Nothing Special   »   [go: up one dir, main page]

You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (6)

Search Parameters:
Keywords = thymidylate kinase gene

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
13 pages, 6011 KiB  
Article
Molecular Variation and Phylogeny of Thymidylate Kinase Genes of Candidatus Phytoplasma ziziphi from Different Resistant and Susceptible Jujube Cultivars in China
by Chuan-Sheng Song, Qi-Cong Xu, Cui-Ping Wan, De-Zhi Kong, Cai-Li Lin and Shao-Shuai Yu
Biology 2024, 13(11), 886; https://doi.org/10.3390/biology13110886 - 30 Oct 2024
Viewed by 839
Abstract
The thymidylate kinase (tmk) gene is indispensable for the proliferation and survival of phytoplasma. To reveal the molecular variation and phylogeny of the tmk genes of Candidatus phytoplasma ziziphi, in this study, the tmk genes of 50 phytoplasma strains infecting different [...] Read more.
The thymidylate kinase (tmk) gene is indispensable for the proliferation and survival of phytoplasma. To reveal the molecular variation and phylogeny of the tmk genes of Candidatus phytoplasma ziziphi, in this study, the tmk genes of 50 phytoplasma strains infecting different resistant and susceptible jujube cultivars from different regions in China were amplified and analyzed. Two sequence types, tmk-x and tmk-y, were identified using clone-based sequencing. The JWB phytoplasma strains were classified into three types, type-X, type-Y, and type-XY, based on the sequencing chromatograms of the tmk genes. The type-X and type-Y strains contained only tmk-x and tmk-y genes, respectively. The type-XY strain contained both tmk-x and tmk-y genes. The type-X, type-Y, and type-XY strains comprised 42%, 12%, and 46% of all the strains, respectively. The type-X and type-XY strains were identified in both susceptible and resistant jujube cultivars, while type-Y strain was only identified in susceptible cultivars. Phylogenetic analysis indicated that the tmk genes of the phytoplasmas were divided into two categories: phylo-S and phylo-M. The phylo-S tmk gene was single-copied in the genome, with an evolutionary pattern similar to the 16S rRNA gene; the phylo-M tmk gene was multi-copied, related to PMU-mediated within-genome transposition and between-genome transfer. Furthermore, the phylogenetic tree suggested that the tmk genes shuttled between the genomes of the Paulownia witches’ broom phytoplasma and JWB phytoplasma. These findings provide insights into the evolutionary and adaptive mechanisms of phytoplasmas. Full article
Show Figures

Figure 1

Figure 1
<p>Variation sites of the <span class="html-italic">tmk</span> genes and amino acids of the coding proteins of the JWB phytoplasma. The <span class="html-italic">tmk-x</span> and <span class="html-italic">tmk-y</span> gene sequences, along with the amino acid sequences of their encoded proteins TMK-x and TMK-y, were aligned, respectively, using DNAMAN 7.0 software. The nucleotide sequence is highlighted with a green background. The mutated nucleotides are highlighted with a red background. The amino acid sequences are not highlighted. The mutated amino acids are highlighted with a yellow background.</p>
Full article ">Figure 2
<p>Selective PCR amplification of the different types of the JWB Phytoplasma strains. The 15 strains were amplified by the PCR using the specific primer pair xtmkkf2/xtmkr for the <span class="html-italic">tmk-x</span> gene and xtmkf1/xtmkr for the <span class="html-italic">tmk-y</span> gene. The abbreviation of each strain are shown in <a href="#app1-biology-13-00886" class="html-app">Table S1</a>.</p>
Full article ">Figure 3
<p>Multiple sequence alignment and functional domain analysis of TMK from different phytoplasmas. A total of 15 TMK amino acid sequences were aligned using DNAMAN 7.0 software. P-loop, TMK binding motif, and LID were functional domains of TMK proteins. Different colors represent varying levels of homology. Red, green, yellow and pink represent 100%, ≥70%, ≥50% and ≥33% identity respectively.</p>
Full article ">Figure 4
<p>The type of phytoplasma strain is related to the resistance and geographic distribution of jujube cultivars. (<b>a</b>) The type data of the JWB phytoplasma strains in different regions and host varieties are visualized. The sampling sites of the infected cultivar and the types of JWB phytoplasma strains are shown in <a href="#app1-biology-13-00886" class="html-app">Table S1</a>. (<b>b</b>) The proportion of the different types of JWB phytoplasmas of eight provinces or municipalities. (<b>c</b>) The proportion of resistant and susceptible hosts, respectively, infected with JWB phytoplasma strains with type-X, type-Y and type-XY. The numbers on the columns with different colors in (<b>a</b>,<b>b</b>) represent the number of JWB phytoplasma strains with different types. The numbers on the columns with different colors in (<b>c</b>) represent the number of JWB phytoplasma strains infecting the resistant and susceptible jujube cultivars.</p>
Full article ">Figure 5
<p>The phylogenetic tree of the <span class="html-italic">tmk</span> gene in phytoplasmas. The analysis involved 191 nucleotide sequences, as shown in <a href="#app1-biology-13-00886" class="html-app">Table S2</a>. The evolutionary history was inferred using the neighbor-joining method. The percentages of the replicate trees in which the associated taxa clustered together in the bootstrap test (1000 replicates) are shown next to the branches. Black circles indicate the percentages of replicate trees greater than 90%, gray circles indicate percentages between 60 and 90%, and unmarked circles indicate percentages less than 60%.</p>
Full article ">
10 pages, 1100 KiB  
Communication
Rapid and Cost-Efficient Detection of RET Rearrangements in a Large Consecutive Series of Lung Carcinomas
by Vladislav I. Tiurin, Elena V. Preobrazhenskaya, Natalia V. Mitiushkina, Aleksandr A. Romanko, Aleksandra A. Anuskina, Rimma S. Mulkidjan, Evgeniya S. Saitova, Elena A. Krivosheyeva, Elena D. Kharitonova, Mikhail P. Shevyakov, Ilya A. Tryakin, Svetlana N. Aleksakhina, Aigul R. Venina, Tatiana N. Sokolova, Aleksandr S. Martianov, Anna D. Shestakova, Alexandr O. Ivantsov, Aglaya G. Iyevleva and Evgeny N. Imyanitov
Int. J. Mol. Sci. 2023, 24(13), 10530; https://doi.org/10.3390/ijms241310530 - 23 Jun 2023
Cited by 7 | Viewed by 1927
Abstract
RET-kinase-activating gene rearrangements occur in approximately 1–2% of non-small-cell lung carcinomas (NSCLCs). Their reliable detection requires next-generation sequencing (NGS), while conventional methods, such as immunohistochemistry (IHC), fluorescence in situ hybridization (FISH) or variant-specific PCR, have significant limitations. We developed an assay that [...] Read more.
