Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Biochem Biophys Res Commun ; 444(2): 135-40, 2014 Feb 07.
Article in English | MEDLINE | ID: mdl-24434142

ABSTRACT

Sangivamycin, a structural analog of adenosine and antibiotic exhibiting antitumor and antivirus activities, inhibits protein kinase C and the synthesis of both DNA and RNA. Primary effusion lymphoma (PEL) is an aggressive neoplasm caused by Kaposi's sarcoma-associated herpesvirus (KSHV) in immunosuppressed patients and HIV-infected homosexual males. PEL cells are derived from post-germinal center B cells, and are infected with KSHV. Herein, we asked if sangivamycin might be useful to treat PEL. We found that sangivamycin killed PEL cells, and we explored the underlying mechanism. Sangivamycin treatment drastically decreased the viability of PEL cell lines compared to KSHV-uninfected B lymphoma cell lines. Sangivamycin induced the apoptosis of PEL cells by activating caspase-7 and -9. Further, sangivamycin suppressed the phosphorylation of Erk1/2 and Akt, thus inhibiting activation of the proteins. Inhibitors of Akt and MEK suppressed the proliferation of PEL cells compared to KSHV-uninfected cells. It is known that activation of Erk and Akt signaling inhibits apoptosis and promotes proliferation in PEL cells. Our data therefore suggest that sangivamycin induces apoptosis by inhibiting Erk and Akt signaling in such cells. We next investigated whether sangivamycin, in combination with an HSP90 inhibitor geldanamycin (GA) or valproate (valproic acid), potentiated the cytotoxic effects of the latter drugs on PEL cells. Compared to treatment with GA or valproate alone, the addition of sangivamycin enhanced cytotoxic activity. Our data thus indicate that sangivamycin may find clinical utility as a novel anti-cancer agent targeting PEL.


Subject(s)
Apoptosis/drug effects , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Pyrimidine Nucleosides/pharmacology , Blotting, Western , Caspase 7/metabolism , Caspase 9/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Gene Expression Regulation, Viral , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/physiology , Host-Pathogen Interactions , Humans , Lymphoma, Primary Effusion/metabolism , Lymphoma, Primary Effusion/pathology , Lymphoma, Primary Effusion/virology , Male , Phosphorylation/drug effects , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Reverse Transcriptase Polymerase Chain Reaction
2.
J Virol ; 86(22): 12198-207, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22933294

ABSTRACT

A novel anti-varicella-zoster virus compound, a derivative of pyrazolo[1,5-c]1,3,5-triazin-4-one (coded as 35B2), was identified from a library of 9,600 random compounds. This compound inhibited both acyclovir (ACV)-resistant and -sensitive strains. In a plaque reduction assay under conditions in which the 50% effective concentration of ACV against the vaccine Oka strain (V-Oka) in human fibroblasts was 4.25 µM, the 50% effective concentration of 35B2 was 0.75 µM. The selective index of the compound was more than 200. Treatment with 35B2 inhibited neither immediate-early gene expression nor viral DNA synthesis. Twenty-four virus clones resistant to 35B2 were isolated, all of which had a mutation(s) in the amino acid sequence of open reading frame 40 (ORF40), which encodes the major capsid protein (MCP). Most of the mutations were located in the regions corresponding to the "floor" domain of the MCP of herpes simplex virus 1. Treatment with 35B2 changed the localization of MCP in the fibroblasts infected with V-Oka but not in the fibroblasts infected with the resistant clones, although it did not affect steady-state levels of MCP. Overexpression of the scaffold proteins restored the normal MCP localization in the 35B2-treated infected cells. The compound did not inhibit the scaffold protein-mediated translocation of MCP from the cytoplasm to the nucleus. Electron microscopic analysis demonstrated the lack of capsid formation in the 35B2-treated infected cells. These data indicate the feasibility of developing a new class of antivirals that target the herpesvirus MCPs and inhibit normal capsid formation by a mechanism that differs from those of the known protease and encapsidation inhibitors. Further biochemical studies are required to clarify the precise antiviral mechanism.


Subject(s)
Antiviral Agents/pharmacology , Capsid/metabolism , Herpesvirus 3, Human/genetics , Virus Replication , Acyclovir/pharmacology , Amino Acid Sequence , Antiviral Agents/chemistry , Capsid Proteins/chemistry , Cell Line , Cell Line, Tumor , DNA, Viral/metabolism , Drug Evaluation, Preclinical/methods , Drug Resistance, Viral , Genes, Reporter , HEK293 Cells , Herpesvirus 1, Human/metabolism , Humans , Microscopy, Electron/methods , Molecular Sequence Data , Protein Structure, Tertiary , Protein Transport , Sequence Analysis, DNA , Sequence Homology, Amino Acid
3.
Biol Pharm Bull ; 35(5): 725-30, 2012.
Article in English | MEDLINE | ID: mdl-22687408

