Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Front Immunol ; 15: 1337520, 2024.
Article in English | MEDLINE | ID: mdl-38562937

ABSTRACT

This study investigates the neutralizing activity against the XBB1.5 variant and the ancestral strain in a population post-bivalent vaccination using a pseudo virus assay validated with authentic virus assay. While bivalent booster vaccination and past infections enhanced neutralization against the XBB 1.5 strain, individuals with comorbidities showed reduced responses. The study suggests the need for continuous vaccine updates to address emerging SARS-CoV-2 variants and highlights the importance of monitoring real-world immune responses.


Subject(s)
COVID-19 , Humans , Japan/epidemiology , COVID-19/prevention & control , SARS-CoV-2 , Vaccination , Surveys and Questionnaires , RNA, Messenger
2.
Nat Commun ; 13(1): 2405, 2022 05 03.
Article in English | MEDLINE | ID: mdl-35504920

ABSTRACT

Human T-cell leukemia virus type 1 (HTLV-1) is a retrovirus that causes adult T-cell leukemia/lymphoma (ATL), a cancer of infected CD4+ T-cells. There is both sense and antisense transcription from the integrated provirus. Sense transcription tends to be suppressed, but antisense transcription is constitutively active. Various efforts have been made to elucidate the regulatory mechanism of HTLV-1 provirus for several decades; however, it remains unknown how HTLV-1 antisense transcription is maintained. Here, using proviral DNA-capture sequencing, we found a previously unidentified viral enhancer in the middle of the HTLV-1 provirus. The transcription factors, SRF and ELK-1, play a pivotal role in the activity of this enhancer. Aberrant transcription of genes in the proximity of integration sites was observed in freshly isolated ATL cells. This finding resolves certain long-standing questions concerning HTLV-1 persistence and pathogenesis. We anticipate that the DNA-capture-seq approach can be applied to analyze the regulatory mechanisms of other oncogenic viruses integrated into the host cellular genome.


Subject(s)
Human T-lymphotropic virus 1 , Leukemia-Lymphoma, Adult T-Cell , DNA , Human T-lymphotropic virus 1/genetics , Humans , Leukemia-Lymphoma, Adult T-Cell/genetics , Proviruses/genetics , Regulatory Sequences, Nucleic Acid
3.
PLoS Pathog ; 17(11): e1010126, 2021 11.
Article in English | MEDLINE | ID: mdl-34843591

ABSTRACT

Human T-cell leukemia virus type 1 (HTLV-1) infects target cells primarily through cell-to-cell routes. Here, we provide evidence that cellular protein M-Sec plays a critical role in this process. When purified and briefly cultured, CD4+ T cells of HTLV-1 carriers, but not of HTLV-1- individuals, expressed M-Sec. The viral protein Tax was revealed to mediate M-Sec induction. Knockdown or pharmacological inhibition of M-Sec reduced viral infection in multiple co-culture conditions. Furthermore, M-Sec knockdown reduced the number of proviral copies in the tissues of a mouse model of HTLV-1 infection. Phenotypically, M-Sec knockdown or inhibition reduced not only plasma membrane protrusions and migratory activity of cells, but also large clusters of Gag, a viral structural protein required for the formation of viral particles. Taken together, these results suggest that M-Sec induced by Tax mediates an efficient cell-to-cell viral infection, which is likely due to enhanced membrane protrusions, cell migration, and the clustering of Gag.


Subject(s)
Cell Membrane/virology , Disease Models, Animal , Gene Products, tax/metabolism , HTLV-I Infections/transmission , Human T-lymphotropic virus 1/physiology , Tumor Necrosis Factors/metabolism , Viral Structural Proteins/metabolism , Animals , Cell Membrane/metabolism , Cell Movement , Coculture Techniques , Gene Products, tax/genetics , HTLV-I Infections/metabolism , HTLV-I Infections/virology , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Tumor Necrosis Factors/genetics , Viral Structural Proteins/genetics
4.
PLoS Pathog ; 17(5): e1009577, 2021 05.
Article in English | MEDLINE | ID: mdl-34019588