RET-kinase-activating gene rearrangements occur in approximately 1–2% of non-small-cell lung carcinomas (NSCLCs). Their reliable detection requires next-generation sequencing (NGS), while conventional methods, such as immunohistochemistry (IHC), fluorescence in situ hybridization (FISH) or variant-specific PCR, have significant limitations. We developed an assay that compares the level of RNA transcripts corresponding to 5′- and 3′-end portions of the RET gene; this test relies on the fact that RET translocations result in the upregulation of the kinase domain of the gene and, therefore, the 5′/3′-end expression imbalance. The present study included 16,106 consecutive NSCLC patients, 14,449 (89.7%) of whom passed cDNA quality control. The 5′/3′-end unbalanced RET expression was observed in 184 (1.3%) tumors, 169 of which had a sufficient amount of material for the identification of translocation variants. Variant-specific PCR revealed RET rearrangements in 155/169 (91.7%) tumors. RNA quality was sufficient for RNA-based NGS in 10 cases, 8 of which carried exceptionally rare or novel (HOOK1::RET and ZC3H7A::RET) RET translocations. We also applied variant-specific PCR for eight common RET rearrangements in 4680 tumors, which emerged negative upon the 5′/3′-end unbalanced expression test; 33 (0.7%) of these NSCLCs showed RET fusion. While the combination of the analysis of 5′/3′-end RET expression imbalance and variant-specific PCR allowed identification of RET translocations in approximately 2% of consecutive NSCLCs, this estimate approached 120/2361 (5.1%) in EGFR/KRAS/ALK/ROS1/BRAF/MET-negative carcinomas. RET-rearranged tumors obtained from females, but not males, had a decreased level of expression of thymidylate synthase (p < 0.00001), which is a known predictive marker of the efficacy of pemetrexed. The results of our study provide a viable alternative for RET testing in facilities that do not have access to NGS due to cost or technical limitations. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Workflow for detection of <span class="html-italic">RET</span> rearrangements in a large series of NSCLCs.</p>
Full article ">Figure 2
<p><span class="html-italic">TYMS</span> RNA expression in RET-rearranged and wild-type NSCLCs. (<b>A</b>) The analysis of the entire cohort of tumors with (<span class="html-italic">n</span> = 88) and without (<span class="html-italic">n</span> = 234) <span class="html-italic">RET</span> fusions; (<b>B</b>,<b>C</b>) comparison of <span class="html-italic">TYMS</span> RNA expression in females (wild-type: <span class="html-italic">n</span> = 73; <span class="html-italic">RET</span>-associated: <span class="html-italic">n</span> = 60) and males (wild-type: <span class="html-italic">n</span> = 161; <span class="html-italic">RET</span>-associated: <span class="html-italic">n</span> = 28); (<b>D</b>,<b>E</b>) gender-related differences in <span class="html-italic">TYMS</span> expression in <span class="html-italic">RET</span>-rearranged NSCLCs (males: <span class="html-italic">n</span> = 28; females: <span class="html-italic">n</span> = 60) and tumors lacking common driver mutational events (males: <span class="html-italic">n</span> = 161; females: 73). <span class="html-italic">p</span>-values were calculated using the Mann–Whitney <span class="html-italic">U</span> test.</p>
Full article ">
15 pages, 3812 KiB  
Article
Phosphorylation of Thymidylate Synthase and Dihydrofolate Reductase in Cancer Cells and the Effect of CK2α Silencing
by Patrycja Wińska, Anna Sobiepanek, Katarzyna Pawlak, Monika Staniszewska and Joanna Cieśla
Int. J. Mol. Sci. 2023, 24(3), 3023; https://doi.org/10.3390/ijms24033023 - 3 Feb 2023
Cited by 1 | Viewed by 2453
Abstract
Our previous research suggests an important regulatory role of CK2-mediated phosphorylation of enzymes involved in the thymidylate biosynthesis cycle, i.e., thymidylate synthase (TS), dihydrofolate reductase (DHFR), and serine hydroxymethyltransferase (SHMT). The aim of this study was to show whether silencing of the CK2α [...] Read more.
