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











Publication year range
1.
J Virol ; 89(4): 2415-24, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25505075

ABSTRACT

UNLABELLED: Members of the APOBEC3 family of cytidine deaminases vary in their proportions of a virion-incorporated enzyme that is localized to mature retrovirus cores. We reported previously that APOBEC3F (A3F) was highly localized into mature human immunodeficiency virus type 1 (HIV-1) cores and identified that L306 in the C-terminal cytidine deaminase (CD) domain contributed to its core localization (C. Song, L. Sutton, M. Johnson, R. D'Aquila, J. Donahue, J Biol Chem 287:16965-16974, 2012, http://dx.doi.org/10.1074/jbc.M111.310839). We have now determined an additional genetic determinant(s) for A3F localization to HIV-1 cores. We found that one pair of leucines in each of A3F's C-terminal and N-terminal CD domains jointly determined the degree of localization of A3F into HIV-1 virion cores. These are A3F L306/L368 (C-terminal domain) and A3F L122/L184 (N-terminal domain). Alterations to one of these specific leucine residues in either of the two A3F CD domains (A3F L368A, L122A, and L184A) decreased core localization and diminished HIV restriction without changing virion packaging. Furthermore, double mutants in these leucine residues in each of A3F's two CD domains (A3F L368A plus L184A or A3F L368A plus L122A) still were packaged into virions but completely lost core localization and anti-HIV activity. HIV virion core localization of A3F is genetically separable from its virion packaging, and anti-HIV activity requires some core localization. IMPORTANCE: Specific leucine-leucine interactions are identified as necessary for A3F's core localization and anti-HIV activity but not for its packaging into virions. Understanding these signals may lead to novel strategies to enhance core localization that may augment effects of A3F against HIV and perhaps of other A3s against retroviruses, parvoviruses, and hepatitis B virus.


Subject(s)
Cytosine Deaminase/analysis , Cytosine Deaminase/genetics , HIV-1/physiology , Virus Assembly , Cell Line , Cytosine Deaminase/immunology , DNA Mutational Analysis , Genes, Reporter , HIV-1/chemistry , HIV-1/immunology , Humans , Luciferases/analysis , Models, Molecular , Mutagenesis, Site-Directed , Mutation, Missense , Staining and Labeling , beta-Galactosidase/analysis
2.
Tumori ; 100(4): 112e-7e, 2014.
Article in English | MEDLINE | ID: mdl-25296601

ABSTRACT

AIMS AND BACKGROUND: APOBEC3B is a deaminase that possesses DNA C-to-T editing activity. A recent report showed that APOBEC3B mRNA was overexpressed in breast cancer and that its expression was responsible for the high C-to-T mutation spectrum in breast cancer, suggesting that APOBEC3B could serve as a source for producing mutations. To see whether APOBEC3B is overexpressed in other common cancers at the protein level, we investigated APOBEC3 protein expression in 100 gastric, 103 colorectal and 107 prostate cancer tissues as well as in 10 breast cancers by immunohistochemistry using antibody that could detect APOBEC3B, APOBEC3F and APOBEC3D proteins. RESULTS: In the cancers, APOBEC3 expression was detected in 100% of breast cancers, 67% of gastric, 84% of colorectal and 67% of prostate cancer. Also, it was expressed in 100% of normal breast, 90% of normal stomach, 82% of normal colon and 93% of normal prostate tissues. In contrast to earlier data that showed an increased APOBEC3B expression in breast cancer cells compared to normal breast cells, APOBEC3 expression in cancers was lower than in normal tissues (gastric and prostate cancer) or was not different from normal tissues (colorectal and breast cancer). There was no significant association of APOBEC3 expression with clinocopathological parameters, including histology, metastasis and stage. CONCLUSIONS: Our data indicate that APOBEC3 overexpression might not be restricted to specific cancer types. Also, APOBEC3 expression in many normal epithelial cells suggests that there might be a mutation unrelated function of APOBEC3 in the normal cells.


