Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
ACS Infect Dis ; 9(8): 1582-1592, 2023 08 11.
Article in English | MEDLINE | ID: mdl-37415514

ABSTRACT

The HIV-1 infection epidemic remains a global health problem. Current antiretroviral treatments are effective in controlling the progression of a severe infection. However, the emergence of drug resistance requires an urgent identification of new treatment regimes. HIV-1 reverse transcriptase (RTs) has been a successful therapeutic target owing to its high specificity and potent antiviral properties; therefore, it has become an essential component of current HIV-1 standard treatments. This study identified a new HIV-1 RTs inhibitor (Compound #8) that is structurally unique and greatly effective against HIV-1 through chemical library screening and a medicinal chemistry program by analyzing the structure-activity relationship (SAR). Further analysis of molecular docking and mechanisms of action demonstrated that Compound #8 is a novel type of HIV-1 non-nucleoside reverse transcriptase inhibitor (NNRTI) with a flexible binding mode. Therefore, it exhibits great therapeutic potential when combined with other existing HIV-1 drugs. Our current studies suggest that Compound #8 is a promising novel scaffold for the development of new HIV-1 treatments.


Subject(s)
HIV Infections , HIV-1 , Humans , Reverse Transcriptase Inhibitors/pharmacology , Reverse Transcriptase Inhibitors/therapeutic use , Reverse Transcriptase Inhibitors/chemistry , Molecular Docking Simulation , Antiviral Agents/pharmacology , HIV Infections/drug therapy
2.
Int J Mol Sci ; 23(21)2022 Nov 04.
Article in English | MEDLINE | ID: mdl-36362286

ABSTRACT

Although the pathogenesis of atopic dermatitis (AD) remains to be fully deciphered, skin barrier abnormality and immune dysregulation are known to be involved. Recently, the vascular endothelial growth factor (VEGF)/VEGF receptor (VEGFR) system has also been implicated in the pathogenesis of this multifactorial chronic inflammatory skin disorder. Previously, we showed that a novel tetrapeptide, N-acetyl-Arg-Leu-Tyr-Glu (Ac-RLYE), inhibits angiogenesis and vascular permeability effectively by selectively antagonizing VEGFR-2. The current study aimed to investigate the pharmacological effect of Ac-RLYE on AD in vitro and in vivo. The in vitro experiments demonstrated that Ac-RLYE inhibited VEGF-induced vascular permeability in endothelial cells. Moreover, in an in vivo animal model of AD, Ac-RLYE relieved AD-like symptoms such as ear thickness and dermatitis severity scores and infiltration of immune cells, including mast cells and eosinophils. Ac-RLYE inhibited IgE secretion, restored the skin barrier protein filaggrin level, and markedly downregulated gene expression of AD-related Th1, Th2, and Th17 cytokines. Collectively, these findings suggest that Ac-RLYE would be useful for the treatment of AD and associated inflammatory skin disorders.


Subject(s)
Dermatitis, Atopic , Mice , Animals , Dermatitis, Atopic/pathology , Vascular Endothelial Growth Factor A/metabolism , Capillary Permeability , Disease Models, Animal , Endothelial Cells/metabolism , Skin/metabolism , Administration, Topical , Cytokines/metabolism , Immunity
3.
BMB Rep ; 55(12): 639-644, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36330710

ABSTRACT

Serine-arginine-rich splicing factors (SRSFs) are members of RNA processing proteins in the serine-arginine-rich (SR) family that could regulate the alternative splicing of the human immunodeficiency virus-1 (HIV-1). Whether SRSF9 has any effect on HIV-1 regulation requires elucidation. Here, we report for the first time the effects and mechanisms of SRSF9 on HIV-1 regulation. The overexpression of SRSF9 inhibits viral production and infectivity in both HEK293T and MT-4 cells. Deletion analysis of SRSF9 determined that the RNA regulation motif domain of SRSF9 is important for anti-HIV-1 effects. Furthermore, overexpression of SRSF9 increases multiple spliced forms of viral mRNA, such as Vpr mRNA. These data suggest that SRSF9 overexpression inhibits HIV-1 production by inducing the imbalanced HIV-1 mRNA splicing that could be exploited further for a novel HIV-1 therapeutic molecule. [BMB Reports 2022; 55(12): 639-644].


