Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Publication year range
1.
Rinsho Ketsueki ; 62(12): 1661-1665, 2021.
Article in Japanese | MEDLINE | ID: mdl-35022333

ABSTRACT

A 46-year-old woman was diagnosed with acute promyelocytic leukemia (APL). The patient was given remission induction therapy with all-trans retinoic acid, and complete remission was achieved. Despite consolidation therapies with arsenic trioxide, daunorubicin and cytosine arabinoside (AraC), and gemtuzumab ozogamicin as well as maintenance therapy with tamibarotene, the patient experienced a relapse 6 months after the start of maintenance therapy. She was then given re-induction therapy with idarubicin+AraC and high-dose AraC, but remission was not achieved. Since the coordination of the unrelated donor had been completed at this time, she then underwent bone marrow transplantation with pre-conditioning of 4 Gy total body irradiation, fludarabine, and busulfan. However, on the 12th day after the transplantation, APL cells appeared in the peripheral blood and the disease progressed rapidly leading to the patient's death on the 15th day after the transplantation. APL usually has a good prognosis, and relapsed cases are often cured by autologous stem cell transplantation. However, this case was highly refractory to treatment and the patient deteriorated rapidly after the transplantation, suggesting a different pathogenesis from the usual from of APL.


Subject(s)
Antineoplastic Agents , Arsenicals , Hematopoietic Stem Cell Transplantation , Leukemia, Promyelocytic, Acute , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Arsenicals/therapeutic use , Female , Humans , Karyotype , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/genetics , Middle Aged , Oxides/therapeutic use , Transplantation, Autologous , Treatment Outcome , Tretinoin/therapeutic use
2.
Sci Adv ; 1(3): e1400203, 2015 Apr.
Article in English | MEDLINE | ID: mdl-26601161

ABSTRACT

Adult T cell leukemia (ATL) is an aggressive T cell malignancy caused by human T cell leukemia virus type 1 (HTLV-1) and has a poor prognosis. We analyzed the cytotoxic effects of various nucleoside analog reverse transcriptase inhibitors (NRTIs) for HIV-1 on ATL cells and found that abacavir potently and selectively kills ATL cells. Although NRTIs have minimal genotoxicities on host cells, the therapeutic concentration of abacavir induced numerous DNA double-strand breaks (DSBs) in the chromosomal DNA of ATL cells. DSBs persisted over time in ATL cells but not in other cell lines, suggesting impaired DNA repair. We found that the reduced expression of tyrosyl-DNA phosphodiesterase 1 (TDP1), a repair enzyme, is attributable to the cytotoxic effect of abacavir on ATL cells. We also showed that TDP1 removes abacavir from DNA ends in vitro. These results suggest a model in which ATL cells with reduced TDP1 expression are unable to excise abacavir incorporated into genomic DNA, leading to irreparable DSBs. On the basis of the above mechanism, we propose abacavir as a promising chemotherapeutic agent for ATL.

3.
Virol J ; 11: 122, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24986077

ABSTRACT

BACKGROUND: HIV-1 Vif is essential for virus replication in natural target cells such as T cells and macrophages. Vif recruits a ubiquitin ligase to degrade restrictive APOBEC3 proteins. APOBEC3G is one of the most potent retroviral restriction factors targeted by Vif and, as such, the Vif-APOBEC3G interaction has emerged as a promising HIV-1 therapeutic target. METHODS: 20,000 small molecules were used in live-cell screens for those that preserve EGFP-APOBEC3G fluorescence and luciferase-APOBEC3G luminescence in the presence of HIV-1 Vif. RESULTS: 2 compounds with similar core structures preserved APOBEC3G levels in the presence of Vif. 10 µM of compound restored APOBEC3G to levels sufficient for incorporation into vif-proficient virus particles and restriction of virus infectivity. Vif-dependent APOBEC3G polyubiquitination and general proteasomal activity were unaffected at the same concentration. CONCLUSIONS: The small molecules described here preserve APOBEC3G levels and activity in the presence of Vif. These molecules are starting points for further development as antiretrovirals.


