Reporter 8.5 Keygen [BETTER]
DOWNLOAD ::: https://urlca.com/2sXXnS
The InSpec json-file reporter is no longer enabled by default in Compliance Phase. Outputting compliance data to file by default potentially exposed sensitive data to the filesystem, without much upside. If you rely on this file for processing by external systems you can produce it by setting the reporter attribute node['audit']['reporter'] to %w{json-file cli}.
The compliance phase will now default to using both the json-file and the new cli reporter by default. This gives you a visual indication of the success of the Compliance Phase and is perfect for running both on the CLI and in Test Kitchen.
Effect of NPM1 mA on the NF-κB activity in THP-1 cells after drug treatment. (A) THP-1 cells expressing NPM1mA or pEGFPC1 were treated with DNR (0.25 μM, 24h). (B) THP-1 cells expressing NPM1mA or pEGFPC1 were treated with Ara-C (8.5 μM, 24h). NF-κB transcription activity was detected by luciferase reporter assays in the presence and absence of drug. Asterisks (*) indicate significant differences in relation to Blank or pEGFPC1 group.
TNF-α stimulation attenuated the effect of NPM1mA on THP-1 cells sensitive to drug. (A) THP-1 cells expressing NPM1 mA cells were treated with TNF-α (100 μM, 12h) and NF-κB transcription activity was detected by luciferase reporter assays. (B) THP-1 cells expressing NPM1 mA cells were treated with or without TNF-α (100 μM, 12h) following DNR (0.25 μM, 24h) incubation. (C) THP-1 cells expressing NPM1 mA cells were treated with or without TNF-α (100 μM, 12h) following Ara-C (8.5 μM, 24h) incubation. The apoptosis rate was assessed by flow cytometry using Annexin V-FITC. Asterisks (*) indicate significant differences in relation to control untreated cells.
We used two solid tumor cell lines (HCT-8 colorectal cancer cells and MCF-7 breast cancer cells) and one hematologic tumor cell line (K562 chronic myeloid leukemia cells), which are resistant to the chemotherapeutic drugs vincristine and adriamycin respectively, and two xenograft mice models, including the solid tumor model in nude mice with the resistant HCT-8 cells and the leukemia model in NOD/SCID mice with the resistant K562 cells to investigate the reversal effect of IVM on the resistance in vitro and in vivo. MTT assay was used to investigate the effect of IVM on cancer cells growth in vitro. Flow cytometry, immunohistochemistry, and immunofluorescence were performed to investigate the reversal effect of IVM in vivo. Western blotting, qPCR, luciferase reporter assay and ChIP assay were used to detect the molecular mechanism of the reversal effect. Octet RED96 system and Co-IP were used to determine the interactions between IVM and EGFR.
A 1, 637 bp region encompassing the NF-κB binding site and the annotated transcription start of ABCB1 (-1468 to +168 bp, chr7-: 87713155-87714791) was cloned into a Gaussia luciferase (GLuc) reporter vector (pEZX-PG04, Genecopoeia), which contains a reference reporter gene, secreted alkaline phosphatase (SeAP).
The cells in 24-well plates were co-transfected with the above reporter vector with pcDNA3.1(+)-P65 expression vector or siRNA targeting NF-κB using transfection reagent VigoFect (Vigorous Biotech). After 12 h, the cells were treated with 3 μM IVM and/or 25 nM VCR for 48 h. The activities of GLuc and SeAP were quantified with the secrete-pair dual luminescence assay kit (Genecopoeia).
Our previous results suggested that oroxylin A could inhibit p53 degradation by downregulating MDM2 expression instead of influencing p53 mRNA level, which was mediated by SIRT3. The result that oroxylin A increased p53 levels suggested that oroxylin A must act through negative feedback on MDM2 transcription through the P2 promoter in wt-p53 cancer cells. However, the expression of SIRT3 still influenced the level of MDM2 in p53-null cells (Figure 4B). This inferred the possibility that oroxylin A may function via a p53-independent mechanism to regulate MDM2 transcription. For this purpose, a genomic DNA fragment containing the P1 promoter regions of the MDM2 gene only was ligated to a luciferase reporter gene. A luciferase assay showed that oroxylin A inhibited the transcription of MDM2 in wt-p53 MCF-7 and HCT116 cells (Figure 5A) as well as in p53-null H1299 cells (Figure 5B).