Our previous research suggests an important regulatory role of CK2-mediated phosphorylation of enzymes involved in the thymidylate biosynthesis cycle, i.e., thymidylate synthase (TS), dihydrofolate reductase (DHFR), and serine hydroxymethyltransferase (SHMT). The aim of this study was to show whether silencing of the CK2α gene affects TS and DHFR expression in A-549 cells. Additionally, we attempted to identify the endogenous kinases that phosphorylate TS and DHFR in CCRF-CEM and A-549 cells. We used immunodetection, immunofluorescence/confocal analyses, reverse transcription–quantitative polymerase chain reaction (RT-qPCR), in-gel kinase assay, and mass spectrometry analysis. Our results demonstrate that silencing of the CK2α gene in lung adenocarcinoma cells significantly increases both TS and DHFR expression and affects their cellular distribution. Additionally, we show for the first time that both TS and DHFR are very likely phosphorylated by endogenous CK2 in two types of cancer cells, i.e., acute lymphoblastic leukaemia and lung adenocarcinoma. Moreover, our studies indicate that DHFR is phosphorylated intracellularly by CK2 to a greater extent in leukaemia cells than in lung adenocarcinoma cells. Interestingly, in-gel kinase assay results indicate that the CK2α’ isoform was more active than the CK2α subunit. Our results confirm the previous studies concerning the physiological relevance of CK2-mediated phosphorylation of TS and DHFR. Full article
(This article belongs to the Section Biochemistry)
Show Figures

Figure 1

Figure 1
<p>Thymidylate synthesis cycle. TS—thymidylate synthase; DHFR—dihydrofolate reductase; SHMT—serine hydroxymethyltransferase; dUMP—2′-deoxyuridine-5′-monophosphate; TMP, TDP, TTP—2′-deoxythymidine-5′-mono-, di-, and triphosphate, respectively; DHF—dihydrofolate; THF—tetrahydrofolate; meTHF—N5,10-methylenetetrahydrofolate.</p>
Full article ">Figure 2
<p>Immunodetection of CK2α, TS, and DHFR proteins in A-549 treated with 25 nM, 50 nM, and 100 nM <span class="html-italic">CSNK2A1</span> siRNA for 48 h and 72 h. (<b>a</b>) A representative Western blot. (<b>b</b>–<b>e</b>) Densitometry analysis data for the tested proteins in A-549 treated with <span class="html-italic">CSNK2A1</span> siRNA for 48 h (<b>b</b>), and for TS (<b>c</b>), DHFR (<b>d</b>), and CK2α (<b>e</b>) after 72 h of treatment with <span class="html-italic">CSNK2A1</span> siRNA. Tubulin was used as a loading control for each sample. <a href="#sec4-ijms-24-03023" class="html-sec">Section 4</a> describes the preparation of cell extracts and protein detection. Densitometry quantifications for each tested protein, given under each cell line panel, were calculated with untreated cells (CTRL) as the reference point. Graphs represent mean values ± s.e.m. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, relative to control; ns—not significant.</p>
Full article ">Figure 3
<p>The relative gene expression of thymidylate synthase (<span class="html-italic">TYMS</span>) and dihydrofolate reductase (DHFR) in A-549 treated with 50 nM and 100 nM <span class="html-italic">CSNK2A1</span> siRNA for 48 h (<b>a</b>) and with 25 nM, 50 nM, and 100 nM <span class="html-italic">CSNK2A1</span> siRNA for 72 h (<b>b</b>). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, relative to control; ns—not significant.</p>
Full article ">Figure 4
<p>The effect of silencing of <span class="html-italic">CSNK2A1</span> on localisation of DHFR (<b>e</b>) and TS (<b>d</b>) in A-549 cells. Detection of TS, DHFR and CK2α in control cells are shown in (<b>a</b>), (<b>b</b>), (<b>c</b>), respectively. The cells were treated with 100 nM <span class="html-italic">CSNK2A1</span> siRNA for 72 h (<b>d</b>–<b>f</b>), fixed, and after blocking, probed with the primary anti-DHFR or anti-TS antibodies. Subsequently, the cells were treated with Alexa Fluor 488-conjugated anti-mouse secondary antibody (green fluorescence) and Hoechst 33, 342 (nuclei, blue fluorescence).</p>
Full article ">Figure 5
<p>In-gel kinase assay results for TS (<b>a</b>) and DHFR (<b>c</b>). CCRF-CEM and A-549 lysates were prepared in NP-40 buffer (20 mM Tris-HCl pH 8.0; 137 mM NaCl; 1% Nonidet P-40; 2 mM EDTA). The left parts of (<b>a</b>) and (<b>c</b>) represent a fragment of gel stained with CBB with visible protein markers (M, PageRuler Prestained Protein Ladder, Thermo Scientific) and recombinant forms of CK2α and CK2α’. Human recombinant catalytic subunits of CK2, i.e., CK2α (29 µg) and CK2α’ (8 µg), were used as positive controls. Lysates containing proteins at three different levels were loaded in the range of 89–22 µg/well. Co-polymerisation of TS and DHFR was at acrylamide:bis-acrylamide ratio 37.5:1 and 19:1, respectively. Autoradiograms shown in (<b>a</b>,<b>b</b>), and (<b>c</b>,<b>d</b>) were obtained after 18 h and 6 h of X-ray films exposure, respectively.</p>
Full article ">Figure 6
<p>The molecular weight distribution of the proteins in the excised gel band against the Score describes the probability of matching experimental data with proteins in the database. The bubble size corresponds to emPAI value—the parameter dependent on the protein concentration. Most abundant proteins are indicated with numbers: 1. Actin cytoplasmic 1/2, 2. Fructose-bisphosphate aldolase A, 3. Phosphoglycerate kinase 1, 4. POTE ankyrin domain family member E, 5. Actin, alpha cardiac muscle.</p>
Full article ">
20 pages, 2839 KiB  
Article
Development of Robust Varicella Zoster Virus Luciferase Reporter Viruses for In Vivo Monitoring of Virus Growth and Its Antiviral Inhibition in Culture, Skin, and Humanized Mice
by Megan G. Lloyd, Michael B. Yee, Joseph S. Flot, Dongmei Liu, Brittany W. Geiler, Paul R. Kinchington and Jennifer F. Moffat
Viruses 2022, 14(4), 826; https://doi.org/10.3390/v14040826 - 15 Apr 2022
Cited by 6 | Viewed by 3459
Abstract
There is a continued need to understand varicella-zoster virus (VZV) pathogenesis and to develop more effective antivirals, as it causes chickenpox and zoster. As a human-restricted alphaherpesvirus, the use of human skin in culture and mice is critical in order to reveal the [...] Read more.