ABSTRACT

Human T-cell leukemia virus type 1 (HTLV-1) is a retrovirus that causes an aggressive T-cell malignancy and a variety of inflammatory conditions. The integrated provirus includes a single binding site for the epigenomic insulator, CCCTC-binding protein (CTCF), but its function remains unclear. In the current study, a mutant virus was examined that eliminates the CTCF-binding site. The mutation did not disrupt the kinetics and levels of virus gene expression, or establishment of or reactivation from latency. However, the mutation disrupted the epigenetic barrier function, resulting in enhanced DNA CpG methylation downstream of the CTCF binding site on both strands of the integrated provirus and H3K4Me3, H3K36Me3, and H3K27Me3 chromatin modifications both up- and downstream of the site. A majority of clonal cell lines infected with wild type HTLV-1 exhibited increased plus strand gene expression with CTCF knockdown, while expression in mutant HTLV-1 clonal lines was unaffected. These findings indicate that CTCF binding regulates HTLV-1 gene expression, DNA and histone methylation in an integration site dependent fashion.


Subject(s)
Epigenesis, Genetic , Genome, Viral/genetics , HTLV-I Infections/virology , Human T-lymphotropic virus 1/genetics , Leukemia, T-Cell/virology , Binding Sites , CCCTC-Binding Factor/genetics , CCCTC-Binding Factor/metabolism , Cell Line , Chromatin/genetics , DNA Methylation , Epigenomics , Human T-lymphotropic virus 1/physiology , Humans , Mutation , Virus Integration , Virus Latency/genetics
5.
Cell Rep ; 29(3): 724-735.e4, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31618639

ABSTRACT

The retrovirus human T-cell leukemia virus type 1 (HTLV-1) integrates into the host DNA, achieves persistent infection, and induces human diseases. Here, we demonstrate that viral DNA-capture sequencing (DNA-capture-seq) is useful to characterize HTLV-1 proviruses in naturally virus-infected individuals, providing comprehensive information about the proviral structure and the viral integration site. We analyzed peripheral blood from 98 naturally HTLV-1-infected individuals and found that defective proviruses were present not only in patients with leukemia, but also in those with other clinical entities. We further demonstrated that clones with defective-type proviruses exhibited a higher degree of clonal abundance than those with full-length proviruses. The frequency of defective-type proviruses in HTLV-1-infected humanized mice was lower than that in infected individuals, indicating that defective proviruses were rare at the initial phase of infection but preferentially selected during persistent infection. These results demonstrate the robustness of viral DNA-capture-seq for HTLV-1 infection and suggest potential applications for other virus-associated cancers in humans.


Subject(s)
Genome, Viral , HTLV-I Infections/diagnosis , Human T-lymphotropic virus 1/genetics , Animals , HTLV-I Infections/virology , High-Throughput Nucleotide Sequencing , Human T-lymphotropic virus 1/physiology , Humans , Jurkat Cells , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/virology , Mice , Models, Animal , Sequence Analysis, DNA , Virus Integration
6.
PLoS Pathog ; 13(11): e1006722, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29186194