In addition to the modification of p53, MDM2 can be modified post-transcriptionally to disrupt the p53-MDM2 interaction. Similar to p53, the activity of MDM2 can be regulated by acetylation [40]. Oroxylin A could not inhibit the binding of p53 with MDM2 (Figure 3D). However, the transcription of MDM2 was significantly suppressed by oroxylin A (Figures 2C and 3B). The human MDM2, HDM2, is controlled by two different promoters [41,42]. Transcription from the first promoter, P1, is independent of p53, whereas transcription from the second promoter, P2, is p53-dependent. As shown in Figure 4B, oroxylin A decreased the level of MDM2 in null-p53 cancer cells, suggesting that it modulates MDM2 levels in a p53-independent manner. Therefore, we constructed a luciferase reporter gene consisting of the P1 promoter DNA fragment of the MDM2 gene only and showed that oroxylin A could inhibit the transcriptional activity of MDM2 through a p53-independent mechanism (Figure 5A, B). In vivo, we showed that oroxylin A inhibited the tumor growth of wt-p53 MCF-7 and HCT116 cells (Figure 8). And the inhibitory rate of 100 mg/kg oroxylin A for mut-p53 MDA-MB-231 cells was 56.78% (data not shown) and 36.16% for HT29 cells [16]. These results suggested that oroxylin A had the presence of a p53-independent mechanism as well.
MicroRNAs (miRNAs) play a vital role in regulating gene expression and are associated with a variety of cancers, including breast cancer. Their distorted and unique expression is a potential marker in clinical diagnoses and prognoses. Thus, accurate determination of miRNA expression levels is a prerequisite for their applications. However, the assays currently available for miRNA detection typically require pre-enrichment, amplification and labeling steps, and most of the assays are only semi-quantitative. Therefore, we developed a quasi-direct liquid chromatography-tandem mass spectrometry (LC-MS/MS)-based targeted proteomics approach to quantify target miRNA by innovatively converting the miRNA signal into the mass response of a reporter peptide via a covalently immobilized DNA-peptide probe. Specifically, the probe containing the targeted proteomics-selected substrate/reporter peptide, GDRAVQLGVDPFR/AVQLGVDPFR, and the DNA sequence complementary to the target miRNA (i.e., miR-21) was first immobilized on APMTS modified silica nanoparticles using PDITC. After the immobilized probe was recognized and hybridized with the target miRNA, the excess probe was degraded using MBN and followed by a trypsin digestion of the hybrids. The reporter peptide was released and quantified using LC-MS/MS. The obtained LOQ was 5 pM. Finally, the developed assay was used for the quantitative analysis of miR-21 in breast cells and tissue samples.
In this study, we developed a quasi-direct targeted proteomics approach by covalently immobilizing a DNA-peptide probe on amino-modified silica nanoparticles (Fig. 1). A substrate peptide containing the reporter peptide and tryptic cleavage site was first designed using a targeted proteomics rationale and then conjugated with a DNA sequence complementary to the target miRNA. After the p-phenylene diisothiocyanate (PDITC) reaction, the newly formed DNA-peptide probe was immobilized on silica beads that were amino modified using (3-aminopropyl) trimethoxysilane (APTMS) in advance. This immobilized probe was then hybridized with the target miRNA (i.e., miR-21). After removing the excess probe using mung bean nuclease (MBN), the reporter peptide was liberated from the hybrids using trypsin and ultimately quantified using LC-MS/MS. The new assay for miR-21 was optimized for the parameters including conjugation, immobilization, hybridization and digestion. Finally, miR-21 was quantified in 3 breast cancer cell lines and 36 pairs of human breast primary tumors and adjacent normal tissue samples. The obtained data were compared with the quantitative reverse transcription PCR (qRT-PCR) results.
In our previous work, the criteria for the selection of suitable reporter and substrate peptides were extensively described38. While evidence indicated that our previously designed surrogate/reporter peptide was applicable to targeted proteomics analysis, these peptides may not be suitable for the present work because of the probe immobilization process.
In this study, we immobilized the DNA-peptide probe on amino-modified silica beads and developed a quasi-direct targeted proteomics assay for miRNA quantification. Using this assay, the target miR-21 was quantified in 3 breast cell lines and 36 pairs of breast tissue samples. The advantages of the quasi-direct targeted proteomics approach were demonstrated by converting the miRNA signal to the mass response of the reporter peptide. More importantly, immobilization of the DNA-peptide probe circumvents the miRNA biotinylation and subsequent dependence on the biotin-streptavidin interaction and allows the use of RNA samples without any further manipulation. Technically, this quasi-direct targeted proteomics method can be easily applied to other miRNAs by replacing the DNA sequence with one that is complementary to the target miRNA while keeping the reporter peptide the same. However, the parameters, including conjugation, immobilization, hybridization and digestion, deserve careful optimization to achieve the highest sensitivity and specificity for each miRNA. Furthermore, this assay has more potential for simultaneous detection of multiple miRNAs. Indeed, a key advantage of the LC-MS/MS-based quasi-targeted proteomics assay is its multiplexing ability, which is valid as long as a mass spectrometer can manage the concomitant analysis of multiple reporter peptides while retaining a degree of selectivity. However, the challenges of optimizing the assay format for each peptide, selecting a common dilution factor, addressing the variability and cross-interference, and establishing a robust quality control algorithm are substantial and require further analytical and statistical development. We anticipate that the quasi-targeted proteomics approach described here can ultimately be applied to the profiling of miRNAs in biological samples. This type of quasi-direct analysis may increase the quantitative accuracy and precision, but more evidence is required to confirm this feature. 2b1af7f3a8