Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Eur J Med Chem ; 216: 113318, 2021 Apr 15.
Article in English | MEDLINE | ID: mdl-33730624

ABSTRACT

Identifying a pharmacological agent that targets only one of more than 500 kinases present in humans is an important challenge. One potential solution to this problem is the development of bivalent kinase inhibitors, which consist of two connected fragments, each bind to a dissimilar binding site of the bisubstrate enzyme. The main advantage of bivalent (type V) kinase inhibitors is generating more interactions with target enzymes that can enhance the molecules' selectivity and affinity compared to single-site inhibitors. Earlier type V inhibitors were not suitable for the cellular environment and were mostly used in in vitro studies. However, recently developed bivalent compounds have high kinase affinity, high biological and chemical stability in vivo. This review summarized the hetero-bivalent kinase inhibitors described in the literature from 2014 to the present. We attempted to classify the molecules by serine/threonine and tyrosine kinase inhibitors, and then each target kinase and its hetero-bivalent inhibitor was assessed in depth. In addition, we discussed the analysis of advantages, limitations, and perspectives of bivalent kinase inhibitors compared with the monovalent kinase inhibitors.


Subject(s)
Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/metabolism , Receptor, EphA1/antagonists & inhibitors , Receptor, EphA1/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
2.
Brain Inj ; 33(10): 1385-1401, 2019.
Article in English | MEDLINE | ID: mdl-31319723

ABSTRACT

Primary Objective: Eph/Ephrin signaling is inhibitory for developing axons and blocking Eph pathways enhances regeneration after spinal cord injury. It was hypothesized that inhibition of Eph signaling promotes cellular and behavioral recovery after traumatic brain injury (TBI). Research design: Lateral fluid percussion (LFP) injury was performed on wildtype (WT) and EphA6 knockout (KO) mice. EphA6-Fc, Ephrin-A5-Fc fusion proteins, and sodium orthovanadate were used to alter the signaling pathway. Immunohistochemistry and tissue explants revealed cellular changes. Rotarod tests demonstrated vestibulomotor function. Outcomes: The EphA6 receptor expression is upregulated following LFP. Uninjured EphA6 KO mice exhibit greater neurite density and clustered Ephrin-A5-Fc causes growth cone collapse in vitro. After LFP, EphA6 KO mice demonstrate longer neurites and decreased neuronal cell death and astrocytosis compared to WT mice. Blocking EphA signaling by soluble EphA6-Fc fusion protein reduces cell death and improves motor function following LFP whereas clustered Ephrin-A5-Fc exacerbates cell death and neurodegeneration. Sodium orthovanadate rescues growth cone collapse in vitro as well as cell death and neurodegeneration in vivo. Conclusions: Eph/Ephrin signaling plays an inhibitory role following TBI. Targeting the Eph signaling pathway with Fc fusion proteins and pharmacological agents can be a novel strategy to counter the damaging effects of TBI. Abbreviations: LFP: lateral fluid percussion; TBI: traumatic brain injury; KO: knockout; WT: wildtype; PTP2: protein phosphotyrosine phosphatase 2; Tg: transgenic; YFP: yellow fluorescent protein; ATM: atmospheres; RT-qPCR: Real-time-quantitative PCR; dpi: days post injury; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; DAPI: 4',6-diamidino-2-phenylindole; PBS: phosphate buffered saline; GFAP: glial fibrillary acidic protein; FLJC: fluorojade C; CA: cornu ammonis; SEM: standard error of the mean; ANOVA: analysis of variance; PLSD: posthoc least significant difference.


Subject(s)
Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/therapy , Genetic Therapy/methods , Receptor, EphA1/antagonists & inhibitors , Receptor, EphA1/genetics , Animals , Astrocytes/pathology , Brain Injuries, Traumatic/pathology , Cell Death , Immunoglobulin G/pharmacology , Male , Mice , Mice, Knockout , Nerve Degeneration/genetics , Nerve Degeneration/prevention & control , Neurites/pathology , Neurons/metabolism , Postural Balance , Receptor, EphA1/biosynthesis , Signal Transduction/drug effects , Signal Transduction/genetics , Vanadates/therapeutic use
3.
Exp Eye Res ; 170: 92-100, 2018 05.
Article in English | MEDLINE | ID: mdl-29476773