Subject(s)
HIV-1 , Serine-Arginine Splicing Factors , Humans , Alternative Splicing/genetics , HEK293 Cells , HIV-1/metabolism , RNA Splicing/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Serine-Arginine Splicing Factors/genetics , Serine-Arginine Splicing Factors/metabolism
4.
Int J Mol Sci ; 23(20)2022 Oct 15.
Article in English | MEDLINE | ID: mdl-36293198

ABSTRACT

The positive transcription elongation factor b (P-TEFb) is an essential factor that induces transcription elongation and is also negatively regulated by the cellular factor HEXIM1. Previously, the chimeric protein HEXIM1-Tat (HT) was demonstrated to inhibit human immunodeficiency virus-1 (HIV)-1 transcription. In this study, we attempted to develop an improved antiviral protein that specifically binds viral RNA (vRNA) by fusing HT to HIV-1 nucleocapsid (NC). Thus, we synthesized NC-HEXIM1-Tat (NHT) and HEXIM1-Tat-NC (HTN). NHT and HTN inhibited virus proliferation more effectively than HT, and they did not attenuate the function of HT. Notably, NHT and HTN inhibited the infectivity of the progeny virus, whereas HT had no such effect. NHT and HTN selectively and effectively interacted with vRNA and inhibited the proper packaging of the HIV-1 genome. Taken together, our results illustrated that the novel NC-fused chimeric proteins NHT and HTN display novel mechanisms of anti-HIV effects by inhibiting both HIV-1 transcription and packaging.


Subject(s)
HIV-1 , Positive Transcriptional Elongation Factor B , Humans , Positive Transcriptional Elongation Factor B/metabolism , HIV-1/genetics , HIV-1/metabolism , tat Gene Products, Human Immunodeficiency Virus/metabolism , RNA, Viral/metabolism , Nucleocapsid Proteins/genetics , Nucleocapsid Proteins/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Virus Replication , Nucleocapsid/metabolism , Antiviral Agents/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/metabolism , Transcription Factors/metabolism
5.
Clin Transl Sci ; 15(5): 1123-1130, 2022 05.
Article in English | MEDLINE | ID: mdl-35133064

ABSTRACT

For the long-term efficacy of dry eye disease treatment, relieving underlying inflammation is necessary. Imatinib mesylate is a novel ophthalmic formulation of imatinib mesylate, which is expected to alleviate inflammation by inhibiting the discoidin domain receptor 1 activity. This study aims to evaluate the safety and pharmacokinetics of imatinib mesylate in healthy subjects. A randomized, double-blind, placebo-controlled study was conducted. In a single ascending dose, 16 subjects received a single eye drop of imatinib mesylate 0.1%, 0.3%, or matching placebo. In the multiple ascending dose (MAD), subjects received multiple eye drops of imatinib mesylate 0.1%, 0.3%, or matching placebo once daily for 7 days. Safety and tolerability were assessed by ophthalmic examination, including the visual analog scale (VAS) to monitor the burning sensation in the eyes. A total of four treatment-emergent adverse events (TEAEs) occurred during the study. All TEAEs were mildly severe with no serious cases. VAS results in the 0.1% MAD group exhibited highest score of two points, whereas it was less than one point in others. Insignificant difference between the imatinib mesylate and placebo groups in the VAS results was seen. After a single dose administration of imatinib mesylate 0.1%, all plasma concentrations were below the lower limit of quantification. The peak plasma concentrations of imatinib were less than 0.54 µg/L in all groups. In conclusion, a single and multiple topical ophthalmic administration of imatinib mesylate was well-tolerated in healthy subjects. Because there was minimal systemic exposure to imatinib, the adverse effect in the body seems to be insignificant.