Subject(s)
Antiviral Agents/isolation & purification , Cytidine Deaminase/metabolism , Enzyme Inhibitors/isolation & purification , vif Gene Products, Human Immunodeficiency Virus/antagonists & inhibitors , APOBEC-3G Deaminase , Drug Evaluation, Preclinical , Proteolysis/drug effects , vif Gene Products, Human Immunodeficiency Virus/metabolism
4.
PLoS One ; 9(1): e85762, 2014.
Article in English | MEDLINE | ID: mdl-24465689

ABSTRACT

The transcription factor NF-κB plays a key regulatory role in lymphocyte activation and generation of immune response. Stimulation of T cell receptor (TCR) induces phosphorylation of CARMA1 by PKCθ, resulting in formation of CARMA1-Bcl10-MALT1 (CBM) complex at lipid rafts and subsequently leading to NF-κB activation. While many molecular events leading to NF-κB activation have been reported, it is less understood how this activation is negatively regulated. We performed a cell-based screening for negative regulators of TCR-mediated NF-κB activation, using mutagenesis and complementation cloning strategies. Here we show that casein kinase-2 interacting protein-1 (CKIP-1) suppresses PKCθ-CBM-NF-κB signaling. We found that CKIP-1 interacts with CARMA1 and competes with PKCθ for association. We further confirmed that a PH domain of CKIP-1 is required for association with CARMA1 and its inhibitory effect. CKIP-1 represses NF-κB activity in unstimulated cells, and inhibits NF-κB activation induced by stimulation with PMA or constitutively active PKCθ, but not by stimulation with TNFα. Interestingly, CKIP-1 does not inhibit NF-κB activation induced by CD3/CD28 costimulation, which caused dissociation of CKIP-1 from lipid rafts. These data suggest that CKIP-1 contributes maintenance of a resting state on NF-κB activity or prevents T cells from being activated by inadequate signaling. In conclusion, we demonstrate that CKIP-1 interacts with CARMA1 and has an inhibitory effect on PKCθ-CBM-NF-κB signaling.


Subject(s)
CARD Signaling Adaptor Proteins/metabolism , Carrier Proteins/metabolism , Guanylate Cyclase/metabolism , Lymphocyte Activation/immunology , Carrier Proteins/chemistry , Enzyme Activation/drug effects , Humans , Intracellular Signaling Peptides and Proteins , Isoenzymes/metabolism , Jurkat Cells , Lymphocyte Activation/drug effects , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , NF-kappa B/metabolism , Protein Binding/drug effects , Protein Kinase C/metabolism , Protein Kinase C-theta , Protein Structure, Tertiary , T-Lymphocytes/drug effects , T-Lymphocytes/enzymology , T-Lymphocytes/immunology , Tetradecanoylphorbol Acetate/pharmacology
5.
Sci Rep ; 2: 806, 2012.
Article in English | MEDLINE | ID: mdl-23150777

ABSTRACT

Human APOBEC3 proteins play pivotal roles in intracellular defense against viral infection by catalyzing deamination of cytidine residues, leading to base substitutions in viral DNA. Activation-induced cytidine deaminase (AID), another member of the APOBEC family, is capable of editing immunoglobulin (Ig) and non-Ig genes, and aberrant expression of AID leads to tumorigenesis. However, it remains unclear whether APOBEC3 (A3) proteins affect stability of human genome. Here we demonstrate that both A3A and A3B can induce base substitutions into human genome as AID can. A3B is highly expressed in several lymphoma cells and somatic mutations occur in some oncogenes of the cells highly expressing A3B. Furthermore, transfection of A3B gene into lymphoma cells induces base substitutions in cMYC gene. These data suggest that aberrant expression of A3B can evoke genomic instability by inducing base substitutions into human genome, which might lead to tumorigenesis in human cells.