Subject(s)
Biomarkers, Tumor/analysis , Colorectal Neoplasms/chemistry , Cytosine Deaminase/analysis , Prostatic Neoplasms/chemistry , Stomach Neoplasms/chemistry , APOBEC Deaminases , Adult , Aged , Aged, 80 and over , Colorectal Neoplasms/genetics , Cytidine Deaminase , Cytosine Deaminase/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Male , Middle Aged , Mutation , Prostatic Neoplasms/genetics , Stomach Neoplasms/genetics , Tissue Array Analysis
3.
Methods Mol Biol ; 756: 395-425, 2011.
Article in English | MEDLINE | ID: mdl-21870242

ABSTRACT

Protein-fragment Complementation Assays (PCAs) are a family of assays for detecting protein-protein interactions (PPIs) that have been developed to provide simple and direct ways to study PPIs in any living cell, multicellular organism, or in vitro. PCAs can be used to detect PPI between proteins of any molecular weight and expressed at their endogenous levels. Proteins are expressed in their appropriate cellular compartments and can undergo any posttranslational modification or degradation that, barring effects of the PCA fragment fusion, they would normally undergo. Assays can be performed in any cell type or model organism that can be transformed or transfected with gene expression DNA constructs. Here we focus on recent applications of PCA in the budding yeast, Saccharomyces cerevisiae, that cover the gamut of applications one could envision for studying any aspect of PPIs. We present detailed protocols for large-scale analysis of PPIs with the survival-selection dihydrofolate reductase (DHFR), reporter PCA, and a new PCA based on a yeast cytosine deaminase reporter that allows for both survival and death selection. This PCA should prove a powerful way to dissect PPIs. We then present methods to study spatial localization and dynamics of PPIs based on fluorescent protein reporter PCAs.


Subject(s)
Protein Interaction Mapping/methods , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , Animals , Cytosine Deaminase/analysis , Cytosine Deaminase/metabolism , Green Fluorescent Proteins/analysis , Luciferases, Renilla/analysis , Luminescent Agents/analysis , Luminescent Measurements/methods , Microscopy, Fluorescence/methods , Models, Molecular , Renilla/enzymology , Saccharomyces cerevisiae Proteins/analysis , Tetrahydrofolate Dehydrogenase/analysis , Tetrahydrofolate Dehydrogenase/metabolism
4.
J Virol ; 84(21): 11067-75, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20702622

ABSTRACT

APOBEC3F (A3F) is a member of the family of cytidine deaminases that is often coexpressed with APOBEC3G (A3G) in cells susceptible to HIV infection. A3F has been shown to have strong antiviral activity in transient-expression studies, and together with A3G, it is considered the most potent cytidine deaminase targeting HIV. Previous analyses suggested that the antiviral properties of A3F can be dissociated from its catalytic deaminase activity. We were able to confirm the deaminase-independent antiviral activity of exogenously expressed A3F; however, we also noted that exogenous expression was associated with very high A3F mRNA and protein levels. In analogy to our previous study of A3G, we produced stable HeLa cell lines constitutively expressing wild-type or deaminase-defective A3F at levels that were more in line with the levels of endogenous A3F in H9 cells. A3F expressed in stable HeLa cells was packaged into Vif-deficient viral particles with an efficiency similar to that of A3G and was properly targeted to the viral nucleoprotein complex. Surprisingly, however, neither wild-type nor deaminase-defective A3F inhibited HIV-1 infectivity. These results imply that the antiviral activity of endogenous A3F is negligible compared to that of A3G.


Subject(s)
Cytidine Deaminase/immunology , Cytosine Deaminase/immunology , APOBEC-3G Deaminase , Cytosine Deaminase/analysis , Cytosine Deaminase/genetics , HIV Infections , HIV-1/pathogenicity , HeLa Cells , Humans , RNA, Messenger/analysis , Virion , vif Gene Products, Human Immunodeficiency Virus
5.
J Virol ; 84(1): 88-95, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19828612

ABSTRACT

The APOBEC3H gene is polymorphic in humans, with four major population-dependent haplotypes that encode proteins with different levels of antiviral activity. Haplotype II, present most frequently in African populations, encodes the most stable protein and is most active against human immunodeficiency virus type 1 (HIV-1). In contrast to human APOBEC3G, which can be completely counteracted by HIV-1 Vif, the protein encoded by APOBEC3H haplotype II is only partially sensitive to Vif, while the protein encoded by APOBEC3H haplotype I is completely resistant to HIV-1 Vif. We mapped a residue on APOBEC3H that determines this partial Vif sensitivity. However, it is unclear how HIV-1 can replicate in vivo without the ability to neutralize APOBEC3H antiviral activity. In order to directly address this question, we cloned vif genes from HIV-1-infected individuals with different APOBEC3H genotypes and tested them for their ability to inhibit human APOBEC3H. We found that while the APOBEC3H genotype of infected individuals significantly influences the activity of Vif encoded by their virus, none of the Vif variants tested can completely neutralize APOBEC3H as well as they neutralize APOBEC3G. Consistent with this genetic result, APOBEC3H protein expression in human peripheral blood mononuclear cells was below our limit of detection using newly developed antibodies against the endogenous protein. These results demonstrate that human APOBEC3H is not as strong of a selective force for current HIV-1 infections as human APOBEC3G.