Subject(s)
Inflammation , Administration, Ophthalmic , Dose-Response Relationship, Drug , Double-Blind Method , Healthy Volunteers , Humans , Imatinib Mesylate/adverse effects , Ophthalmic Solutions/adverse effects
6.
Transl Vis Sci Technol ; 10(11): 14, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34520512

ABSTRACT

Purpose: Dry eye disease (DED) is a multifactorial disorder of the tears and ocular surface accompanied by ocular discomfort, visual disturbance, tear film instability, and ocular surface inflammation. In the present study, we evaluated the efficacy of the tyrosine kinase inhibitor imatinib mesylate for the treatment of DED. Methods: Experimental models of DED were generated in Sprague Dawley rats using a combination of benzalkonium chloride (BAC) with atropine sulfate and in New Zealand White rabbits using BAC. The animals were treated twice daily with eye drops of vehicle, imatinib (0.01%-0.3%), or a positive control (Restasis). The improvement in DED due to imatinib was assessed by staining with fluorescein, lissamine green, impression cytology, and histological analysis. In addition, immunofluorescence staining was performed at the end of the study to evaluate the inflammatory response in the ocular surface. Results: Topical application of imatinib significantly reduced ocular surface damage compared with vehicle-treated animals. Imatinib restored the morphology and structure of the conjunctival epithelium and reduced the recruitment of immune cells in the corneal epithelium. Furthermore, imatinib significantly reduced the impression cytology score, thus demonstrating that imatinib prevents the loss of goblet cells in DED animal models. The therapeutic efficacy of imatinib was similar to or better than that of cyclosporine treatment. Conclusions: In this study, we provide an animal in vivo proof of concept of the therapeutic potential of imatinib for the treatment of DED. Translational Relevance: With this study we show the possibility of developing imatinib as a new ophthalmic drop to treat DED.


Subject(s)
Dry Eye Syndromes , Epithelium, Corneal , Animals , Dry Eye Syndromes/chemically induced , Imatinib Mesylate , Models, Animal , Protein Kinase Inhibitors/therapeutic use , Rabbits , Rats , Rats, Sprague-Dawley
7.
BMB Rep ; 54(11): 551-556, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34353428

ABSTRACT

In this study, we investigated how Staufen1 influences the HIV-1 production. The overexpression of Staufen1 increased virus production without any negative affect on the viral infectivity. This increase was not caused by transcriptional activation; but by influencing post-transcriptional steps. Using multiple Gag protein derivatives, we confirmed that the zinc-finger domains of the HIV-1 nucleocapsid (NC) are important for its interaction with Staufen1. We also found that Staufen1 colocalized in stress granules with the mature form of the HIV-1 NC protein. [BMB Reports 2021; 54(11): 551-556].


Subject(s)
Cytoskeletal Proteins/metabolism , Gene Products, gag/metabolism , HIV Infections/virology , HIV-1/physiology , Nucleocapsid/metabolism , RNA-Binding Proteins/metabolism , Stress Granules/metabolism , Virus Replication , Cytoskeletal Proteins/genetics , Gene Products, gag/genetics , HeLa Cells , Humans , Nucleocapsid/genetics , Protein Binding , Protein Interaction Domains and Motifs , RNA-Binding Proteins/genetics
8.
Int J Mol Sci ; 22(8)2021 Apr 09.
Article in English | MEDLINE | ID: mdl-33918777

ABSTRACT

It has been shown previously that a novel tetrapeptide, Arg-Leu-Tyr-Glu (RLYE), derived from human plasminogen inhibits vascular endothelial growth factor (VEGF)-induced angiogenesis, suppresses choroidal neovascularization in mice by an inhibition of VEGF receptor-2 (VEGFR-2) specific signaling pathway. In this study, we report that a modified tetrapeptide (Ac-RLYE) showed improved anti-choroidal neovascularization (CNV) efficacy in a number of animal models of neovascular age-related macular degeneration (AMD) which include rat, rabbit, and minipig. The preventive and therapeutic in vivo efficacy of Ac-RLYE via following intravitreal administration was determined to be either similar or superior to that of ranibizumab and aflibercept. Assessment of the intraocular pharmacokinetic and toxicokinetic properties of Ac-RLYE in rabbits demonstrated that it rapidly reached the retina with minimal systemic exposure after a single intravitreal dose, and it did not accumulate in plasma during repetitive dosing (bi-weekly for 14 weeks). Our results suggested that Ac-RLYE has a great potential for an alternative therapeutics for neovascular (wet) AMD. Since the amino acids in human VEGFR-2 targeted by Ac-RLYE are conserved among the animals employed in this study, the therapeutic efficacies of Ac-RLYE evaluated in those animals are predicted to be observed in human patients suffering from retinal degenerative diseases.