ABSTRACT

Human T-cell leukemia virus type 1 (HTLV-1) infects mainly CD4+CCR4+ effector/memory T cells in vivo. However, it remains unknown whether HTLV-1 preferentially infects these T cells or this virus converts infected precursor cells to specialized T cells. Expression of viral genes in vivo is critical to study viral replication and proliferation of infected cells. Therefore, we first analyzed viral gene expression in non-human primates naturally infected with simian T-cell leukemia virus type 1 (STLV-1), whose virological attributes closely resemble those of HTLV-1. Although the tax transcript was detected only in certain tissues, Tax expression was much higher in the bone marrow, indicating the possibility of de novo infection. Furthermore, Tax expression of non-T cells was suspected in bone marrow. These data suggest that HTLV-1 infects hematopoietic cells in the bone marrow. To explore the possibility that HTLV-1 infects hematopoietic stem cells (HSCs), we analyzed integration sites of HTLV-1 provirus in various lineages of hematopoietic cells in patients with HTLV-1 associated myelopathy/tropical spastic paraparesis (HAM/TSP) and a HTLV-1 carrier using the high-throughput sequencing method. Identical integration sites were detected in neutrophils, monocytes, B cells, CD8+ T cells and CD4+ T cells, indicating that HTLV-1 infects HSCs in vivo. We also detected Tax protein in myeloperoxidase positive neutrophils. Furthermore, dendritic cells differentiated from HTLV-1 infected monocytes caused de novo infection to T cells, indicating that infected monocytes are implicated in viral spreading in vivo. Certain integration sites were re-detected in neutrophils from HAM/TSP patients at different time points, indicating that infected HSCs persist and differentiate in vivo. This study demonstrates that HTLV-1 infects HSCs, and infected stem cells differentiate into diverse cell lineages. These data indicate that infection of HSCs can contribute to the persistence and spread of HTLV-1 in vivo.


Subject(s)
HTLV-I Infections/virology , Hematopoietic Stem Cells/virology , Human T-lymphotropic virus 1/physiology , Animals , CD8-Positive T-Lymphocytes/virology , Cells, Cultured , Gene Products, tax/genetics , Gene Products, tax/metabolism , HTLV-I Infections/immunology , Human T-lymphotropic virus 1/genetics , Humans , Macaca mulatta , Neutrophils/virology
7.
Blood ; 123(3): 346-55, 2014 Jan 16.
Article in English | MEDLINE | ID: mdl-24196073

ABSTRACT

Human T-cell leukemia virus type 1 (HTLV-1) is causally associated with adult T-cell leukemia (ATL), an aggressive T-cell malignancy with a poor prognosis. To elucidate ATL pathogenesis in vivo, a variety of animal models have been established; however, the mechanisms driving this disorder remain poorly understood due to deficiencies in each of these animal models. Here, we report a novel HTLV-1-infected humanized mouse model generated by intra-bone marrow injection of human CD133(+) stem cells into NOD/Shi-scid/IL-2Rγc null (NOG) mice (IBMI-huNOG mice). Upon infection, the number of CD4(+) human T cells in the periphery increased rapidly, and atypical lymphocytes with lobulated nuclei resembling ATL-specific flower cells were observed 4 to 5 months after infection. Proliferation was seen in both CD25(-) and CD25(+) CD4 T cells with identical proviral integration sites; however, a limited number of CD25(+)-infected T-cell clones eventually dominated, indicating an association between clonal selection of infected T cells and expression of CD25. Additionally, HTLV-1-specific adaptive immune responses were induced in infected mice and might be involved in the control of HTLV-1-infected cells. Thus, the HTLV-1-infected IBMI-huNOG mouse model successfully recapitulated the development of ATL and may serve as an important tool for investigating in vivo mechanisms of ATL leukemogenesis and evaluating anti-ATL drug and vaccine candidates.


Subject(s)
Disease Models, Animal , HTLV-I Infections/immunology , Leukemia-Lymphoma, Adult T-Cell/immunology , AC133 Antigen , Animals , Antigens, CD/metabolism , Bone Marrow/metabolism , CD4-Positive T-Lymphocytes/cytology , Cell Separation , Female , Fetal Blood/metabolism , Flow Cytometry , Glycoproteins/metabolism , Hematopoietic Stem Cells/cytology , Human T-lymphotropic virus 1/immunology , Humans , Inflammation , Interleukin-2 Receptor alpha Subunit/metabolism , Leukemia-Lymphoma, Adult T-Cell/virology , Mice , Mice, Inbred NOD , Peptides/metabolism , Spleen/cytology , Stem Cells/cytology , Viral Load
8.
Nihon Shokakibyo Gakkai Zasshi ; 108(11): 1902-9, 2011 Nov.
Article in Japanese | MEDLINE | ID: mdl-22056712

ABSTRACT

A 68-year-old man with chronic hepatitis C and of a heavy drinker was admitted to our hospital because of a huge liver tumor (10cm in diameter) in segment-5 detected on CT in July 2009. One month later, the size of liver tumor was reduced to 5cm in diameter and another liver tumor of 1cm in segment-3 was detected on CT. AFP and AFP-L3 spontaneously decreased to normal range. In October, a partial hepatectomy was performed. The resected specimen demonstrated complete necrosis with thick capsule. The tumor in segment-3 became equivocal without resection. We considered this is a case of spontaneous complete necrosis of hepatocellular carcinoma.