There is a continued need to understand varicella-zoster virus (VZV) pathogenesis and to develop more effective antivirals, as it causes chickenpox and zoster. As a human-restricted alphaherpesvirus, the use of human skin in culture and mice is critical in order to reveal the important VZV genes that are required for pathogenesis but that are not necessarily observed in the cell culture. We previously used VZV-expressing firefly luciferase (fLuc), under the control of the constitutively active SV40 promoter (VZV-BAC-Luc), to measure the VZV spread in the same sample. However, the fLuc expression was independent of viral gene expression and viral DNA replication programs. Here, we developed robust reporter VZV viruses by using bacterial artificial chromosome (BAC) technology, expressing luciferase from VZV-specific promoters. We also identified two spurious mutations in VZV-BAC that were corrected for maximum pathogenesis. VZV with fLuc driven by ORF57 showed superior growth in cells, human skin explants, and skin xenografts in mice. The ORF57-driven luciferase activity had a short half-life in the presence of foscarnet. This background was then used to investigate the roles for ORF36 (thymidine kinase (TK)) and ORF13 (thymidylate synthase (TS)) in skin. The studies reveal that VZV-∆TS had increased sensitivity to brivudine and was highly impaired for skin replication. This is the first report of a phenotype that is associated with the loss of TS. Full article
Show Figures

Figure 1

Figure 1
<p>Constructs for VZV reporter viruses. The diagram depicts the presence of the recombinant sequences used to alter pOka [<a href="#B14-viruses-14-00826" class="html-bibr">14</a>] with BAC recombineering, as detailed in the text. Viruses shown in (<b>A</b>) include parent Oka and VZV-BAC-Luc, derived from the BAC-Zhu pOka system [<a href="#B14-viruses-14-00826" class="html-bibr">14</a>,<a href="#B19-viruses-14-00826" class="html-bibr">19</a>]. VZV-BAC-Luc was generated from pOka (passage 9), as previously described [<a href="#B19-viruses-14-00826" class="html-bibr">19</a>], and included a small population of TK-virus, which arose through unknown mechanisms. (<b>B</b>)—upper section: viruses derived from an uncorrected BAC system, as detailed by the Osterreider group, which include VZV-ORF9-T2A-Luc; VZV-ORF14-T2A-Luc; and VZV-ORF17-T2A-Luc. (<b>B</b>)—lower section: diagram of the viral protein and luciferase expression mediated by the T2A ribosome skipping motif, which is used to generate two proteins from one mRNA. (<b>C</b>) VZV-ORF57-T2A-Luc [<a href="#B27-viruses-14-00826" class="html-bibr">27</a>], VZV-ORF57-∆TK, VZV-ORF57-∆TS, and VZV-ORF57-∆TK∆TS were generated in a BAC with corrected spurious mutations identified in ORF40 and ORF50 (Tischer VZV-BAC [<a href="#B18-viruses-14-00826" class="html-bibr">18</a>]), as detailed in the text. In VZV-ORF57-Luc, ORF13 (TS), ORF36 (TK), or both, were deleted through site-directed recombination events to replace the partial (ORF36) or entire (ORF13) ORFs with either the mturq2blue (mT2B) fluorescent reporter gene or a kanamycin resistance cassette. The colors used for each virus are carried through the rest of the figures for continuity.</p>
Full article ">Figure 2
<p>Evaluation of VZV-BAC-Luc and VZV-ORFx-Luc growth kinetics and bioluminescence in tissue culture and human fetal skin. Cells and skin were infected with cell-free or cell-associated virus, respectively, and grown at 37 °C. Cell culture experiments (<b>A</b>,<b>C</b>,<b>D</b>) were performed in HFFs, while skin organ culture (<b>B</b>) was performed in fetal skin. (<b>A</b>,<b>B</b>) VZV yield was measured by bioluminescence imaging and calculated as the fold change from the average Total Flux (photons/sec/cm<sup>2</sup>/steradian), divided by the lowest Total Flux value (DPI 1 for cells, or DPI 1-3 for SOC). (<b>C</b>) Correlation coefficients of luciferase and virus plaque number were determined for each VZV reporter virus on the basis of the relationship of pfu/well to average Total Flux per well. (<b>D</b>) HFFs and VZV were co-cultured for approximately 40 h prior to foscarnet treatment (1 mM) to block viral DNA replication. Values next to each curve represent the time (in h) for bioluminescence to decrease by 50% after treatment started and are shown in the corresponding color for each virus (individual points omitted for clarity of graph). Each point represents the mean ± SEM. Statistical analyses included one-way ANOVA with Dunnett’s post hoc test (<b>A</b>,<b>B</b>, *** <span class="html-italic">p</span> &lt; 0.001; **** <span class="html-italic">p</span> &lt; 0.0001). Nonlinear regression analysis with (<b>C</b>) log–log line or (<b>D</b>) dose response—inhibition was used for best-fit lines. <span class="html-italic">n</span> = 3 biological replicates for cell-based assays; <span class="html-italic">n</span> = 6 biological replicates for skin organ culture.</p>
Full article ">Figure 3
<p>Evaluation of VZV-BAC-Luc and VZV-ORF57-Luc growth kinetics and responses to antiviral treatment in a SCIDhu mouse model. SCID <span class="html-italic">beige</span> mice were implanted with a single subcutaneous xenograft of fetal human skin. Xenografts were inoculated 3–4 weeks later with (<b>A</b>) VZV-BAC-Luc or (<b>B</b>) VZV-ORF57-Luc by intra-xenograft injection (1 × 10<sup>4</sup>–10<sup>5</sup> pfu/mL; 60 µL injection; grown in HFFs). Mice were treated with vehicle (water) or 10 mg/kg/day cidofovir (CDV) by intraperitoneal injection from DPI 3 to 9 (Tx Phase). VZV yield was measured daily by bioluminescence imaging and the fold change was calculated as Total Flux each day divided by the lowest Total Flux value per mouse (usually taken on DPI 2 or 3). Virus growth kinetics were evaluated for statistical significance on DPI 9. Symbols represent mean ± SEM. Student’s <span class="html-italic">t</span>-test; asterisks indicate significance between vehicle and cidofovir groups (* <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01). <span class="html-italic">n</span> = 6–10 mice per group.</p>