Subject(s)
Macular Degeneration/etiology , Macular Degeneration/metabolism , Oligopeptides/pharmacology , Acetylation , Animals , Choroidal Neovascularization/drug therapy , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Disease Models, Animal , Disease Susceptibility , Fluorescein Angiography , Humans , Macular Degeneration/diagnosis , Macular Degeneration/drug therapy , Male , Mice , Oligopeptides/chemistry , Promoter Regions, Genetic , Rabbits , Ranibizumab/pharmacology , Receptors, Vascular Endothelial Growth Factor , Recombinant Fusion Proteins/pharmacology , Retina/metabolism , Retina/pathology , Retinal Neovascularization/drug therapy , Retinal Neovascularization/metabolism , Retinal Neovascularization/pathology , Swine , Treatment Outcome , Vascular Endothelial Growth Factor A/chemistry , Vascular Endothelial Growth Factor A/metabolism
9.
Biomed Pharmacother ; 134: 111110, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33338749

ABSTRACT

Targeting the vascular endothelial growth factor (VEGF)/its receptor-2 (VEGFR-2) system has become a mainstay of treatment for many human diseases, including retinal diseases. We examined the therapeutic effect of recently developed N-acetylated Arg-Leu-Tyr-Glu (Ac-RLYE), a human plasminogen kringle-5 domain-derived VEGFR-2 antagonists, on the pathogenesis of diabetic retinopathy. Ac-RLYE inhibited VEGF-A-mediated VEGFR-2 activation and endothelial nitric oxide synthase (eNOS)-derived NO production in the retinas of diabetic mice. In addition, Ac-RLYE prevented the disruption of adherens and tight junctions and vascular leakage by inhibiting S-nitrosylation of ß-catenin and tyrosine nitration of p190RhoGAP in the retinal vasculature of diabetic mice. Peptide treatment preserved the pericyte coverage of retinal capillaries by upregulating angiopoietin-2. These results suggest that Ac-RLYE potentially prevents blood-retinal barrier breakdown and vascular leakage by antagonizing VEGFR-2; Ac-RLYE can be used as a potential therapeutic drug for the treatment of diabetic retinopathy.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Blood-Retinal Barrier/drug effects , Diabetic Retinopathy/drug therapy , Oligopeptides/pharmacology , Retinal Vessels/drug effects , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Adherens Junctions/drug effects , Adherens Junctions/metabolism , Adherens Junctions/pathology , Animals , Blood-Retinal Barrier/metabolism , Blood-Retinal Barrier/pathology , Capillary Permeability/drug effects , Cells, Cultured , Diabetes Mellitus, Experimental/complications , Diabetic Retinopathy/etiology , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Humans , Male , Mice, Inbred C57BL , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Retinal Vessels/metabolism , Retinal Vessels/pathology , Signal Transduction , Tight Junctions/drug effects , Tight Junctions/metabolism , Tight Junctions/pathology , Vascular Endothelial Growth Factor Receptor-2/metabolism
10.
BMB Rep ; 53(5): 248-253, 2020 May.
Article in English | MEDLINE | ID: mdl-31818358

ABSTRACT

Gene expression in HIV-1 is regulated by the promoters in 5' long-terminal repeat (LTR) element, which contain multiple DNA regulatory elements that serve as binding sites for cellular transcription factors. YY1 could repress HIV-1 gene expression and latent infection. Here, however, we observed that virus production can be increased by YY1 over-expression and decreased under YY1 depleted condition by siRNA treatment. To identify functional domain(s) of YY1 activation, we constructed a number of YY1 truncated mutants. Our data show that full-length YY1 enhances the viral transcription both through U3 and U3RU5 promoters. Moreover, the C-terminal region (296-414 residues) of YY1 is responsible for the transcriptional upregulation, which could be enhanced further in the presence of the viral Tat protein. The central domain of YY1 (155-295 residues) does not affect LTR activity but has a negative effect on HIV-1 gene expression. Taken together, our study shows that YY1 could act as a transcriptional activator in HIV-1 replication, at least in the early stages of infection. [BMB Reports 2020; 53(5): 248-253].