ABSTRACT

Primary effusion lymphoma (PEL) is an aggressive neoplasm caused by Kaposi's sarcoma-associated herpesvirus (KSHV) in immunosuppressed patients and human immunodeficiency virus (HIV)-infected homosexual males. We evaluated the cytotoxic effects of heat shock protein 90 (HSP90) inhibitors on PEL cells. The HSP90 inhibitors geldanamycin (GA), 17(allylamino)-17-demethoxygeldanamycin (17-AAG), and radicicol dramatically inhibited cell proliferation and induced apoptosis of PEL cells through caspase activation. Furthermore, GA induced the stabilization of inhibitor of κB (IκB)α and reduced the phosphorylation of IκBα in PEL cells. HSP90 inhibitors suppressed the transcriptional activity of nuclear factor-kappa B (NF-κB) in PEL cells. It is known that the constitutive activation of NF-κB signaling is essential for the survival of PEL cells and HSP90 contributes to promote activation of NF-κB signaling. The suppression of NF-κB signaling by HSP90 inhibitors may contribute to the induction of apoptosis in PEL cells. In addition, HSP90 activity is required for KSHV replication in KSHV latently infected PEL cells. GA, 17-AAG and radicicol reduced the production of progeny virus from PEL cells at low concentrations, which do not affect PEL cell growth. Our results suggest that HSP90 activity is required for both the survival of PEL cells and viral replication in PEL cells, and that pharmacologic inhibition of HSP90 may be an effective treatment for PEL and KSHV-related diseases.


Subject(s)
Apoptosis/drug effects , Benzoquinones/therapeutic use , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Herpesvirus 8, Human/drug effects , Lactams, Macrocyclic/therapeutic use , Lymphoma, Primary Effusion/drug therapy , Macrolides/therapeutic use , Virus Replication/drug effects , Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/therapeutic use , Benzoquinones/pharmacology , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic , Herpesvirus 8, Human/physiology , Humans , I-kappa B Proteins/metabolism , Lactams, Macrocyclic/pharmacology , Lymphoma, Primary Effusion/metabolism , Lymphoma, Primary Effusion/virology , Macrolides/pharmacology , NF-kappa B/metabolism , Phosphorylation , Sarcoma, Kaposi/complications , Sarcoma, Kaposi/virology , Signal Transduction , Transcription, Genetic/drug effects
4.
Front Microbiol ; 3: 66, 2012.
Article in English | MEDLINE | ID: mdl-22375140

ABSTRACT

Ubiquitination is a post-translational modification in which one or more ubiquitin molecules are covalently linked to lysine residues of target proteins. The ubiquitin system plays a key role in the regulation of protein degradation, which contributes to cell signaling, vesicular trafficking, apoptosis, and immune regulation. Bacterial and viral pathogens exploit the cellular ubiquitin system by encoding their own proteins to serve their survival and replication in infected cells. Recent studies have revealed that Kaposi's sarcoma-associated herpesvirus (KSHV) manipulates the ubiquitin system of infected cells to facilitate cell proliferation, anti-apoptosis, and evasion from immunity. This review summarizes recent developments in our understanding of the molecular mechanisms used by KSHV to interact with the cellular ubiquitin machinery.

5.
Biochem Biophys Res Commun ; 415(4): 573-8, 2011 Dec 02.
Article in English | MEDLINE | ID: mdl-22074820

ABSTRACT

Primary effusion lymphoma (PEL) is an aggressive neoplasm caused by Kaposi's sarcoma-associated herpesvirus (KSHV). This study provides evidence that proteasomal activity is required for both survival of PEL cells stably harboring the KSHV genome and viral replication of KSHV. We evaluated the cytotoxic effects of proteasome inhibitors on PEL cells. The proteasome inhibitors MG132, lactacystin, and proteasome inhibitor I dramatically inhibited cell proliferation and induced apoptosis of PEL cells through the accumulation of p21 and p27. Furthermore, proteasome inhibitors induced the stabilization of NF-κB inhibitory molecule (IκBα) and suppressed the transcriptional activity of NF-κB in PEL cells. The NF-κB specific inhibitor BAY11-7082 also induced apoptosis in PEL cells. The constitutive activation of NF-κB signaling is essential for the survival and growth of B cell lymphoma cells, including PEL cells. NF-κB signaling is upregulated by proteasome-dependent degradation of IκBα. The suppression of NF-κB signaling by proteasome inhibitors may contribute to the induction of apoptosis in PEL cells. In addition, proteasome activity is required for KSHV replication in KSHV latently infected PEL cells. MG132 reduced the production of progeny virus from PEL cells at low concentrations, which do not affect PEL cell growth. These findings suggest that proteasome inhibitors may represent a novel strategy for the treatment of KSHV infection and KSHV-associated lymphomas.


Subject(s)
Apoptosis/drug effects , Cysteine Proteinase Inhibitors/pharmacology , Herpesvirus 8, Human/drug effects , Lymphoma, Primary Effusion/virology , Proteasome Inhibitors , Virus Replication/drug effects , Acetylcysteine/analogs & derivatives , Acetylcysteine/pharmacology , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Herpesvirus 8, Human/physiology , Humans , Leupeptins/pharmacology , NF-kappa B/antagonists & inhibitors , Nitriles/pharmacology , Oligopeptides/pharmacology , Sulfones/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...