Full article ">Figure 4
<p>VZV TK is dispensable while VZV TS is required for optimal growth in cells and is critical for human skin organ culture. Cell-free VZV was added to ARPE-19 monolayers at 37 °C (<b>A</b>–<b>C</b>; 1:50 ratio) or introduced to adult human skin (<b>D</b>; 1 × 10<sup>4</sup>–10<sup>5</sup> pfu/mL; 30 µL inoculum) by scarification, placed on NetWells, and incubated at 35 °C. Virus spread was monitored daily by bioluminescence imaging. Cell cultures were infected with up to 500 pfu/well. Skin was infected with 300–3000 pfu/piece of skin, depending on the viral titer that could be attained in ARPE-19 cells. VZV-ORF57-Luc was grown independently in ARPE-19 for each assay in (<b>A</b>–<b>C</b>), and directly compared to (<b>A</b>) VZV-ORF57-∆TK, (<b>B</b>) VZV-ORF57-∆TS, or (<b>C</b>) VZV-ORF57-∆TK∆TS under the same conditions. (<b>D</b>) Growth kinetics of ∆TK, ∆TS, and ∆TK∆TS in adult human skin explants were evaluated for significance compared to parental VZV-ORF57-Luc on DPI 7. VZV yield was calculated as the average Total Flux each day divided by the Total Flux on (<b>A</b>–<b>C</b>) DPI 1 or (<b>D</b>) DPI 2. Each point and line represent the mean ± SEM. (<b>A</b>–<b>C</b>) Student’s <span class="html-italic">t</span>-test or (<b>D</b>) one-way ANOVA with Dunnett’s post hoc test; * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01, ns = not significant. <span class="html-italic">n</span> = 6 biological replicates.</p>
Full article ">Figure 5
<p>Loss of VZV TK and TS affect virus sensitivity to select antivirals in cell culture. Cell-free VZV was added to ARPE-19 monolayers at 37 °C and compounds were added at 2 h postinoculation (HPI). Antiviral compounds remained in culture medium until VZV yield was measured on DPI 3. Infected cells were treated with cidofovir (CDV) and brivudine (BVdU) (<b>A</b>) in a range of concentrations, or (<b>B</b>) at specific concentrations of BVdU (1.25, 2.5, or 5 nM). VZV yield was calculated as the average Total Flux of treated wells divided by the average Total Flux of untreated wells. (<b>A</b>) Each point represents the mean Total Flux with the best fit line. (<b>B</b>) Each bar represents the mean + SD. Asterisks indicate significance between VZV viruses for each treatment condition (ns = no significance; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001; **** <span class="html-italic">p</span> &lt; 0.0001). <span class="html-italic">p</span> &lt; 0.05; one-way ANOVA; Tukey post hoc test. <span class="html-italic">n</span> = 3 (<b>A</b>) to 6 (<b>B</b>) biological replicates.</p>
Full article ">
22 pages, 3910 KiB  
Article
SAHA Overcomes 5-FU Resistance in IFIT2-Depleted Oral Squamous Cell Carcinoma Cells
by Prabha Regmi, Kuo-Chu Lai, Chung-Ji Liu and Te-Chang Lee
Cancers 2020, 12(12), 3527; https://doi.org/10.3390/cancers12123527 - 26 Nov 2020
Cited by 5 | Viewed by 3346
Abstract
Interferon-induced protein with tetratricopeptide repeats 2 (IFIT2) is a member of the interferon-stimulated gene family that contains tetratricopeptide repeats (TPRs), which mediate protein–protein interactions in various biological systems. We previously showed the depletion of IFIT2 enhanced cell migration and metastatic activity in oral [...] Read more.
Interferon-induced protein with tetratricopeptide repeats 2 (IFIT2) is a member of the interferon-stimulated gene family that contains tetratricopeptide repeats (TPRs), which mediate protein–protein interactions in various biological systems. We previously showed the depletion of IFIT2 enhanced cell migration and metastatic activity in oral squamous cell carcinoma (OSCC) cells via the activation of atypical PKC signaling. In this study, we found that IFIT2-knockdown cells displayed higher resistance to 5-fluorouracil (5-FU) than control cells. The comet assay and annexin V analysis showed decreased DNA damage and cell death in IFIT2-knockdown cells compared to control cells treated with 5-FU. Cell cycle progression was also perturbed by 5-FU treatment, with the accumulation of IFIT2-depleted cells in S phase in a time-dependent manner. We further observed the overexpression of thymidylate synthase (TS) and thymidine kinase (TK) in IFIT2-knockdown cells. Inhibition of TS alone or double inhibition of TS and TK1 using the siRNA technique increased susceptibility to 5-FU in IFIT2-knockdown cells. We further identified that suberanilohydroxamic acid (SAHA) treatment decreased the expression of TS in IFIT2-knockdown cells and demonstrated that pretreatment with SAHA sensitized IFIT2-knockdown cells to 5-FU in vitro and in vivo. In conclusion, IFIT2 knockdown enhances TS expression, which mediates 5-FU resistance, and SAHA pretreatment suppresses TS expression and hence sensitizes cells to 5-FU. SAHA will be an effective strategy for the treatment of OSCC patients with 5-FU resistance. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>DNA damage in IFIT2-knockdown cells after 5-FU treatment. (<b>A</b>) IFIT2 expression in IFIT2-knockdown cells. The IFIT2 protein levels in sh-control, sh-IFIT2-1 and sh-IFIT2-2 cells were determined by Western blotting. GAPDH was used as a loading control. (<b>B</b>) Comet assay. Quantitative data from the comet assay. Data are presented as the mean ± SEM of three independent experiments. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 compared with sh-control cells. (<b>C</b>) Induction of γH2AX. Cells were treated with 5-FU for 12, 24, 48 and 72 h. The appearance of γH2AX was determined by Western blotting. The right panel is the quantitative data of the of relative γH2AX expression from three independent experiments.</p>
Full article ">Figure 2
