetics , Receptor, EphA3/genetics , Reverse Transcriptase Polymerase Chain Reaction , Trigeminal Nerve/cytology , Trigeminal Nerve/growth & development
6.
BMC Neurosci ; 5: 30, 2004 Aug 31.
Article in English | MEDLINE | ID: mdl-15339341

ABSTRACT

BACKGROUND: We examine results of gain-of-function experiments on retinocollicular maps in knock-in mice [Brown et al. (2000) Cell 102:77]. In wild-type mice the temporal-nasal axis of retina is mapped to the rostral-caudal axis of superior colliculus. The established map is single-valued, which implies that each point in retina maps to a unique termination zone in superior colliculus. In homozygous Isl2/EphA3 knock-in mice the map is double-valued, which means that each point on retina maps to two termination zones in superior colliculus. This is because about 50 percent of cells in retina express Isl2, and two types of projections, wild-type and Isl2/EphA3 positive, form two branches of the map. In heterozygous Isl2/EphA3 knock-ins the map is intermediate between the homozygous and wild-type: it is single-valued in temporal and double-valued in the nasal parts of retina. In this study we address possible reasons for such a bifurcation of the map. RESULTS: We study the map formation using stochastic model based on Markov chains. In our model the map undergoes a series of reconstructions with probabilities dependent upon a set of chemical cues. Our model suggests that the map in heterozygotes is single-valued in temporal region of retina for two reasons. First, the inhomogeneous gradient of endogenous receptor in retina makes the impact of exogenous receptor less significant in temporal retina. Second, the gradient of ephrin in the corresponding region of superior colliculus is smaller, which reduces the chemical signal-to-noise ratio. We predict that if gradient of ephrin is reduced by a genetic manipulation, the single-valued region of the map should extend to a larger portion of temporal retina, i.e. the point of transition between single-and double-valued maps should move to a more nasal position in Isl2-EphA3 heterozygotes. CONCLUSIONS: We present a theoretical model for retinocollicular map development, which can account for intriguing behaviors observed in gain-of-function experiments by Brown et al., including bifurcation in heterozygous Isl2/EphA3 knock-ins. The model is based on known chemical labels, axonal repulsion/competition, and stochasticity. Possible mapping in Isl2/EphB knock-ins is also discussed.


Subject(s)
Markov Chains , Models, Neurological , Retina/anatomy & histology , Superior Colliculi/anatomy & histology , Animals , Axons/physiology , Ephrin-B2/metabolism , Ephrin-B3/metabolism , Ephrins/metabolism , Heterozygote , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Homozygote , LIM-Homeodomain Proteins , Ligands , Mice , Mice, Transgenic , Receptor, EphA3/biosynthesis , Receptor, EphA3/genetics , Retina/growth & development , Retina/metabolism , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism , Stochastic Processes , Superior Colliculi/growth & development , Superior Colliculi/metabolism , Transcription Factors/biosynthesis , Transcription Factors/genetics
7.
J Neurosci ; 24(10): 2542-50, 2004 Mar 10.
Article in English | MEDLINE | ID: mdl-15014130

ABSTRACT

EphA tyrosine kinases are thought to act as topographically specific receptors in the well-characterized projection map from the retina to the tectum. Here, we describe a loss-of-function analysis of EphA receptors in retinotectal mapping. Expressing patches of a cytoplasmically truncated EphA3 receptor in chick retina caused temporal axons to have reduced responsiveness to posterior tectal repellent activity in vitro and to shift more posteriorly within the map in vivo. A gene disruption of mouse EphA5, replacing the intracellular domain with beta-galactosidase, reduced in vitro responsiveness of temporal axons to posterior target membranes. It also caused map abnormalities in vivo, with temporal axons shifted posteriorly and nasal axons anteriorly, but with the entire target still filled by retinal axons. The anterior shift of nasal axons was not accompanied by increased responsiveness to tectal repellent activity, in contrast to the comparable anterior shift in ephrin-A knock-outs, helping to resolve a previous ambiguity in interpreting the ephrin gene knock-outs. The results show the functional requirement for endogenous EphA receptors in retinotectal mapping, show that the receptor intracellular domain is required for a forward signaling response to topographic cues, and provide new evidence for a role of axon competition in topographic mapping.


