Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Phys Chem B ; 126(26): 4914-4924, 2022 07 07.
Article in English | MEDLINE | ID: mdl-35732074

ABSTRACT

Protein-protein interaction plays an important role in the development of almost all cells. Elucidating the dynamic binding and affinity of a protein-protein complex is essential for understanding the biological functions of proteins. EphA6 and Odin proteins are members of the Eph (erythropoietin-producing hepatocyte) receptor family and the Anks (ankyrin repeat and sterile α motif domain-containing) family, respectively. Odin significantly functions in regulating endocytosis, degradation, and stability of EphA receptors. In this work, the key residues of the interaction interface were determined through a hydrogen bond, root-mean-square deviation (RMSD), root-mean-square fluctuation (RMSF), and dynamic correlation analysis of the conventional molecular dynamics (MD) simulations. The calculated standard binding free energy, -7.92 kcal/mol, between EphA6 and Odin is quite consistent with the experimental measurement value, -8.73 kcal/mol. By the combination of several MD simulation techniques, our investigation of the binding process reveals the detailed representative characteristics of the entire binding pathway at the molecular level. Based on the obtained potential of the mean force (PMF) curve, the analysis of the simulation trajectories shows that the residue Arg1013 in the receptor EphA6 is responsible for capturing Asp739 and Asp740 in the ligand Odin during the initial stage of binding. In the later stage of binding, the hydrogen bonds and salt bridges between a series of residues Lys973, Leu1007, Gly1009, His1010, and Arg1012 in the receptor and residues Leu735, Asn736, Asp739, Asp740, and Asp753 in the ligand mainly contribute to the stability of the protein complex. In addition, the specific change process of the receptor-ligand-binding mode is also clarified during the binding process. Our present simulation will promote a deep understanding of the protein-protein interaction, and the identified key interresidue interaction will be theoretical guidance for the design of protein drugs.


Subject(s)
Molecular Dynamics Simulation , Receptor, EphA6 , Carrier Proteins/metabolism , Ligands , Molecular Docking Simulation , Protein Binding , Proteins/metabolism , Receptor, EphA6/metabolism
2.
Article in English | MEDLINE | ID: mdl-32843430

ABSTRACT

Although cutaneous squamous cell carcinoma (cSCC) is treatable in the majority of cases, deadly invasive and metastatic cases do occur. To date there are neither reliable predictive biomarkers of disease progression nor FDA-approved targeted therapies as standard of care. To address these issues, we screened patient-derived primary cultured cells from invasive/metastatic cSCC with 107 small-molecule inhibitors. In-house bioinformatics tools were used to cross-analyze drug responses and DNA mutations in tumors detected by whole-exome sequencing (WES). Aberrations in molecular pathways with evidence of potential drug targets were identified, including the Eph-ephrin and neutrophil degranulation signaling pathways. Using a screening panel of siRNAs, we identified EPHA6 and EPHA7 as targets within the Eph-ephrin pathway responsible for mitigating decreased cell viability. These studies form a plausible foundation for detecting biomarkers of high-risk progressive disease applicable in dermatopathology and for patient-specific therapeutic options for invasive/metastatic cSCC.


Subject(s)
Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Aged , Aged, 80 and over , Carcinoma, Squamous Cell/metabolism , Cell Survival/genetics , Disease Progression , Gene Expression/genetics , Gene Expression Regulation, Neoplastic/genetics , Genomics/methods , Humans , Male , Middle Aged , Mutation/genetics , Receptor, EphA6/antagonists & inhibitors , Receptor, EphA6/metabolism , Receptor, EphA7/antagonists & inhibitors , Receptor, EphA7/metabolism , Signal Transduction/genetics , Skin Neoplasms/genetics , Small Molecule Libraries/pharmacology , Exome Sequencing/methods
3.
Cancer Sci ; 110(11): 3486-3496, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31483918

ABSTRACT

Bone morphogenetic protein (BMP) signaling plays important roles in glioblastoma multiforme (GBM), a lethal form of brain tumor. BMP reduces GBM tumorigenicity through its differentiation- and apoptosis-inducing effects on glioma-initiating cells (GIC). However, some GIC do not respond to the tumor suppressive effects of BMP. Using a phosphoreceptor tyrosine kinase array, we found that EPHA6 (erythropoietin-producing hepatocellular carcinoma receptor A6) phosphorylation was regulated by BMP-2 signaling in some GIC. Analysis of The Cancer Genome Atlas showed that EPHA6 expression was lower in patients with GBM than in the normal brain, and that high EPHA6 expression was correlated with better prognosis. EPHA6 receptor increased the susceptibility of both sensitive and resistant GIC to BMP-2-induced apoptosis. The cooperative effect on apoptosis induction depended on the kinase activity of BMP type I receptor but was independent of EPHA6 kinase function. Overexpression of the EPHA6 receptor in GIC resulted in the formation of a protein complex of EPHA6 receptor and the BMP type I receptor ALK-2, which was associated with BMP-induced apoptosis in GIC. Intracranial injection of GIC into nude mice showed that gain-of-function of EPHA6 together with BMP-2 pretreatment slowed GBM tumor progression in the mouse brain and promoted mouse survival. In summary, EPHA6 together with BMP-2 signaling led to apoptotic cell death in GIC, and thus is a putative tumor suppressor in GBM.