<p>Cell cycle interference by 5-FU. (<b>A</b>) Cell cycle analysis. Growing sh-control, sh-IFIT2-1 and sh-IFIT2-2 cells were treated with 10 µM 5-FU for 24, 48 and 72 h. The cells were harvested, and the cell cycle phase was analyzed by flow cytometry as described in the Materials and Methods. The symbols *, ** and *** denote statistically significant differences compared to sh-control at <span class="html-italic">p</span> &lt; 0.05, <span class="html-italic">p</span> &lt; 0.01 and <span class="html-italic">p</span> &lt; 0.001, respectively. (<b>B</b>) Increased basal levels of pChk1-S317 in IFIT2-knockdown cells. The lysates of sh-control, sh-IFIT2-1 and sh-IFIT2-2 cells were subjected to Western blot analysis using antibodies against Chk1 and pChK1-S317, respectively. The bar values in the right panel is the quantitative SEM values of relative expression of pCHK1.</p>
Full article ">Figure 3
<p>Apoptotic death triggered by 5-FU. (<b>A</b>) Quantitative data analysis of apoptotic cell death. Data are presented as the mean ± SEM of three independent experiments. * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 compared with sh-control cells. (<b>B</b>) Expression of the prosurvival protein cIAP1, the protease caspase 9 and cleaved PARP after 5-FU treatment. The cells were treated with 5-FU for 24, 48 and 72 h. The protein levels were determined by Western blotting. GAPDH was used as a loading control. The bar graphs below depict the quantitative densitometry analysis of Western blot data from three independent experiments. The symbols *, ** and *** denote statistically significant differences compared to sh-control at 0 h at <span class="html-italic">p</span> &lt; 0.05, <span class="html-italic">p</span> &lt; 0.01 and <span class="html-italic">p</span> &lt; 0.001, respectively.</p>
Full article ">Figure 4
<p>Protein and mRNA levels of 5-FU metabolic enzymes. (<b>A</b>) Protein levels. The whole cell lysates of sh-control, sh-IFIT2-1 and sh-IFIT2-1 cells were immunoblotted for TS, TK1, OPRT and TP. Bar graph indicates quantitative densitometry of Western blot analysis of three independent experiments relative to sh-control. The symbols ** and *** denote statistically significant differences compared to sh-control at <span class="html-italic">p</span> &lt; 0.01 and <span class="html-italic">p</span> &lt; 0.001, respectively. (<b>B</b>) mRNA levels. The relative mRNA expression levels of 5-FU metabolic enzymes in sh-control, sh-FIT2-1 and sh-IFIT2-2 cells were determined by Q-PCR. GAPDH mRNA was used as an internal control. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 denotes a statistically significant difference compared to sh-control. (<b>C</b>) Immunostaining. The intracellular distribution of TS in sh-control, sh-FIT2-1 and sh-IFIT2-2 cells with or without 5-FU treatment (10 μM for 48 h) was determined by immunostaining (red). The nuclei were counterstained with DAPI (blue). Quantitative analysis of the mean fluorescence intensity is shown in bar graph. The symbols ** and *** denote statistically significant differences compared to non-treated sh-control at <span class="html-italic">p</span> &lt; 0.01 and <span class="html-italic">p</span> &lt; 0.001, respectively. (<b>D</b>) Formation of the TS/5-FU complex. As described above, the cells were treated with 10 μM 5-FU for various time periods. The upper band (red arrow) denotes the TS/5-FU complex, and the lower band (black arrow) represents free TS. GAPDH was used as a loading control. The numbers represent the relative expression level of Free TS protein in compared to control cells. Quantitative densitometry of Western blot analysis of three independent experiments. *** <span class="html-italic">p</span> &lt; 0.001 denotes a statistically significant difference compared to sh-control at 0 h.</p>
Full article ">Figure 4 Cont.
<p>Protein and mRNA levels of 5-FU metabolic enzymes. (<b>A</b>) Protein levels. The whole cell lysates of sh-control, sh-IFIT2-1 and sh-IFIT2-1 cells were immunoblotted for TS, TK1, OPRT and TP. Bar graph indicates quantitative densitometry of Western blot analysis of three independent experiments relative to sh-control. The symbols ** and *** denote statistically significant differences compared to sh-control at <span class="html-italic">p</span> &lt; 0.01 and <span class="html-italic">p</span> &lt; 0.001, respectively. (<b>B</b>) mRNA levels. The relative mRNA expression levels of 5-FU metabolic enzymes in sh-control, sh-FIT2-1 and sh-IFIT2-2 cells were determined by Q-PCR. GAPDH mRNA was used as an internal control. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 denotes a statistically significant difference compared to sh-control. (<b>C</b>) Immunostaining. The intracellular distribution of TS in sh-control, sh-FIT2-1 and sh-IFIT2-2 cells with or without 5-FU treatment (10 μM for 48 h) was determined by immunostaining (red). The nuclei were counterstained with DAPI (blue). Quantitative analysis of the mean fluorescence intensity is shown in bar graph. The symbols ** and *** denote statistically significant differences compared to non-treated sh-control at <span class="html-italic">p</span> &lt; 0.01 and <span class="html-italic">p</span> &lt; 0.001, respectively. (<b>D</b>) Formation of the TS/5-FU complex. As described above, the cells were treated with 10 μM 5-FU for various time periods. The upper band (red arrow) denotes the TS/5-FU complex, and the lower band (black arrow) represents free TS. GAPDH was used as a loading control. The numbers represent the relative expression level of Free TS protein in compared to control cells. Quantitative densitometry of Western blot analysis of three independent experiments. *** <span class="html-italic">p</span> &lt; 0.001 denotes a statistically significant difference compared to sh-control at 0 h.</p>
Full article ">Figure 4 Cont.