ABSTRACT

The corneal endothelium is a monolayer of epithelial cells that lines the posterior surface of the cornea and is essential for maintenance of corneal transparency. Wound healing within the corneal endothelium typically occurs through cell spreading and migration rather than through proliferation. The mechanisms that control corneal endothelial cell migration are unclear. In this study we demonstrate that cultures of corneal endothelial cells display reduced migration in scratch wound assays, and reduced levels of E-cadherin mRNA, following suppression of ligand-activated Eph receptor signalling by treatment with lithocholic acid. Two Eph receptors, EphA1 and EphA2, were subsequently detected in corneal endothelial cells, and their potential involvement during migration was explored through gene silencing using siRNAs. EphA2 siRNA reduced levels of mRNA for both EphA2 and N-cadherin, but increased levels of mRNA for both EphA1 and E-cadherin. No effect, however, was observed for EphA2 siRNA on migration. Our results indicate a potential role for Eph receptor signalling during corneal endothelial cell migration via changes in cadherin expression. Nevertheless, defining a precise role for select Eph receptors is likely to be complicated by crosstalk between Eph-mediated signalling pathways.


Subject(s)
Cell Movement/physiology , Endothelium, Corneal/cytology , Receptor, EphA1/physiology , Receptor, EphA2/physiology , Signal Transduction/physiology , Animals , Cadherins/metabolism , Cell Line , Cell Survival , Detergents/pharmacology , Endothelium, Corneal/drug effects , Endothelium, Corneal/metabolism , Ephrin-A1/genetics , Ephrin-A1/metabolism , Fluorescent Antibody Technique, Indirect , Gene Silencing , Humans , Lithocholic Acid/pharmacology , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Receptor, EphA1/antagonists & inhibitors , Receptor, EphA2/antagonists & inhibitors , Sheep
4.
Sci Rep ; 8(1): 1539, 2018 01 24.
Article in English | MEDLINE | ID: mdl-29367676

ABSTRACT

Angiogenesis plays an important role in bladder cancer (BCa). The immunosuppressive drug leflunomide has attracted worldwide attention. However, the effects of leflunomide on angiogenesis in cancer remain unclear. Here, we report the increased expression of soluble ephrin-A1 (sEphrin-A1) in supernatants of BCa cell lines (RT4, T24, and TCCSUP) co-cultured with human umbilical vein endothelial cells (HUVECs) compared with that in immortalized uroepithelial cells (SV-HUC-1) co-cultured with HUVECs. sEphrin-A1 is released from BCa cells as a monomeric protein that is a functional form of the ligand. The co-culture supernatants containing sEphrin-A1 caused the internalization and down-regulation of EphA2 on endothelial cells and dramatic functional activation of HUVECs. This sEphrin-A1/EphA2 system is mainly functional in regulating angiogenesis in BCa tissue. We showed that leflunomide (LEF) inhibited angiogenesis in a N-butyl-N-(4-hydroxybutyl)-nitrosamine (BBN)-induced bladder carcinogenesis model and a tumor xenograft model, as well as in BCa cell and HUVEC co-culture systems, via significant inhibition of the sEphrin-A1/EphA2 system. Ephrin-A1 overexpression could partially reverse LEF-induced suppression of angiogenesis and subsequent tumor growth inhibition. Thus, LEF has a significant anti-angiogenesis effect on BCa cells and BCa tissue via its inhibition of the functional angiogenic sEphrin-A1/EphA2 system and may have potential for treating BCa beyond immunosuppressive therapy.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Endothelial Cells/drug effects , Leflunomide/administration & dosage , Neovascularization, Pathologic/drug therapy , Receptor, EphA1/antagonists & inhibitors , Receptor, EphA2/antagonists & inhibitors , Urinary Bladder Neoplasms/drug therapy , Angiogenesis Inhibitors/pharmacology , Animals , Cell Line, Tumor , Coculture Techniques , Disease Models, Animal , Endothelial Cells/physiology , Epithelial Cells/physiology , Heterografts , Humans , Leflunomide/pharmacology , Models, Biological , Neoplasm Transplantation , Treatment Outcome
5.
Sci Rep ; 7(1): 6519, 2017 07 26.
Article in English | MEDLINE | ID: mdl-28747680