Subject(s)
Cytidine Deaminase/metabolism , Genome, Human , Base Sequence , Cell Line, Tumor , Cytidine Deaminase/genetics , Genomic Instability , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Minor Histocompatibility Antigens , Molecular Sequence Data , Mutation , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Transfection
6.
J Mol Biol ; 413(3): 667-83, 2011 Oct 28.
Article in English | MEDLINE | ID: mdl-21893069

ABSTRACT

Dysiherbaine (DH) and neodysiherbaine A (NDH) selectively bind and activate two kainate-type ionotropic glutamate receptors, GluK1 and GluK2. The ligand-binding domains of human GluK1 and GluK2 were crystallized as bound forms with a series of DH analogues including DH, NDH, 8-deoxy-NDH, 9-deoxy-NDH and 8,9-dideoxy-NDH (MSVIII-19), isolated from natural sources or prepared by total synthesis. Since the DH analogues exhibit a wide range of binding affinities and agonist efficacies, it follows that the detailed analysis of crystal structure would provide us with a significant opportunity to elucidate structural factors responsible for selective binding and some aspects of gating efficacy. We found that differences in three amino acids (Thr503, Ser706 and Ser726 in GluK1 and Ala487, Asn690 and Thr710 in GluK2) in the ligand-binding pocket generate differences in the binding modes of NDH to GluK1 and GluK2. Furthermore, deletion of the C(9) hydroxy group in NDH alters the ligand conformation such that it is no longer suited for binding to the GluK1 ligand-binding pocket. In GluK2, NDH pushes and rotates the side chain of Asn690 (substituted for Ser706 in GluK1) and disrupts an interdomain hydrogen bond with Glu409. The present data support the idea that receptor selectivities of DH analogues resulted from the differences in the binding modes of the ligands in GluK1/GluK2 and the steric repulsion of Asn690 in GluK2. All ligands, regardless of agonist efficacy, induced full domain closure. Consequently, ligand efficacy and domain closure did not directly coincide with DH analogues and the kainate receptors.


Subject(s)
Alanine/analogs & derivatives , Bridged Bicyclo Compounds, Heterocyclic/metabolism , Receptors, Kainic Acid/chemistry , Receptors, Kainic Acid/metabolism , Alanine/metabolism , Crystallography, X-Ray , Glutamic Acid/metabolism , Humans , Marine Toxins/metabolism , Models, Chemical , Models, Molecular , Protein Binding , Protein Conformation , GluK2 Kainate Receptor
7.
Proc Natl Acad Sci U S A ; 107(48): 20798-803, 2010 Nov 30.
Article in English | MEDLINE | ID: mdl-21071676

ABSTRACT

Viral infectivity factor, an accessory protein encoded in the HIV-1 genome, induces G2 cell cycle arrest; however, the biological significance and mechanism(s) remain totally unclear. Here we demonstrate that the TP53 pathway is involved in Vif-mediated G2 cell cycle arrest. Vif enhances the stability and transcriptional activity of TP53 by blocking the MDM2-mediated ubiquitination and nuclear export of TP53. Furthermore, Vif causes G2 cell cycle arrest in a TP53-dependent manner. HXB2 Vif lacks these activities toward TP53 and cannot induce G2 cell cycle arrest. Using mutagenesis, we demonstrate that the critical residues for this function are located in the N-terminal region of Vif. Finally, we construct a mutant NL4-3 virus with an NL4-3/HXB2 chimeric Vif defective for the ability to induce cell cycle arrest and show that the mutant virus replicates less effectively than the wild-type NL4-3 virus in T cells expressing TP53. These data imply that Vif induces G2 cell cycle arrest through functional interaction with the TP53/MDM2 axis and that the G2 cell cycle arrest induced by Vif has a positive effect on HIV-1 replication. This report demonstrates the molecular mechanisms and the biological significance of Vif-mediated G2 cell cycle arrest for HIV-1 infection.


Subject(s)
G2 Phase , HIV Infections/metabolism , HIV Infections/pathology , HIV-1/physiology , Tumor Suppressor Protein p53/metabolism , Virus Replication/physiology , vif Gene Products, Human Immunodeficiency Virus/metabolism , HCT116 Cells , Humans , Protein Stability , Proto-Oncogene Proteins c-mdm2/metabolism , Signal Transduction , Structure-Activity Relationship , T-Lymphocytes/virology , vif Gene Products, Human Immunodeficiency Virus/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...