Subject(s)
Gene Expression Regulation, Viral/genetics , HIV-1/genetics , Up-Regulation , YY1 Transcription Factor/metabolism , HIV-1/metabolism , Humans , Virus Replication/genetics
11.
BMB Rep ; 51(8): 388-393, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29636121

ABSTRACT

The activating transcription factor (ATF) 4 belongs to the ATF/CREB (cAMP Response Element Binding bZIP [Basic Leucine Zipper]) transcription factor family, and plays a central role in the UPR (Unfolded Protein Response) process in cells. The induction of ATF4 expression has previously been shown to increase the replication of HIV-1. However, the detailed mechanism underlying this effect and the factors involved in the regulation of ATF4 function are still unknown. Here, we demonstrate first that knocking out ATF4 using siRNA shows a strong negative effect on HIV-1 production, indicating that ATF4 is a functional positive cellular factor in HIV-1 production. To determine the mechanism by which ATF4 regulates the HIV-1 life cycle, we assessed the effect of the overexpression of wild type ATF4 and its various derivatives on HIV-1 LTR-mediated transcriptional activation and the production of HIV-1 particles. This effect was studied through co-transfection experiments with either reporter vectors or proviral DNA. We found that the N-terminal domains of ATF4 are involved in HIV-1 LTR-mediated transcriptional activation, and thus in HIV-1 production. [BMB Reports 2018; 51(8): 388-393].


Subject(s)
Activating Transcription Factor 4/physiology , HIV-1/physiology , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Gene Expression Regulation, Viral , HEK293 Cells , HIV Infections/genetics , HIV Infections/metabolism , HIV Infections/virology , HIV Long Terminal Repeat , HIV-1/genetics , HIV-1/metabolism , Humans , Transcription, Genetic , Transcriptional Activation , Unfolded Protein Response
12.
BMB Rep ; 51(7): 338-343, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29555014

ABSTRACT

Transcription termination factor-1 (TTF-I) is an RNA polymerase 1-mediated transcription terminator and consisting of a C-terminal DNA-binding domain, central domain, and N-terminal regulatory domain. This protein binds to a so-called 'Sal box' composed of an 11-base pair motif. The interaction of TTF-I with the 'Sal box' is important for many cellular events, including efficient termination of RNA polymerase-1 activity involved in pre-rRNA synthesis and formation of a chromatin loop. To further understand the role of TTF-I in human immunodeficiency virus (HIV)-I virus production, we generated various TTF-I mutant forms. Through a series of studies of the over-expression of TTF-I and its derivatives along with co-transfection with either proviral DNA or HIV-I long terminal repeat (LTR)-driven reporter vectors, we determined that wild-type TTF-I downregulates HIV-I LTR activity and virus production, while the TTF-I Myb-like domain alone upregulated virus production, suggesting that wild-type TTF-I inhibits virus production and trans-activation of the LTR sequence; the Myb-like domain of TTF-I increased virus production and trans-activated LTR activity. [BMB Reports 2018; 51(7): 338-343].


Subject(s)
DNA-Binding Proteins/metabolism , HIV-1/physiology , Transcription Factors/metabolism , DNA-Binding Proteins/genetics , Genetic Vectors/genetics , Genetic Vectors/metabolism , HEK293 Cells , HIV Long Terminal Repeat/genetics , HIV-1/genetics , HeLa Cells , Humans , Mutagenesis , Promoter Regions, Genetic , RNA Polymerase I/metabolism , RNA, Viral/metabolism , Transcription Factors/genetics , Transcriptional Activation , Virus Replication
13.
BMB Rep ; 51(6): 290-295, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29429449

ABSTRACT

Y-box binding protein 1 (YB-1) is a member of the cold-shock domain (CSD) protein superfamily. It participates in a wide variety of cellular events, including transcription, RNA splicing, translation, DNA repair, drug resistance, and stress responses. We investigated putative functions of YB-1 in HIV-1 replication. Functional studies using overexpression or knockdown of YB-1 in conjunction with transfection of proviral DNA showed that YB-1 enhances virus production. We found YB-1 regulates HIV-1 production by stimulating viral transcription using HIV-1 LTR sequence U3RU5 with Luciferase assay. We also identified a specific region from amino acids 1 to 324 of YB-1 as necessary for the participation of the protein in the production of virions. [BMB Reports 2018; 51(6): 290-295].