ABSTRACT

Eph receptors have emerged as targets for therapy in both neoplastic and non-neoplastic disease, however, particularly in non-neoplastic diseases, redundancy of function limits the effectiveness of targeting individual Eph proteins. We have shown previously that a soluble fusion protein, where the EphA4 ectodomain was fused to IgG Fc (EphA4 Fc), was an effective therapy in acute injuries and demonstrated that EphA4 Fc was a broad spectrum Eph/ephrin antagonist. However, a very short in vivo half-life effectively limited its therapeutic development. We report a unique glycoengineering approach to enhance the half-life of EphA4 Fc. Progressive deletion of three demonstrated N-linked sites in EphA4 progressively increased in vivo half-life such that the triple mutant protein showed dramatically improved pharmacokinetic characteristics. Importantly, protein stability, affinity for ephrin ligands and antagonism of cell expressed EphA4 was fully preserved, enabling it to be developed as a broad spectrum Eph/ephrin antagonist for use in both acute and chronic diseases.


Subject(s)
Immunoglobulin Fc Fragments/metabolism , Receptor, EphA1/antagonists & inhibitors , Receptor, EphA4/metabolism , Receptor, EphA4/pharmacokinetics , DNA Mutational Analysis , Glycosylation , Half-Life , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin Fc Fragments/genetics , Mutagenesis, Site-Directed , Protein Binding , Receptor, EphA4/chemistry , Receptor, EphA4/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacokinetics
6.
Molecules ; 18(10): 13043-60, 2013 Oct 21.
Article in English | MEDLINE | ID: mdl-24152675

ABSTRACT

The Eph-ephrin system plays a critical role in tumor growth and vascular functions during carcinogenesis. We had previously identified cholanic acid as a competitive and reversible EphA2 antagonist able to disrupt EphA2-ephrinA1 interaction and to inhibit EphA2 activation in prostate cancer cells. Herein, we report the synthesis and biological evaluation of a set of cholanic acid derivatives obtained by conjugation of its carboxyl group with a panel of naturally occurring amino acids with the aim to improve EphA2 receptor inhibition. Structure-activity relationships indicate that conjugation of cholanic acid with linear amino acids of small size leads to effective EphA2 antagonists whereas the introduction of aromatic amino acids reduces the potency in displacement studies. The b-alanine derivative 4 was able to disrupt EphA2-ephrinA1 interaction in the micromolar range and to dose-dependently inhibit EphA2 activation on PC3 cells. These findings may help the design of novel EphA2 antagonists active on cancer cell lines.


Subject(s)
Cholic Acids/pharmacology , Receptor, EphA2/antagonists & inhibitors , Binding Sites , Cell Line, Tumor , Cholic Acids/chemical synthesis , Cholic Acids/chemistry , Humans , Hydrogen Bonding , Inhibitory Concentration 50 , Molecular Docking Simulation , Phosphorylation , Protein Binding , Protein Processing, Post-Translational/drug effects , Protein Structure, Secondary , Receptor, EphA1/antagonists & inhibitors , Receptor, EphA1/chemistry , Receptor, EphA1/metabolism , Receptor, EphA2/chemistry , Receptor, EphA2/metabolism , Structure-Activity Relationship
7.
Biochim Biophys Acta ; 1835(2): 243-57, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23396052

ABSTRACT

Although at present, there is a high incidence of prostate cancer, particularly in the Western world, mortality from this disease is declining and occurs primarily only from clinically significant late stage tumors with a poor prognosis. A major current focus of this field is the identification of new biomarkers which can detect earlier, and more effectively, clinically significant tumors from those deemed "low risk", as well as predict the prognostic course of a particular cancer. This strategy can in turn offer novel avenues for targeted therapies. The large family of Receptor Tyrosine Kinases, the Ephs, and their binding partners, the ephrins, has been implicated in many cancers of epithelial origin through stimulation of oncogenic transformation, tumor angiogenesis, and promotion of increased cell survival, invasion and migration. They also show promise as both biomarkers of diagnostic and prognostic value and as targeted therapies in cancer. This review will briefly discuss the complex roles and biological mechanisms of action of these receptors and ligands and, with regard to prostate cancer, highlight their potential as biomarkers for both diagnosis and prognosis, their application as imaging agents, and current approaches to assessing them as therapeutic targets. This review demonstrates the need for future studies into those particular family members that will prove helpful in understanding the biology and potential as targets for treatment of prostate cancer.