Subject(s)
Receptors, Eph Family/physiology , Retinal Ganglion Cells/metabolism , Superior Colliculi/metabolism , Visual Pathways/metabolism , Animals , Axons/metabolism , Axons/physiology , Chick Embryo , Gene Targeting , Genes, Reporter , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Mice , Mice, Mutant Strains , Receptor, EphA3/biosynthesis , Receptor, EphA3/genetics , Receptor, EphA3/physiology , Receptor, EphA5/biosynthesis , Receptor, EphA5/genetics , Receptor, EphA5/physiology , Receptors, Eph Family/deficiency , Receptors, Eph Family/genetics , Retinal Ganglion Cells/cytology , Sequence Deletion , Superior Colliculi/cytology , Visual Pathways/cytology
8.
Mol Cell Neurosci ; 24(4): 984-99, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14697663

ABSTRACT

The Eph family of tyrosine kinase receptors and their ligands, ephrins, are distributed in gradients and serve as molecular guidance cues for axonal patterning during neuronal development. Most of these molecules are also expressed in mature brain. Thus, we examine here the potential roles of such molecules in plasticity and activity-dependent mossy fiber sprouting of adult CNS. We show that the ligand ephrin-A3 and the receptor EphA5 are expressed in complementary gradients in the adult rat mossy fiber system. Using the kindling model, we demonstrate that exogenous immunoadhesins that affect the interaction of endogenous EphA receptors and ephrin-A ligands modulate the development of kindling, one type of long-term plasticity, in mature rat brain. These immunoadhesins, combined with epileptogenic stimulations, alter both the extent and the pattern of collateral axonal sprouting in the mossy fiber pathway. Our results suggest that EphA receptors and ephrin-A ligands modify neuronal plasticity and may serve as spatial cues that modulate the development and pattern of activation-dependent axonal growth in adult CNS.


Subject(s)
Axons/metabolism , Epilepsy/metabolism , Kindling, Neurologic/metabolism , Receptor, EphA3/physiology , Receptor, EphA5/physiology , Animals , Epilepsy/genetics , Hippocampus/metabolism , Immunoglobulins/pharmacology , Kindling, Neurologic/genetics , Male , Neuronal Plasticity/physiology , Rats , Rats, Long-Evans , Receptor, EphA3/biosynthesis , Receptor, EphA3/genetics , Receptor, EphA5/biosynthesis , Receptor, EphA5/genetics
9.
Development ; 129(24): 5647-58, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12421705

ABSTRACT

Neural maps in the vertebrate central nervous system often show discontinuously segregated, domain-to-domain patterns. However, the molecular mechanism that establishes such maps is not well understood. Here we show that in the chicken olivocerebellar system, EphA receptors and ephrin-As are expressed with distinct levels and combinations in mapping domains. When ephrin-A2 is retrovirally overexpressed in the cerebellum, the olivocerebellar map is disrupted, excluding axons with high receptor activity from ectopic expression domains. Conversely, overexpression of a truncated EphA3 receptor in the cerebellum reduces endogenous ligand activity to undetectable levels and causes aberrant mapping, with high receptor axons invading high ligand domains. In vitro, ephrin-A2 inhibits outgrowth of inferior olive axons in a region-specific manner. These results suggest that Eph receptors and ephrins constitute domain-specific positional information, and the spatially accurate receptor-ligand interaction is essential to guide inferior olive axons to their correct target domains.


Subject(s)
Receptor, EphA2/metabolism , Receptor, EphA3/metabolism , Receptors, Eph Family/metabolism , Animals , Axons , Cell Line , Cell Membrane , Central Nervous System/embryology , Cerebellum/metabolism , Chickens , Humans , In Situ Hybridization , Ligands , Models, Biological , Protein Binding , Protein Structure, Tertiary , Purkinje Cells/metabolism , Receptor, EphA3/biosynthesis , Retroviridae/genetics , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...