Subject(s)
Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Aged , Carcinoma, Hepatocellular/surgery , Humans , Liver Neoplasms/surgery , Male , Necrosis , Neoplasm Regression, Spontaneous
9.
FEBS Lett ; 585(7): 1103-11, 2011 Apr 06.
Article in English | MEDLINE | ID: mdl-21402072

ABSTRACT

Pharmacological manipulations to purge human immunodeficiency virus (HIV) from latent reservoirs have been considered as an adjuvant therapeutic approach to highly-active antiretroviral therapy for the eradication of HIV. Our novel histone deacetylase inhibitor NCH-51 induced expression of latent HIV-1 with minimal cytotoxicity. Using chromatin immunoprecipitation assays, we observed a reduction of HDAC1 occupancy, histone hyperacetylation and the recruitment of positive transcription factors at the HIV-1 promoter in latently infected-cells under the treatment with NCH-51. Mutation studies of the long terminal repeat (LTR) revealed NCH-51 mediated gene expression through the Sp1 sites. When Sp1 expression was knocked-down by small interfering RNA, the NCH-51-mediated activation of a stably integrated HIV-1 LTR was attenuated. Moreover, the Sp1 inhibitor mithramycin A abolished the effects of NCH-51.


Subject(s)
Gene Expression Regulation, Viral/drug effects , HIV-1/drug effects , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Sulfhydryl Compounds/pharmacology , Virus Latency/drug effects , Virus Latency/genetics , Acetylation/drug effects , Chromatin Assembly and Disassembly/drug effects , Gene Knockdown Techniques , HIV-1/genetics , HIV-1/physiology , HL-60 Cells , Histones/metabolism , Humans , Nucleosomes/drug effects , Nucleosomes/metabolism , Promoter Regions, Genetic/genetics , Sp1 Transcription Factor/deficiency , Sp1 Transcription Factor/genetics , Terminal Repeat Sequences/genetics , Transcriptional Activation/drug effects , Virus Replication/drug effects
10.
J Biol Chem ; 283(12): 7834-43, 2008 Mar 21.
Article in English | MEDLINE | ID: mdl-18178962

ABSTRACT

In this study, we have identified protein kinase A-interacting protein 1 (AKIP1) as a binding partner of NF-kappaB p65 subunit, and AKIP1 enhances the NF-kappaB-mediated gene expression. AKIP1 is a nuclear protein and known to interact with the catalytic subunit of PKA (PKAc). We identified AKIP1 by a yeast two-hybrid screen using the N terminus region of p65 as bait. The interaction between AKIP1 and p65 was confirmed by glutathione S-transferase pull-down assay in vitro and immunoprecipitation-Western blotting assay in vivo. We found that the PKAc was present in the AKIP1.p65 complex and enhanced the transcriptional activity of NF-kappaB by phosphorylating p65. In a transient luciferase assay, AKIP1 cotransfection efficiently increased the transcriptional activity of NF-kappaB induced by phorbol 12-myristate 13-acetate (PMA). When AKIP1 was knocked down by RNA interference, the PMA-mediated NF-kappaB-dependent gene expression was abolished, indicating a physiological role of AKIP1. We found that PKAc, which is maintained in an inactive form by binding to IkappaBalpha and NF-kappaB in resting cells, was activated by PMA-induced signaling and could phosphorylate p65. Overexpression of AKIP1 increased the PKAc binding to p65 and enhanced the PKAc-mediated phosphorylation of p65 at Ser-276. Interestingly, this p65 phosphorylation promoted nuclear translocation of p65 and enhanced NF-kappaB transcription. In fact, we observed that AKIP1 colocalized with p65 within the cells and appeared to retain p65 in nucleus. These findings indicate a positive role of AKIP1 in NF-kappaB signaling and suggest a novel mechanism by which AKIP1 augments the transcriptional competence of NF-kappaB.