Subject(s)
Cytosine Deaminase/antagonists & inhibitors , Gene Products, vif/physiology , HIV-1/chemistry , Polymorphism, Genetic , APOBEC-3G Deaminase , Aminohydrolases , Cloning, Molecular , Cytidine Deaminase/antagonists & inhibitors , Cytosine Deaminase/analysis , Gene Products, vif/genetics , Genotype , Haplotypes , Human Immunodeficiency Virus Proteins/physiology , Humans , Leukocytes, Mononuclear/chemistry , Leukocytes, Mononuclear/virology
6.
Acta Oncol ; 47(7): 1211-20, 2008.
Article in English | MEDLINE | ID: mdl-18661431

ABSTRACT

INTRODUCTION: Increased expression of cytosine deaminase (CD) and uracil phosphoribosyltransferase (UPRT) may improve the antitumoral effect of 5-fluorouracil (5-FU) and 5-fluorocytosine (5-FC), and thereby enhance the potential of gene-directed enzyme prodrug therapy. For the applicability of gene-directed enzyme prodrug therapy in a clinical setting, it is essential to be able to monitor the transgene expression and function in vivo. Thus, we developed a preclinical tumor model to investigate the feasibility of using magnetic resonance spectroscopy and optical imaging to measure non-invasively CD and UPRT expression and function. MATERIALS AND METHODS: Expression vectors of CD or CD/UPRT fused to monomeric DsRed (mDsRed) were constructed and rat prostate carcinoma (R3327-AT) cell lines stably expressing either CD/mDsRed or CD/UPRT/mDsRed were generated. The expression of the fusion proteins was evaluated by flow cytometry, fluorescence microscopy, and Western blot analysis. The function of the fusion protein was confirmed in vitro by assessing 5-FC and 5-FU cytotoxicity. In vivo fluorine-19 magnetic resonance spectroscopy ((19)F MRS) was used to monitor the conversion of 5-FC to 5-FU in mice bearing the R3327-CD/mDsRed and R3327-CD/UPRT/mDsRed tumor xenografts. RESULTS: Sensitivity to 5-FC and 5-FU was higher in cells stably expressing the CD/UPRT/mDsRed fusion gene than in cells stably expressing CD/mDsRed alone or wild-type cells. Whole tumor (19)F MRS measurements showed rapid conversion of 5-FC to 5-FU within 20 min after 5-FC was administered intravenously in both CD/mDsRed and CD/UPRT/mDsRed tumors with subsequent anabolism to cytotoxic fluoronucleotides (FNucs). CD/UPRT/mDsRed tumor was more efficient in these processes. CONCLUSION: This study demonstrates the utility of these tumor models stably expressing CD or CD/UPRT to non-invasively evaluate the efficacy of the transgene expression/activity by monitoring drug metabolism in vivo using MRS, with potential applications in preclinical and clinical settings.


Subject(s)
Cytosine Deaminase/analysis , Luminescent Proteins/analysis , Magnetic Resonance Spectroscopy , Pentosyltransferases/analysis , Animals , Blotting, Western , Cell Line, Tumor , Flow Cytometry , Fluorine Radioisotopes , Genes, Reporter/physiology , Male , Mice , Mice, Nude , Prostatic Neoplasms , Rats , Sensitivity and Specificity , Transfection , Transgenes/physiology , Red Fluorescent Protein
7.
Nucleic Acids Res ; 35(9): 2955-64, 2007.
Article in English | MEDLINE | ID: mdl-17439959

ABSTRACT

Approximately 17% of the human genome is comprised of long interspersed nuclear element 1 (LINE-1, L1) non-LTR retrotransposons. L1 retrotransposition is known to be the cause of several genetic diseases, such as hemophilia A, Duchene muscular dystrophy, and so on. The L1 retroelements are also able to cause colon cancer, suggesting that L1 transposition could occur not only in germ cells, but also in somatic cells if innate immunity would not function appropriately. The mechanisms of L1 transposition restriction in the normal cells, however, are not fully defined. We here show that antiretroviral innate proteins, human APOBEC3 (hA3) family members, from hA3A to hA3H, differentially reduce the level of L1 retrotransposition that does not correlate either with antiviral activity against Vif-deficient HIV-1 and murine leukemia virus, or with patterns of subcellular localization. Importantly, hA3G protein inhibits L1 retrotransposition, in striking contrast to the recent reports. Inhibitory effect of hA3 family members on L1 transposition might not be due to deaminase activity, but due to novel mechanism(s). Thus, we conclude that all hA3 proteins act to differentially suppress uncontrolled transposition of L1 elements.