Subject(s)
HIV Infections/metabolism , Y-Box-Binding Protein 1/metabolism , Y-Box-Binding Protein 1/physiology , DNA/metabolism , DNA-Binding Proteins/metabolism , HIV/metabolism , HIV Long Terminal Repeat/genetics , Human Immunodeficiency Virus Proteins/metabolism , Humans , Transcriptional Activation , Transfection , Y-Box-Binding Protein 1/genetics
14.
RNA ; 24(2): 219-236, 2018 02.
Article in English | MEDLINE | ID: mdl-29127210

ABSTRACT

The nucleocapsid (NC) is an N-terminal protein derived from the HIV-1 Gag precursor polyprotein, pr55Gag NC possesses key functions at several pivotal stages of viral replication. For example, an interaction between NC and the host double-stranded RNA-binding protein Staufen1 was shown to regulate several steps in the viral replication cycle, such as Gag multimerization and genomic RNA encapsidation. In this work, we observed that the overexpression of NC leads to the induction of stress granule (SG) assembly. NC-mediated SG assembly was unique as it was resistant to the SG blockade imposed by the HIV-1 capsid (CA), as shown in earlier work. NC also reduced host cell mRNA translation, as judged by a puromycylation assay of de novo synthesized proteins, and this was recapitulated in polysome profile analyses. Virus production was also found to be significantly reduced. Finally, Staufen1 expression completely rescued the blockade to NC-mediated SG assembly, global mRNA translation as well as virus production. NC expression also resulted in the phosphorylation of protein kinase R (PKR) and eIF2α, and this was inhibited with Staufen1 coexpression. This work sheds light on an unexpected function of NC in host cell translation. A comprehensive understanding of the molecular mechanisms by which a fine balance of the HIV-1 structural proteins NC and CA act in concert with host proteins such as Staufen1 to modulate the host stress response will aid in the development of new antiviral therapeutics.


Subject(s)
Protein Biosynthesis , RNA-Binding Proteins/metabolism , gag Gene Products, Human Immunodeficiency Virus/metabolism , Cell Line , Cytoplasmic Granules/metabolism , DNA Helicases/metabolism , Eukaryotic Initiation Factor-2/metabolism , Eukaryotic Initiation Factor-3/metabolism , HIV-1/physiology , HeLa Cells , Humans , Poly-ADP-Ribose Binding Proteins/metabolism , RNA Helicases/metabolism , RNA Recognition Motif Proteins/metabolism , RNA, Messenger/metabolism , eIF-2 Kinase/metabolism , gag Gene Products, Human Immunodeficiency Virus/antagonists & inhibitors
15.
Oncotarget ; 8(7): 11763-11777, 2017 Feb 14.
Article in English | MEDLINE | ID: mdl-28052029

ABSTRACT

The tetrapeptide Arg-Leu-Tyr-Glu (RLYE) is known to inhibit vascular endothelial growth factor-A (VEGF-A)-induced angiogenesis in vitro. Herein, we examined its underlying mechanism and antitumor activity associated with vascular remodeling. RLYE inhibited VEGF-A-induced angiogenesis in a mouse model and suppressed VEGF-A-induced angiogenic signal cascades in human endothelial cells. However, RLYE showed no inhibitory effect on VEGF-A-induced proliferation and migration of multiple myeloma cells expressing VEGF receptor (VEGFR)-1, but not VEGFR-2. In addition, RLYE showed no inhibitory effect on angiogenic activities induced by VEGF-B, basic fibroblast growth factor, epithermal growth factor, sphingosine-1-phosphate, and placental growth factor. RLYE bound specifically to VEGFR-2 at the VEGF-A binding site, thereby blocking VEGF-A-VEGFR-2 binding and VEGF-A-induced VEGFR-2 internalization. The RLYE peptide inhibited tumor growth and metastasis via suppression of tumor angiogenesis in tumor-bearing mice. Moreover, RLYE showed a synergistic effect of the cytotoxic agent irinotecan on tumor cell apoptosis and tumor progression via tumor vessel normalization due to stabilization of VE-cadherin-mediated adherens junction, improvement of pericyte coverage, and inhibition of vascular leakage in tumors. Our results suggest that RLYE can be used as an antiangiogenic and tumor blood vessel remodeling agent for inhibition of tumor growth and metastasis by antagonizing VEGFR-2, with the synergistic anti-cancer effect via enhancement of drug delivery and therapeutic efficacy.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Colonic Neoplasms/drug therapy , Melanoma, Experimental/drug therapy , Oligopeptides/pharmacology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Animals , Capillary Permeability/drug effects , Colonic Neoplasms/blood supply , Colonic Neoplasms/metabolism , Disease Progression , HCT116 Cells , Humans , Male , Melanoma, Experimental/blood supply , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Mice, Nude , Neovascularization, Pathologic/drug therapy , Rats, Sprague-Dawley , Vascular Endothelial Growth Factor Receptor-2/metabolism
16.
Virology ; 492: 204-12, 2016 May.
Article in English | MEDLINE | ID: mdl-26967976