<p>Protein and mRNA levels of 5-FU metabolic enzymes. (<b>A</b>) Protein levels. The whole cell lysates of sh-control, sh-IFIT2-1 and sh-IFIT2-1 cells were immunoblotted for TS, TK1, OPRT and TP. Bar graph indicates quantitative densitometry of Western blot analysis of three independent experiments relative to sh-control. The symbols ** and *** denote statistically significant differences compared to sh-control at <span class="html-italic">p</span> &lt; 0.01 and <span class="html-italic">p</span> &lt; 0.001, respectively. (<b>B</b>) mRNA levels. The relative mRNA expression levels of 5-FU metabolic enzymes in sh-control, sh-FIT2-1 and sh-IFIT2-2 cells were determined by Q-PCR. GAPDH mRNA was used as an internal control. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 denotes a statistically significant difference compared to sh-control. (<b>C</b>) Immunostaining. The intracellular distribution of TS in sh-control, sh-FIT2-1 and sh-IFIT2-2 cells with or without 5-FU treatment (10 μM for 48 h) was determined by immunostaining (red). The nuclei were counterstained with DAPI (blue). Quantitative analysis of the mean fluorescence intensity is shown in bar graph. The symbols ** and *** denote statistically significant differences compared to non-treated sh-control at <span class="html-italic">p</span> &lt; 0.01 and <span class="html-italic">p</span> &lt; 0.001, respectively. (<b>D</b>) Formation of the TS/5-FU complex. As described above, the cells were treated with 10 μM 5-FU for various time periods. The upper band (red arrow) denotes the TS/5-FU complex, and the lower band (black arrow) represents free TS. GAPDH was used as a loading control. The numbers represent the relative expression level of Free TS protein in compared to control cells. Quantitative densitometry of Western blot analysis of three independent experiments. *** <span class="html-italic">p</span> &lt; 0.001 denotes a statistically significant difference compared to sh-control at 0 h.</p>
Full article ">Figure 5
<p>Silencing TS expression sensitizes IFIT2-knockdown cells to 5-FU. (<b>A</b>) Silencing TS or TK1 by the siRNA technique. The specific siRNAs for TS and TK1 were transfected into cells either individually or in combination. TS and TK1 expression were confirmed after siRNA transfection in sh-IFIT2-1 cells for 48 h and replating in fresh medium for 72 h. The knockdown efficiency of siRNA transfection measured by detecting the protein levels of TS and TK1 by Western blotting. The quantitative densitometry of Western blot analysis of two independent experiment is shown in the graph. (<b>B</b>) IFIT2-knockdown cells were sensitized to 5-FU by silencing TS expression. Transfected siRNAs were seeded into 96-well plates at 3000 cells per well. The cells were treated with 5-FU in a serial dilution from 100 µM for 72 h. At the end of treatment, an aliquot of the Presto blue solution was added, and the fluorescence intensity was measured. IC<sub>50</sub> values were calculated using CompuSyn software. The symbols *, ** and *** denote statistically significant differences compared to control siRNA at <span class="html-italic">p</span> &lt; 0.05, <span class="html-italic">p</span> &lt; 0.01 and <span class="html-italic">p</span> &lt; 0.001, respectively.</p>
Full article ">Figure 6
<p>Sensitizing cells to 5-FU with SAHA. (<b>A</b>) Reduced expression of TS by SAHA. The cells were treated with SAHA at concentrations of 0, 0.375 and 0.75 μM for 24 h. The protein levels of TS were determined by Western blotting. Bar graph is the quantitative densitometry of Western blot analysis of three independent experiments. The symbols *, ** and *** denote statistically significant differences compared to non-treated cells at <span class="html-italic">p</span> &lt; 0.05, <span class="html-italic">p</span> &lt; 0.01 and <span class="html-italic">p</span> &lt; 0.001, respectively (<b>B</b>–<b>D</b>). Synergistic cytotoxicity of SAHA and 5-FU. A combination cytotoxicity assay of 5-FU and SAHA was performed at a ratio of 33:1 in sh-IFIT2-1 cells, sh-IFIT2-2 cells and 10:1 in sh-control cells. The CI shown in parentheses was calculated using CompuSyn software, where CI values denote the following: CI = 1, additive effects; CI &lt; 1, synergism; and CI &gt; 1, antagonism.</p>
Full article ">Figure 7
<p>Synergistic inhibition of SAHA and 5-FU in a xenograft tumor model. (<b>A</b>–<b>C</b>) An aliquot of 10<sup>7</sup> cells (sh-IFIT2-1, sh-IFIT2-2 and sh-control) was subcutaneously implanted into nude mice. When the tumor reached approximately 100 mm<sup>3</sup>, the mice were randomly divided into four groups (<span class="html-italic">n</span> = 4–5 for each group) and treated with vehicle (<span style="color:#000099">●</span>), SAHA (<span style="color:#7030A0">▲</span>, 80 mg/kg orally), 5-FU (<span style="color:#538135">▲</span>, 100 m/kg i.p.) or a combination of the two drugs (<span style="color:red">▼</span>). The tumor volume (left) and body weight change (right) of the mice were recorded every other day. The values are shown as the mean ± SD of each group.</p>
Full article ">
12 pages, 2313 KiB  
Article
A Nymphalid-Infecting Group I Alphabaculovirus Isolated from the Major Passion Fruit Caterpillar Pest Dione juno juno (Lepidoptera: Nymphalidae)
by Bergmann Morais Ribeiro, Ethiane Rozo dos Santos, Luana Beló Trentin, Leonardo Assis da Silva, Fernando Lucas de Melo, Elliot Watanabe Kitajima and Daniel M. P. Ardisson-Araújo
Viruses 2019, 11(7), 602; https://doi.org/10.3390/v11070602 - 3 Jul 2019
Cited by 6 | Viewed by 4080
Abstract
Baculoviruses are capable of infecting a wide diversity of insect pests. In the 1990s, the Dione juno nucleopolyhedrovirus (DijuNPV) was isolated from larvae of the major passionfruit defoliator pest Dione juno juno (Nymphalidae) and described at ultrastructural and pathological levels. In this study, [...] Read more.