Subject(s)
Cytosine Deaminase/metabolism , Long Interspersed Nucleotide Elements , APOBEC Deaminases , Base Sequence , Cell Line , Cytidine Deaminase , Cytosine Deaminase/analysis , HeLa Cells , Humans , Molecular Sequence Data , RNA, Messenger/metabolism , Retroviridae/physiology , Reverse Transcription , Sequence Analysis, DNA
8.
Blood ; 110(1): 393-400, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17371941

ABSTRACT

HIV-1 recognition by, interaction with, and/or infection of CD4(+)CCR5(+) tissue macrophages and dendritic cells (DCs) play important roles in HIV-1 transmission and pathogenesis. By comparison, circulating CD4(+)CCR5(+) monocytes appear relatively resistant to HIV-1, and a fundamental unresolved question involves deciphering restriction factors unique to this precursor population. Not only do monocytes, relative to macrophages, possess higher levels of the innate resistance factor APOBEC3G, but we uncovered APOBEC3A, not previously associated with anti-HIV activity, as being critical in monocyte resistance. Inversely correlated with susceptibility, silencing of APOBEC3A renders monocytes vulnerable to HIV-1. Differences in promiscuity of monocytes, macrophages, and DCs can be defined, at least partly, by disparities in APOBEC expression, with implications for enhancing cellular defenses against HIV-1.


Subject(s)
Cell Differentiation , Cytosine Deaminase/immunology , Disease Susceptibility/immunology , HIV Infections/etiology , Immunity, Innate , Monocytes/immunology , Myeloid Cells/cytology , APOBEC Deaminases , Cytidine Deaminase , Cytosine Deaminase/analysis , Dendritic Cells/chemistry , HIV Infections/immunology , HIV-1 , Humans , Macrophages/chemistry , Monocytes/chemistry , Myeloid Cells/chemistry
9.
Gene Ther ; 13(2): 127-37, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16163381

ABSTRACT

A major limitation in cancer gene therapy, specifically gene-dependent enzyme prodrug therapy (GDEPT), is inefficient gene delivery and expression. The suicide gene cytosine deaminase (CD) and its substrate, 5-fluorocytosine (5-FC), have been extensively explored due to the inherent 'bystander' effect achieved through diffusion of the toxic metabolite 5-fluorouracil (5-FU). In this study, we aimed to enhance this 'bystander' effect by fusing the Saccharomyces cerevisiae CD to the HSV-1 tegument protein vp22, a novel translocating protein. Two constructs were created: one with vp22 fused to CD (vp22CD) and a second wherein a truncated vp22, lacking the necessary residues for trafficking, fused to CD (delvp22CD). The generated 9L stable lines exhibited similar growth rates, enzyme expression, CD activity, and sensitivity to 5-FC and 5-FU. However, mixed population colony formation assays demonstrated greater bystander effect with the vp22CD fusion as compared to delvp22CD. This enhancement was maintained in vivo where 9L tumors expressing 20 or 50% vp22CD exhibited increased growth delay compared to the respective delvp22CD tumors. Moreover, adenoviral transduction of established wild-type 9L tumors showed increased growth delay with vp22CD (Ad-EF_vp22CD) as compared to equivalent CD (Ad-EF_CD) transduced tumors. Finally, confirming the increased efficacy, (19)F magnetic resonance spectroscopy (MRS) of vp22CD-expressing tumors demonstrated increased 5-FU levels as compared to tumors expressing the nontranslocating CD. These results together demonstrated that fusion of vp22 to CD resulted in CD translocation, which in turn amplified conversion of 5-FC to 5-FU in vivo and enhanced the therapeutic benefit of this GDEPT strategy.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Bystander Effect , Cytosine Deaminase/genetics , Fluorouracil/therapeutic use , Genetic Therapy/methods , Viral Structural Proteins/genetics , Adenoviridae/genetics , Animals , Cell Line , Cytosine Deaminase/analysis , Cytosine Deaminase/metabolism , Flucytosine/therapeutic use , Gene Fusion , Genetic Engineering , Humans , Immunohistochemistry/methods , Magnetic Resonance Spectroscopy , Male , Mice , Mice, Nude , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Transduction, Genetic/methods , Translocation, Genetic
10.
Blood ; 107(6): 2470-3, 2006 Mar 15.
Article in English | MEDLINE | ID: mdl-16269615