Subject(s)
Cell Nucleus/metabolism , Gene Expression Regulation/physiology , Multiprotein Complexes/metabolism , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Transcription Factor RelA/metabolism , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/physiology , Adaptor Proteins, Signal Transducing , Carcinogens/pharmacology , Cell Nucleus/genetics , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Expression Regulation/drug effects , HeLa Cells , Humans , Multiprotein Complexes/genetics , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Phosphorylation , Protein Structure, Tertiary/physiology , RNA Interference , Signal Transduction/drug effects , Signal Transduction/physiology , Tetradecanoylphorbol Acetate/pharmacology , Transcription Factor RelA/genetics , Two-Hybrid System Techniques
11.
Microbes Infect ; 8(4): 984-93, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16500131

ABSTRACT

Most human papillomavirus (HPV)-positive cervical cancers contain integrated copies of the viral genome in their chromosomes and express the viral oncoproteins E6 and E7. A virus-encoded transcription factor, E2, is known to repress E6/E7 expression in HPV-positive cancer cells, leading to growth inhibition, which indicates that E6/E7 is required for the survival of the cells. We found that the E2-mediated growth inhibition of HeLa cells, an HPV18-positive cancer cell line, was coupled with a reduction in telomerase activity, an effect which was rescued by the complementation of E7 expression, but not E6 expression, indicating that the cell viability and the telomerase activity in HeLa cells are maintained by an E7-associated function. Analysis of E7 mutants suggested that the binding to the pRB family of pocket proteins was involved in the ability of E7 to rescue the growth potential and telomerase activity inhibited by E2 expression. We also showed that the telomerase activity upregulated by E7 expression was determined by the hTERT promoter activity, and that c-Myc upregulation caused by pRB inactivation could account for the promoter activity. The activation of p53 and consequent accumulation of p21Cip1, which were triggered by the downregulation of E6, appeared not to be essential for the E2-mediated growth arrest.


Subject(s)
DNA-Binding Proteins/physiology , Oncogene Proteins, Viral/physiology , Cell Cycle Proteins , Cell Survival , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Genes, myc/genetics , HeLa Cells/physiology , Humans , Nuclear Proteins/metabolism , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Promoter Regions, Genetic/genetics , Repressor Proteins/metabolism , Telomerase/metabolism , Up-Regulation
12.
Virology ; 311(2): 316-25, 2003 Jul 05.
Article in English | MEDLINE | ID: mdl-12842621

ABSTRACT

Downregulation of virus receptors on the cell surface is considered to be important in preventing superinfection. HIV-1 encodes multiple gene products, Env, Vpu, and Nef, involved in downregulation of CD4, a major HIV-1 receptor. We found that simultaneous mutations in both vpu and nef severely impaired virus replication. We examined the involvement of CD4 downregulation mediated by Vpu and Nef in the modification of virus infectivity. The mutation in vpu increased CD4 incorporation into virions without affecting the Env content in it, inhibiting the attachment step of virions to the CD4-positive cell surface. Although a single mutation in nef suppresses virus infectivity via a CD4-independent mechanism, it could augment CD4 incorporation in virions in combination with a vpu mutation. These results indicated that CD4 downregulation was necessary for maintenance of Env function in the virion.