Subject(s)
Ephrins/physiology , Prostatic Neoplasms/drug therapy , Receptor, EphA1/physiology , Biomarkers , Ephrins/analysis , Humans , Male , Neoplastic Cells, Circulating/chemistry , Prostatic Neoplasms/etiology , Receptor, EphA1/analysis , Receptor, EphA1/antagonists & inhibitors , Signal Transduction
8.
Mol Biol Cell ; 22(18): 3508-19, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21795402

ABSTRACT

Insulin-like growth factor-I (IGF-I) activates not only the phosphatidylinositol 3-kinase (PI3K)-AKT cascade that is essential for myogenic differentiation but also the extracellular signal-regulated kinase (ERK) 1/2 cascade that inhibits myogenesis. We hypothesized that there must be a signal that inhibits ERK1/2 upon cell-cell contact required for skeletal myogenesis. Cell-cell contact-induced engagement of ephrin ligands and Eph receptors leads to downregulation of the Ras-ERK1/2 pathway through p120 Ras GTPase-activating protein (p120RasGAP). We therefore investigated the significance of the ephrin/Eph signal in IGF-I-induced myogenesis. EphrinA1-Fc suppressed IGF-I-induced activation of Ras and ERK1/2, but not that of AKT, in C2C12 myoblasts, whereas ephrinB1-Fc affected neither ERK1/2 nor AKT activated by IGF-I. IGF-I-dependent myogenic differentiation of C2C12 myoblasts was potentiated by ephrinA1-Fc. In p120RasGAP-depleted cells, ephrinA1-Fc failed to suppress the Ras-ERK1/2 cascade by IGF-I and to promote IGF-I-mediated myogenesis. EphrinA1-Fc did not promote IGF-I-dependent myogenesis when the ERK1/2 was constitutively activated. Furthermore, a dominant-negative EphA receptor blunted IGF-I-induced myogenesis in C2C12 and L6 myoblasts. However, the inhibition of IGF-I-mediated myogenesis by down-regulation of ephrinA/EphA signal was canceled by inactivation of the ERK1/2 pathway. Collectively, these findings demonstrate that the ephrinA/EphA signal facilitates IGF-I-induced myogenesis by suppressing the Ras-ERK1/2 cascade through p120RasGAP in myoblast cell lines.


Subject(s)
Cell Differentiation , Insulin-Like Growth Factor I/physiology , MAP Kinase Signaling System , Myoblasts/physiology , Receptor, EphA1/metabolism , Receptor, EphA2/metabolism , Animals , Cell Fusion , Cell Line , Ephrins/metabolism , Gene Knockdown Techniques , Immunoglobulin Fc Fragments/pharmacology , Insulin-Like Growth Factor I/pharmacology , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Myoblasts/metabolism , Myosin Heavy Chains/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , RNA Interference , Rats , Receptor, EphA1/antagonists & inhibitors , Receptor, EphA2/genetics , Sequence Deletion , p120 GTPase Activating Protein/genetics , p120 GTPase Activating Protein/metabolism
9.
Oncol Rep ; 23(2): 563-70, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20043122

ABSTRACT

The Eph family of receptor tyrosine kinases has emerged as one of the pivotal regulators of tumor angiogenesis. EphA1, the first identified member of the Eph receptor family, has been found to be overexpressed in several types of human tumors. A recent report indicated that EphA1 was overexpressed in hepatocellular carcinoma (HCC) and that elevated expression of EphA1 can promote proliferation of HCC cells through stimulation by exogenous Ephrin-A1. To investigate the role of EphA1 in angiogenesis and progression of HCC, we down-regulated EphA1 by RNA interference (RNAi) technology, in an HCC-derived cell line with a high level of EphA1 expression. We established a stable knockdown clone named SiEphA1/Huh-7. The knockdown resulted in decreased proliferation of Huh-7 cells, as well as decreased motility and invasion capability in vitro. siRNA-based EphA1 knockdown also down-regulated the expression of vascular endothelial growth factor (VEGF) and matrix metalloproteinase (MMP)-2 and -9. Interestingly, the suppression of EphA1 expression in Huh-7 cells reduced their outgrowth when inoculated in the subcutaneous space in the flank of nude mice, presumably through angiogenesis inhibition since microvessel density was found to be inhibited.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , RNA, Small Interfering/therapeutic use , Receptor, EphA1/antagonists & inhibitors , Angiogenesis Inhibitors/pharmacology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Liver Neoplasms/blood supply , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , NIH 3T3 Cells , RNA, Small Interfering/pharmacology , Receptor, EphA1/genetics , Treatment Outcome , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A/genetics , Xenograft Model Antitumor Assays
10.
Angiogenesis ; 7(1): 17-28, 2004.
Article in English | MEDLINE | ID: mdl-15302992