ABSTRACT

Neoplastic transformation of mature B cells can be triggered by class-switch recombination of the immunoglobulin gene, which aberrantly targets a protooncogene and promotes translocation. Class-switch recombination is initiated by the B-cell-specific protein activation-induced cytidine deaminase (AID). Using immunohistochemistry with a newly generated monoclonal antibody and quantitative reverse-transcription-polymerase chain reaction (RT-PCR) on microdissected tissue from lymph node, tonsil, and thymus, we demonstrate that AID expression is found in secondary lymphoid organs outside germinal centers and in the thymic medulla at substantial levels. This is accompanied by the presence of circle transcripts, indicating class-switch recombination to be active at these sites. The dominant AID-expressing cell population outside germinal centers displays cytomorphologic properties corresponding to those that define the recently characterized interfollicular large B-cell subset. These findings indicate that interfollicular large B cells and AID-expressing B lymphocytes of the thymic medulla could give rise to mature B-cell malignancies.


Subject(s)
B-Lymphocytes/cytology , Cytosine Deaminase/analysis , Leukemia, B-Cell/pathology , Lymphoid Tissue/cytology , Lymphoma, B-Cell/pathology , Cell Transformation, Neoplastic/immunology , Cytidine Deaminase , Cytosine Deaminase/genetics , Humans , Immunoglobulin Class Switching , Immunohistochemistry , Leukemia, B-Cell/etiology , Lymphoma, B-Cell/etiology , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction
11.
Cancer Gene Ther ; 10(10): 737-44, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14502226

ABSTRACT

We performed a pilot trial in refractory cancer patients to investigate the feasibility of intratumoral injection of TAPET-CD, an attenuated Salmonella bacterium expressing the E. coli cytosine deaminase gene. A total of three patients received three dose levels of TAPET-CD (3 x 10(6)-3 x 10(7) CFU/m(2)) via intratumoral injection once every 28 days as long as progression of disease or intolerable toxicity was not observed. From days 4 to 14 of each 28 day cycle, patients also received 5-fluorocytosine (5-FC) at a dose of 100 mg/kg/day p.o. divided three times daily. Six cycles of treatment were administered. No significant adverse events clearly attributable to TAPET-CD were demonstrated. Two patients had intratumor evidence of bacterial colonization with TAPET-CD, which persisted for at least 15 days after initial injection. Conversion of 5-FC to 5-fluorouracil (5-FU) as a result of cytosine deaminase expression was demonstrated in these two patients. The tumor to plasma ratio of 5-FU for these two colonized patients was 3.0, demonstrating significantly increased levels of 5-FU at the site of TAPET-CD colonization and insignificant systemic spread of the bacteria. In contrast, the tumor to plasma ratio of 5-FU of the patient who did not show colonization of TAPET-CD was less than 1.0. These results support the principle that a Salmonella bacterium can be utilized as a delivery vehicle of the cytosine deaminase gene to malignant tissue and that the delivered gene is functional (i.e. able to convert 5-FC to 5-FU) at doses at or below 3 x 10(7) CFU/m(2).


Subject(s)
Cytosine Deaminase/genetics , Cytosine Deaminase/therapeutic use , Genetic Therapy/methods , Neoplasms/genetics , Neoplasms/therapy , Salmonella/genetics , Aged , Aged, 80 and over , Cytosine Deaminase/administration & dosage , Cytosine Deaminase/analysis , Escherichia coli/enzymology , Escherichia coli/genetics , Female , Flucytosine/analysis , Flucytosine/blood , Flucytosine/metabolism , Fluorouracil/analysis , Fluorouracil/blood , Fluorouracil/metabolism , Genetic Therapy/adverse effects , Head/pathology , Head and Neck Neoplasms/blood , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/therapy , Humans , Male , Middle Aged , Neck/pathology , Neoplasms/blood , Neoplasms/metabolism , Pilot Projects , Salmonella/physiology
SELECTION OF CITATIONS
SEARCH DETAIL