ABSTRACT

The HIV-1 nucleocapsid (NC) is an essential viral protein containing two highly conserved retroviral-type zinc finger (ZF) motifs, which functions in multiple stages of the HIV-1 life cycle. Although a number of functions for NC either in its mature form or as a domain of Gag have been revealed, little is known about the intracellular localization of NC and, moreover, its role in Gag protein trafficking. Here, we have investigated various forms of HIV-1 NC protein for its cellular localization and found that the NC has a strong nuclear and nucleolar localization activity. The linker region, composed of a stretch of basic amino acids between the two ZF motifs, was necessary and sufficient for the activity.


Subject(s)
Cell Nucleolus/virology , HIV-1/genetics , Nucleocapsid Proteins/genetics , Nucleocapsid/genetics , Virion/genetics , Amino Acid Sequence , Calnexin/genetics , Calnexin/metabolism , Cell Nucleolus/metabolism , Cell Nucleolus/ultrastructure , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Gene Expression , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , HIV-1/metabolism , HIV-1/ultrastructure , HeLa Cells , Host-Pathogen Interactions , Humans , Molecular Sequence Data , Nucleocapsid/metabolism , Nucleocapsid Proteins/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Virion/metabolism , Virion/ultrastructure , Virus Assembly/genetics , Zinc/chemistry , Zinc/metabolism , Zinc Fingers , gag Gene Products, Human Immunodeficiency Virus/genetics , gag Gene Products, Human Immunodeficiency Virus/metabolism
17.
BMB Rep ; 49(1): 51-6, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26246283

ABSTRACT

Glycogen synthase kinase-3ß (GSK-3ß) is a serine/threonine protein kinase that is known to mediate cancer cell death. Here, we show that B-cell lymphoma 2 (Bcl-2), an anti-apoptotic protein, is regulated by GSK-3ß and that GSK-3ß-mediated regulation of Bcl-2 is crucial for mitochondrial-dependent cell death in paclitaxel-stimulated cells. We demonstrate that MCF7 GSK-3ß siRNA cells are more sensitive to cell death than MCF7 GFP control cells and that in the absence of GSK-3ß, Bcl-2 levels are reduced, a result enhanced by paclitaxel. Paclitaxel-induced JNK (c-Jun N-terminal kinase) activation is critical for Bcl-2 modulation. In the absence of GSK-3ß, Bcl-2 was unstable in an ubiquitination-dependent manner in both basal- and paclitaxeltreated cells. Furthermore, we demonstrate that GSK-3ß-mediated regulation of Bcl-2 influences cytochrome C release and mitochondrial membrane potential. Taken together, our data suggest that GSK-3ß-dependent regulation of Bcl-2 is crucial for mitochondria-dependent cell death in paclitaxel-mediated breast cancer therapy.


Subject(s)
Apoptosis/drug effects , Glycogen Synthase Kinase 3/metabolism , Paclitaxel/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Cytochromes c/metabolism , DNA Fragmentation/drug effects , Enzyme Activation/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , MCF-7 Cells , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Protein Stability/drug effects , RNA Interference , RNA, Small Interfering/metabolism , Ubiquitination
18.
Retrovirology ; 12: 90, 2015 Nov 06.
Article in English | MEDLINE | ID: mdl-26545586