ABSTRACT

Angiogenesis, the process by which new blood vessels sprout and branch from existing vasculature, is crucial for vascular remodeling during embryogenesis and in normal tissue homeostasis, such as in the female reproductive tract. Angiogenesis can also contribute to the pathogenesis of diseases such as cancer and retinopathy. The Eph family of receptor tyrosine kinases and their ligands, called ephrins, has emerged as critical regulators of vascular remodeling in the embryo. More recently, these molecules have been associated with post-natal angiogenic remodeling and tumor neovascularization. This review provides an overview of recent advances in our understanding of Eph/ephrins in angiogenesis, with an emphasis on development and disease, and the potential for targeting these molecules in anti-angiogenic therapy.


Subject(s)
Neovascularization, Pathologic/etiology , Neovascularization, Physiologic/physiology , Receptor, EphA1/physiology , Animals , Humans , Ligands , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/enzymology , Neovascularization, Physiologic/drug effects , Receptor, EphA1/antagonists & inhibitors
11.
J Immunol ; 172(2): 843-50, 2004 Jan 15.
Article in English | MEDLINE | ID: mdl-14707054

ABSTRACT

Gene expression screening showed decreased ephrin-A1 expression in CD4+ T cells of asthma patients. Ephrin-A1 is the ligand of the Eph receptor family of tyrosine kinases, forming the largest family of receptor tyrosine kinases. Their immune regulatory properties are largely unknown. This study demonstrates significantly reduced ephrin-A1 expression in T cells of asthma patients using real time-PCR. Immunohistological analyses revealed strong ephrin-A1 expression in lung tissue and low expression in cortical areas of lymph nodes. It is absent in T cell/B cell areas of the spleen. Colocalization of ephrin-A1 and its receptors was found only in the lung, but not in lymphoid tissues. In vitro activation of T cells reduced ephrin-A1 at mRNA and protein levels. T cell proliferation, activation-induced, and IL-2-dependent cell death were inhibited by cross-linking ephrin-A1, and not by engagement of Eph receptors. However, anti-EphA1 receptor slightly enhances Ag-specific and polyclonal proliferation of PBMC cultures. Furthermore, activation-induced CD25 up-regulation was diminished by ephrin-A1 engagement. Ephrin-A1 engagement reduced IL-2 expression by 82% and IL-4 reduced it by 69%; the IFN-gamma expression remained unaffected. These results demonstrate that ephrin-A1 suppresses T cell activation and Th2 cytokine expression, while preventing activation-induced cell death. The reduced ephrin-A1 expression in asthma patients may reflect the increased frequency of activated T cells in peripheral blood. That the natural ligands of ephrin-A1 are most abundantly expressed in the lung may be relevant for Th2 cell regulation in asthma and Th2 cell generation by mucosal allergens.


Subject(s)
Down-Regulation/physiology , Ephrin-A1/physiology , Lung/cytology , Lung/physiology , Lymphocyte Activation/physiology , Respiratory Mucosa/cytology , Respiratory Mucosa/physiology , Th2 Cells/physiology , Animals , Asthma/immunology , Asthma/metabolism , Asthma/pathology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Cell Death/immunology , Cell Division/physiology , Cells, Cultured , Ephrin-A1/biosynthesis , Ephrin-A1/metabolism , Growth Inhibitors/biosynthesis , Growth Inhibitors/metabolism , Growth Inhibitors/physiology , Humans , Immune Sera/pharmacology , Interferon-gamma/biosynthesis , Interleukin-2/antagonists & inhibitors , Interleukin-2/biosynthesis , Interleukin-4/antagonists & inhibitors , Interleukin-4/biosynthesis , Lung/immunology , Lymphoid Tissue/metabolism , Mice , Mice, Inbred BALB C , Receptor, EphA1/antagonists & inhibitors , Receptor, EphA1/immunology , Respiratory Mucosa/immunology , Th2 Cells/cytology , Th2 Cells/immunology , Th2 Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...