Baculoviruses are capable of infecting a wide diversity of insect pests. In the 1990s, the Dione juno nucleopolyhedrovirus (DijuNPV) was isolated from larvae of the major passionfruit defoliator pest Dione juno juno (Nymphalidae) and described at ultrastructural and pathological levels. In this study, the complete genome sequence of DijuNPV was determined and analyzed. The circular genome presents 122,075 bp with a G + C content of 50.9%. DijuNPV is the first alphabaculovirus completely sequenced that was isolated from a nymphalid host and may represent a divergent species. It appeared closely related to Orgyia pseudotsugata multiple nucleopolyhedrovirus (OpMNPV) and other Choristoneura-isolated group I alphabaculoviruses. We annotated 153 open reading frames (ORFs), including a set of 38 core genes, 26 ORFs identified as present in lepidopteran baculoviruses, 17 ORFs unique in baculovirus, and several auxiliary genes (e.g., bro, cathepsin, chitinase, iap-1, iap-2, and thymidylate kinase). The thymidylate kinase (tmk) gene was present fused to a dUTPase (dut) gene in other baculovirus genomes. DijuNPV likely lost the dut portion together with the iap-3 homolog. Overall, the genome sequencing of novel alphabaculoviruses enables a wide understanding of baculovirus evolution. Full article
(This article belongs to the Special Issue Emerging Viruses: Surveillance, Prevention, Evolution and Control)
Show Figures

Figure 1

Figure 1
<p>Ultrastructural analysis of the <span class="html-italic">Dione juno</span> nucleopolyhedrovirus (DijuNPV) occlusion bodies (OBs). (<b>a</b>) Passionfruit caterpillars were fed on contaminated passionfruit leaves, and when the first symptoms of infection appeared, the insects were frozen and dissected for fat body extraction. The tissue was fixed and prepared for transmission electron microscopy. A section of a single cell inset on the nucleus is presented here. The nuclear envelope (NE) surrounds several polyhedral OBs. The virogenic stroma (VS) presents non-enveloped rod-shaped nucleocapsids (white arrowhead). (<b>b</b>) Cross section of a purified DijuNPV OB showing several occlusion-derived viruses containing multiple nucleocapsids.</p>
Full article ">Figure 2
<p>DijuNPV is an alphabaculovirus. Maximum likelihood inference based on the concatenated amino acid sequences of 38 core genes of all complete baculovirus genomes (<a href="#app1-viruses-11-00602" class="html-app">Table S1</a>). The branch support was determined by the Shimodaira-Hasegawa-like method. Some branches were collapsed for clarity: members inside genus <span class="html-italic">Gammabaculovirus</span> (orange), members inside genus <span class="html-italic">Betabaculovirus</span> (pink), and members inside genus <span class="html-italic">Alphabaculovirus</span> (dark blue). The deltabaculovirus CuniNPV was used as the root (light blue). We collapsed gammabaculovirus, betabaculovirus, some alphabaculovirus, and the Autographa californica multiple nucleopolyhedrovirus (AcMNPV)-like viruses.</p>
Full article ">Figure 3
<p>Genome comparison of DijuNPV and its related viruses. DijuNPV is compared to three alphabaculovirus genomes, including Hyphantria cunea nucleopolyhedrovirus (HycuNPV), Orgyia pseudotsugata multiple nucleopolyhedrovirus (OpMNPV), and Choristoneura fumiferana multiple nucleopolyhedrovirus (CfMNPV). The same colors depict the same LCBs across the genomes. Seven Locally Collinear Blocks (LCB) numbered from 1 to 7 were found. The DijuNPV genome lost LCB4 (purple) and present LCB7 (blue) instead. The white regions depict sequence loss.</p>
Full article ">Figure 4
<p>Venn diagram comparing the gene content among DijuNPV and some selected relatives. Total number of genes of the five alphabaculovirus (DijuNPV, AnpeNPV-L2, CfMNPV, OpMNPV, and HycuNPV) were compared by BLASTX to find homologies among the genes of the different genomes. A total of 251 genes were found, 25 were present only in the DijuNPV genome (eight are found in other baculovirus, and 17 are unique to DijuNPV); 117 are shared between the five viruses and only one single gene is found between DijuNPV and OpMNPV, the <span class="html-italic">tmk</span> homolog, which in the OpMNPV genome is found fused to the <span class="html-italic">dut</span>.</p>
Full article ">Figure 5
<p>Syntenic genomic context chart containing both <span class="html-italic">iap3</span> and <span class="html-italic">tmk</span> orthologs in DijuNPV-related species. We show the genomic context together with the phylogeny among the species. The arrowhead shape represents gene orientation, and similar colors represent ortholog genes. Autapomorphies are colored black, and dashed lines concatenate the genes.</p>
Full article ">Figure 6
<p>Phylogeny of a putative thymidylate kinase homolog found in the DijuNPV genome. Phylogeny of DijuNPV-ORF-126, a homolog of cp016-like. Erinnyis ello granulovirus (ErelGV), OpMNPV, Dasychira pudibunda nucleopolyhedrovirus (DapuNPV), Perigonia lusca single nucleopolyhedrovirus (PeluSNPV), Catopsilia pomona nucleopolyhedrovirus (CapoNPV), and DijuNPV-derived proteins clustered together, indicating common ancestry. The tree was rooted at the midpoint for clarity.</p>
Full article ">
Back to TopTop