ABSTRACT

BACKGROUND: The human immunodeficiency virus type-1 (HIV-1) nucleocapsid protein (NC) is an essential and multifunctional protein involved in multiple stages of the viral life cycle such as reverse transcription, integration of proviral DNA, and especially genome RNA packaging. For this reason, it has been considered as an attractive target for the development of new anti-HIV drugs. Although a number of inhibitors of NC have been reported thus far, the search for NC-specific and functional inhibitor(s) with a good antiviral activity continues. RESULTS: In this study, we report the identification of A1752, a small molecule with inhibitory action against HIV-1 NC, which shows a strong antiviral efficacy and an IC50 around 1 µM. A1752 binds directly to HIV-1 NC, thereby inhibiting specific chaperone functions of NC including Psi RNA dimerization and complementary trans-activation response element (cTAR) DNA destabilization, and it also disrupts the proper Gag processing. Further analysis of the mechanisms of action of A1752 also showed that it generates noninfectious viral particles with defects in uncoating and reverse transcription in the infected cells. CONCLUSIONS: These results demonstrate that A1752 is a specific and functional inhibitor of NC with a novel mode of action and good antiviral efficacy. Thus, this agent provides a new type of anti-HIV NC inhibitor candidate for further drug development.


Subject(s)
Anti-HIV Agents/pharmacology , HIV-1/drug effects , Nucleocapsid Proteins/antagonists & inhibitors , Propionates/pharmacology , Thiazolidines/pharmacology , gag Gene Products, Human Immunodeficiency Virus/antagonists & inhibitors , Amino Acid Sequence , Anti-HIV Agents/chemistry , Anti-HIV Agents/metabolism , Dimerization , Drug Discovery , HIV-1/physiology , Humans , Molecular Chaperones/metabolism , Nucleocapsid Proteins/metabolism , Propionates/chemistry , Propionates/metabolism , RNA, Viral/chemistry , RNA, Viral/genetics , Thiazolidines/chemistry , Thiazolidines/metabolism , gag Gene Products, Human Immunodeficiency Virus/metabolism
19.
Biochem Biophys Res Commun ; 457(2): 227-33, 2015 Feb 06.
Article in English | MEDLINE | ID: mdl-25559349

ABSTRACT

Although cis-acting packaging signal RNA sequences for the influenza virus NP encoding vRNA have been identified recently though genetic studies, little is known about the interaction between NP and the vRNA packaging signals either in vivo or in vitro. Here, we provide evidence that NP is able to interact specifically with the vRNA packaging sequence RNA within living cells and that the specific RNA binding activity of NP in vivo requires both the N-terminal and central region of the protein. This assay established would be a valuable tool for further detailed studies of the NP-packaging signal RNA interaction in living cells.


Subject(s)
Biological Assay/methods , Influenza A virus/metabolism , Nucleoproteins/metabolism , Protein Sorting Signals , RNA, Viral/metabolism , Virus Assembly , Gene Deletion , Kinetics , Lac Operon , Protein Binding
20.
BMB Rep ; 48(3): 178-83, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25560695

ABSTRACT

Dendritic cells play an important role in determining whether naïve T cells mature into either Th1 or Th2 cells. We determined whether heat-shock protein X (HspX) purified from Mycobacterium tuberculosis regulates the Th1/Th2 immune response in an ovalbumin (OVA)-induced murine model of asthma. HspX increased interferon-gamma, IL-17A, -12 and transforming growth factor (TGF)-ß production and T-bet gene expression but reduced IL-13 production and GATA-3 gene expression. HspX also inhibited asthmatic reactions as demonstrated by an increase in the number of eosinophils in bronchoalveolar lavage fluid, inflammatory cell infiltration in lung tissues, airway luminal narrowing, and airway hyper-responsiveness. Furthermore, HspX enhanced OVA-induced decrease of regulatory T cells in the mediastinal lymph nodes. This study provides evidence that HspX plays critical roles in the amelioration of asthmatic inflammation in mice. These findings provide new insights into the immunotherapeutic role of HspX with respect to its effects on a murine model of asthma.


Subject(s)
Asthma/therapy , Dendritic Cells/immunology , Heat-Shock Proteins/isolation & purification , Hypersensitivity/therapy , Immunotherapy , Mycobacterium tuberculosis/metabolism , Adoptive Transfer , Animals , GATA3 Transcription Factor/metabolism , Inflammation Mediators/metabolism , Lung/metabolism , Mice , Ovalbumin/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...