Subject(s)
Activin Receptors, Type I/metabolism , Apoptosis , Bone Morphogenetic Protein 2/metabolism , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Receptor, EphA6/metabolism , Animals , Bone Morphogenetic Protein 2/pharmacology , Bone Morphogenetic Protein Receptors, Type I/metabolism , Bone Morphogenetic Proteins/metabolism , Brain/drug effects , Brain/metabolism , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Cell Line, Tumor , Disease Progression , Glioblastoma/mortality , Glioblastoma/pathology , Glioma/metabolism , Glioma/pathology , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Phosphorylation , Prognosis , Tumor Suppressor Proteins/metabolism
4.
Oncotarget ; 6(26): 22587-97, 2015 Sep 08.
Article in English | MEDLINE | ID: mdl-26041887

ABSTRACT

Metastasis is the primary cause of prostate cancer (CaP)-related death. We investigate the molecular, pathologic and clinical outcome associations of EphA6 expression and CaP metastasis. The expression profiling of Eph receptors (Ephs) and their ephrin ligands was performed in parental and metastatic CaP cell lines. Among Ephs and ephrins, only EphA6 is consistently overexpressed in metastatic CaP cells. Metastatic potential of EphA6 is assessed by RNAi in a CaP spontaneous metastasis mouse model. EphA6 knock-down in human PC-3M cells causes decreased invasion in vitro and reduced lung and lymph node metastasis in vivo. In addition, knock-down of EphA6 decreases tube formation in vitro and angiogenesis in vivo. EphA6 mRNA expression is higher in 112 CaP tumor samples compared with benign tissues from 58 benign prostate hyperplasia patients. Positive correlation was identified between EphA6 expression and vascular invasion, neural invasion, PSA level, and TNM staging in CaP cases. Further, genome-wide gene expression analysis in EphA6 knock-down cells identified a panel of differentially regulated genes including PIK3IPA, AKT1, and EIF5A2, which could contribute to EphA6-regulated cancer progression. These findings identify EphA6 as a potentially novel metastasis gene which positively correlates with CaP progression. EphA6 may be a therapeutic target in metastatic CaP.


Subject(s)
Prostatic Neoplasms/blood supply , Prostatic Neoplasms/metabolism , Receptor, EphA6/metabolism , Animals , Cell Line, Tumor , Disease Progression , Heterografts , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Transfection
5.
PLoS One ; 9(6): e100096, 2014.
Article in English | MEDLINE | ID: mdl-24945320

ABSTRACT

HMX1 is a homeobox-containing transcription factor implicated in eye development and responsible for the oculo-auricular syndrome of Schorderet-Munier-Franceschetti. HMX1 is composed of two exons with three conserved domains in exon 2, a homeobox and two domains called SD1 and SD2. The function of the latter two domains remains unknown. During retinal development, HMX1 is expressed in a polarized manner and thus seems to play a role in the establishment of retinal polarity although its exact role and mode of action in eye development are unknown. Here, we demonstrated that HMX1 dimerized and that the SD1 and homeodomains are required for this function. In addition, we showed that proper nuclear localization requires the presence of the homeodomain. We also identified that EPHA6, a gene implicated in retinal axon guidance, is one of its targets in eye development and showed that a dimerized HMX1 is needed to inhibit EPHA6 expression.


Subject(s)
Homeodomain Proteins/metabolism , Protein Multimerization , Receptor, EphA4/metabolism , Receptor, EphA6/metabolism , Retina/metabolism , Transcription Factors/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Cell Nucleus/metabolism , Green Fluorescent Proteins/metabolism , HEK293 Cells , Homeodomain Proteins/chemistry , Humans , Mice, Inbred C57BL , Mutant Proteins/metabolism , Promoter Regions, Genetic/genetics , Protein Structure, Tertiary , Protein Transport , Transcription Factors/chemistry , Zebrafish Proteins/chemistry
6.
Dev Dyn ; 236(4): 951-60, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17304517

ABSTRACT

Hypospadias, a common defect affecting the growth and closure of the external genitalia, is often accompanied by gross enlargements of the genital tubercle (GT) vasculature. Because Hoxa13 homozygous mutant mice also exhibit hypospadias and GT vessel expansion, we examined whether genes playing a role in angiogenesis exhibit reduced expression in the GT. From this analysis, reductions in EphA6 and EphA7 were detected. Characterization of EphA6 and EphA7 expression in the GT confirmed colocalization with HOXA13 in the GT vascular endothelia. Analysis of the EphA6 and EphA7 promoter regions revealed a series of highly conserved cis-regulatory elements bound by HOXA13 with high affinity. GT chromatin immunoprecipitation confirmed that HOXA13 binds these gene-regulatory elements in vivo. In vitro, HOXA13 activates gene expression through the EphA6 and EphA7 gene-regulatory elements. Together these findings indicate that HOXA13 directly regulates EphA6 and EphA7 in the developing GT and identifies the GT vascular endothelia as a novel site for HOXA13-dependent expression of EphA6 and EphA7.


Subject(s)
Endothelium, Vascular/embryology , Gene Expression Regulation, Developmental , Genitalia/blood supply , Genitalia/embryology , Homeodomain Proteins/physiology , Receptor, EphA6/genetics , Receptor, EphA7/genetics , Animals , Base Sequence , Binding Sites , Cells, Cultured , Endothelium, Vascular/metabolism , Genitalia/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Mice, Mutant Strains , Molecular Sequence Data , Promoter Regions, Genetic , Receptor, EphA6/metabolism , Receptor, EphA7/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Homology, Nucleic Acid , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...