Subject(s)
CD4 Antigens/metabolism , Down-Regulation , HIV-1/physiology , HIV-1/pathogenicity , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , Gene Products, env/metabolism , Gene Products, nef/metabolism , HIV-1/chemistry , HIV-1/genetics , Human Immunodeficiency Virus Proteins , Humans , Superinfection/metabolism , Superinfection/virology , Viral Regulatory and Accessory Proteins/genetics , Viral Regulatory and Accessory Proteins/metabolism , Virion/chemistry , Virion/metabolism , Virus Replication , nef Gene Products, Human Immunodeficiency Virus
13.
Free Radic Res ; 37(2): 159-69, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12653204

ABSTRACT

In gastric mucosal injury, nitric oxide (NO) plays both cytoprotective and cytotoxic roles, and the NO level is one determinant of these dual roles. We employed electron paramagnetic resonance (EPR)-spectrometry combined with an NO-trapping technique to directly evaluate NO production in ethanol-induced gastric injury in rats. The rat stomach, mounted on an ex vivo chamber, was perfused with ethanol (12.5 and 43%), and NO levels in mucosal tissues were measured during perfusion. Luminal nitrite/nitrate (NOx) content, mucosal blood flow, area of mucosal injury, transmucosal potential difference (PD), and luminal pH were simultaneously monitored with/without preadministration of the NO synthase inhibitor, N(G)-nitro-L-arginine methyl ester (L-NAME). NO levels in the gastric tissue increased during ethanol perfusion, and luminal NOx levels increased after the perfusion, accompanying an increase in the area of mucosal injury and changes in physiological parameters. Preadministration of L-NAME aggravated the gastric mucosal damage and suppressed increases in mucosal blood flow in a dose-dependent manner. These results demonstrate that endogenous NO produced in ethanol-induced gastric injury contributes to maintenance of mucosal integrity via regulation of mucosal blood flow.


Subject(s)
Ethanol/pharmacology , Gastric Mucosa/pathology , Nitric Oxide , Animals , Chromatography, High Pressure Liquid , Electron Spin Resonance Spectroscopy , Gastric Mucosa/injuries , Gastric Mucosa/metabolism , Hydrogen-Ion Concentration , Male , NG-Nitroarginine Methyl Ester/metabolism , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Time Factors
14.
Am J Respir Crit Care Med ; 167(2): 205-10, 2003 Jan 15.
Article in English | MEDLINE | ID: mdl-12524256

ABSTRACT

Alveolar fluid clearance may be inhibited and/or stimulated under pathologic conditions. We examined the early change of alveolar fluid clearance after endotoxin instillation in adult rats. We employed electron paramagnetic resonance nitric oxide (NO) trapping technique with iron complex with N,N-diethyldithiocarbamate as an NO trapping agent. We found that lung NO signals reached the highest magnitude by 6 hours after endotoxin instillation. NO production was accompanied by increases in lung cyclic guanosine monophosphate levels. Alveolar fluid clearance decreased significantly 6 hours after the administration of the endotoxin and increased further at 24 hours. These changes were shown to be related to the function of amiloride-sensitive sodium ion channels. Treatment with gadolinium chloride and aminoguanidine significantly decreased lung NO and cyclic guanosine monophosphate levels and completely ameliorated the decrease in alveolar fluid clearance. In addition, the increase in alveolar fluid clearance at 24 hours returned to normal levels after treatment with gadolinium chloride and aminoguanidine. We found immunoreactive inducible nitric oxide synthase to be abundantly expressed in the cytoplasm of alveolar macrophages. Our results suggest that alveolar endotoxin inhibits alveolar fluid clearance at 6 hours by NO. NO is produced via inducible NO synthase in endotoxin-stimulated alveolar macrophages and was also shown to increase alveolar fluid clearance at 24 hours.


Subject(s)
Lung/pathology , Macrophages, Alveolar/enzymology , Nitric Oxide Synthase/biosynthesis , Nitric Oxide/biosynthesis , Pulmonary Edema/metabolism , Animals , Bronchoalveolar Lavage Fluid/chemistry , Disease Models, Animal , Endotoxins , Escherichia coli , Immunohistochemistry , Instillation, Drug , Male , Nitric Oxide/analysis , Nitric Oxide Synthase/analysis , Nitric Oxide Synthase Type II , Probability , Pulmonary Edema/physiopathology , Random Allocation , Rats , Rats, Sprague-Dawley , Reference Values , Time Factors
15.
J Virol ; 76(21): 10914-20, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12368334

ABSTRACT

The life cycle of human papillomaviruses (HPVs) is tightly coupled to the differentiation program of their host epithelial cells. HPV E4 gene expression is first observed in the parabasal layers of squamous epithelia, suggesting that the E4 gene product contributes to the mechanism of differentiation-dependent virus replication, although its biological function remains unclear. We analyzed the effect of HPV type 18 E4 on cell proliferation and found that E4 expression induced cell cycle arrest at the G(2)/M boundary. The functional region of E4 necessary for the growth arrest activity was located in the central portion of the molecule, and this activity was independent of the E4-mediated collapse of cytokeratin intermediate filament structures.


Subject(s)
Oncogene Proteins, Viral/metabolism , Papillomaviridae/metabolism , Animals , Cell Cycle , Cell Division , Cell Line , Chlorocebus aethiops , G2 Phase , HeLa Cells , Humans , Keratins/metabolism , Mitosis , Oncogene Proteins, Viral/genetics
16.
Nihon Rinsho ; 60(4): 674-80, 2002 Apr.
Article in Japanese | MEDLINE | ID: mdl-11968772

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1) has 4 auxiliary genes, vpr, vpu, nef, and vif, which are dispensable for viral replication in vitro. However, many studies with animal model revealed that these genes play important roles on the viral replication and the development of AIDS in vivo through many complicated mechanisms. Although several key factors involved in the function have been identified, further studies are required for the complete understandings of the action mechanisms. The elucidation of the function of the auxiliary genes on molecular bases leads to the discovery of new therapeutic strategies against HIV and the understanding of basic cellular mechanisms. In this review, we summarize new observations mainly about the interactions between auxiliary genes and host cell functions.


Subject(s)
Genes, nef/physiology , Genes, vif/physiology , Genes, vpr/physiology , Genes, vpu/physiology , HIV-1/genetics , Acquired Immunodeficiency Syndrome/virology , Animals , HIV-1/physiology , Humans , Virus Replication
17.
Biochem Pharmacol ; 63(3): 485-93, 2002 Feb 01.
Article in English | MEDLINE | ID: mdl-11853699

ABSTRACT

Dinitrosyl dithiolato iron complex (DNIC) has been identified as an endogenous NO carrier, yet in vivo mechanisms of NO donation remain undefined. Transnitrosylation, in which a coordinated NO group is transferred to another metal complex, has been observed in transition-metal-nitrosyl chemistry. In this study, we used three kinds of iron dithiocarbamate complexes (Fe-DTCs) as NO acceptors to elucidate in vivo transnitrosylation of diglutathionyl dinitrosyl iron complex [DNIC-(GS)(2)]. Fe-DTCs were administered to mice after the injection of DNIC-(GS)(2) and electron paramagnetic resonance (EPR) spectra were measured both in the resected organs and in the upper abdomen of living mice. The spectral feature gradually changed from an initial DNIC-(GS)(2) signal to mononitrosyl iron dithiocarbamate one, suggesting that NO-Fe-DTC was formed through in vivo reaction of DNIC-(GS)(2) with Fe-DTC. The spectral results in in vitro and in vivo systems indicate that NO-Fe-DTCs can be formed not only by the transfer of coordinated NO-group(s) in DNIC-(GS)(2) but also by the abstraction of Fe-NO group in DNIC-(GS)(2) by free DTC ligands. Transnitrosylation proceeded more rapidly in blood than in liver and kidney; and more efficiently in kidney than in liver. Further, the ability to accept NO from DNIC was dependent on water-solubility of Fe-DTCs. Thus, in vivo transnitrosylation from DNIC to exogenous iron complex could be observed and this reaction was influenced by biological constituents and properties of iron complex. These results demonstrate that the transnitrosylation from DNIC to intrinsic NO acceptors like metalloproteins has a probable significance in in vivo NO transfer process.


Subject(s)
Iron/metabolism , Nitric Oxide/metabolism , Nitrogen Oxides/metabolism , Animals , Biological Transport , Electron Spin Resonance Spectroscopy , Female , Ferric Compounds/metabolism , Mice , Mice, Inbred ICR , Thiocarbamates/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...