The largest database of trusted experimental protocols
> Chemicals & Drugs > Amino Acid > DNMT1 protein, human

DNMT1 protein, human

DNMT1 is a DNA methyltransferase enzyme that plays a crucial role in the maintenance of DNA methylation patterns during cell division.
It is responsible for copying methylation marks from the parental DNA strand to the newly synthesized daughter strand, ensuring the preservation of epigenetic information.
DNMT1 is essential for normal development, genomic imprinting, and the regulation of gene expression.
Disruption of DNMT1 function has been implicated in various diseases, including cancer, neurological disorders, and immune system dysfunctions.
Researchers can leverage PubComapre.ai, an AI-driven platform, to optimize their DNMT1 protein research by locating relevant protocols from literature, preprints, and patents, and using AI-driven comparisons to identify the best protocols and products for enhanced reproducibility and accuracy.

Most cited protocols related to «DNMT1 protein, human»

The Integrated Neuroendocrine Prostate Cancer (NEPC) score estimates the likelihood of a test sample to be CRPC-NE. It is calculated as the Pearson's correlation coefficient between the test vector and a reference CRPC-NE vector based on a set of 70 genes (Supplementary Table 9, Supplementary Fig. 10 and 15) using normalized FPKM values of the test sample. The gene set stems from the integration of differentially deleted/amplified and/or expressed and/or methylated genes in CRPC-NE and CRPC-Adeno. Specifically, 16 differentially deleted genes were selected among putative cancer genes31 (link) (see Differential copy number analysis). The following strategy was used to identify both differentially expressed genes that better distinguish CRPC-NE and CRPC-Adeno samples. We selected differentially expressed protein coding genes with FDR ≤ 1e-2, resulting in a total of 2425 genes, corresponding to 1301 over- and 1124 under-expressed. For each gene, we performed a Receiver Operator Curve (ROC) analysis using the normalized FPKMs as threshold parameter and calculated the Area Under the Curve (AUC). ROCs were built by considering only samples sequenced excluding two samples (7520 and 4240) that were previously published9 (link).leaving 34 CRPC-Adeno and 13 CRPC-NE. Only those differentially expressed genes with AUC ≥ 0.95 and with a fold-change greater than 2 or lower than 0.5 were included in the classifier, resulting in a list of 49 genes (25 over- and 24 under- expressed in CRPC-NE vs. CRPC-Adeno), 21 of which found as differentially methylated between CRPC-NE and CRPC-Adeno. Concordant information between RNA and Methylation was found for 11 genes (see Supplementary Table 9). In addition, we considered 2 genes (MYCN and AURKA) that we previously described as associated with CRPC-NE phenotype9 (link), EZH2 (FDR = 7.9*10−4) and DNMT1 (FDR = 6.9*10−5) for their role in controlling DNA methylation70 (link) and RB1 (FDR = 0.056), reported as a key driver in the pathogenesis of CRPC-NE9 (link),45 (link). For each of the resulting 70 genes, we calculated the mean of the normalized FPKM across the 13 CRPC-NE samples with RNA-seq data and defined the resulting set of averages as reference CRPC-NE vector. The Integrated NEPC score was tested across 719 prostate samples with available transcriptome data from multiple datasets (Supplementary Table 10). RNA-seq data were processed as described above. Processed SU2C-PCF26 (link) and Grasso et al21 (link) (Michigan 2012) data were downloaded from cBioPortal71 (link). Since data for 4 genes (ARHGAP8, BRINP1, C7Orf76 and MAP10) were not available from cBioPortal, for Michigan 2012 we used a reduced version of Integrated NEPC Score (indicated as Integrated NEPC Score*). Samples with Integrated NEPC Score greater than or equal to 0.40 (elevated Integrated NEPC score in main text) were nominated as putative CRPC-NE (Figure 4c, Supplementary Table 14). In order to take into account the lower signal-to-noise ratio and the reduced version of Integrated NEPC Score in Michigan 2012 microarray data, in Figure 4d we consider as CRPC-NE – like those samples with Integrated NEPC Score ≥ 0.25 (significant Integrated NEPC score in Figure 4 legend). AR signaling and Integrated NEPC Score values per sample are reported in Supplementary Table 15.
Publication 2016
Aurora Kinase A Cloning Vectors DNMT1 protein, human EZH2 protein, human Gene Products, Protein Genes Malignant Neoplasms Methylation Microarray Analysis MYCN protein, human Neurosecretory Systems pathogenesis Prostate Prostate Cancer RNA-Seq Stem, Plant Transcriptome
Dataset 1: 450K dataset of a total of 39 methylation laboratory standard control samples reported by [13 (link)]. Human unmethylated DNA (HCT116 double knock out (DKO) of both DNA methyltransferases DNMT1 (-/-) and DNMT3b (-/-)) and fully methylated DNA (HCT116 DKO DNA enzymatically methylated) were obtained commercially (Zymo Research, Irving CA) and mixed together in different proportions to create laboratory control samples with specific methylation levels: 0, 5, 10, 20, 40, 50, 60, 80 and 100% methylated. Replicates for each methylation level (n = 10, 3, 2, 3, 3, 2, 3, 3 and 10, respectively) were independently assayed on different arrays.
Dataset 2: 450K dataset of 22 samples reported by [4 (link)]. These samples included three replicates from the HCT116 WT cell-line, three replicates from the HCT116 DNMT1 and DNMT3B double KO (DKO) cell-line, and 16 other samples (GEO accession number: GSE29290). In particular to evaluate RELIC and other dye-bias correction methods, we used the six replicates from the HCT116 WT and HCT116 DKO cell-lines, and the matched bisulfite pyrosequencing (BPS) data for 15 probes in the two cell-lines reported in the Table one of [4 (link)]. As described in [4 (link)] the fifteen CpGs were selected for technical validation of the 450K array measures (six sites for Infinium I assay and nine sites for Infinium II assay) using the more accurate BPS method as the “gold standard”.
Dataset 3: 450K dataset of 24 samples reported by [6 (link)]. These samples included 12 blood samples and 12 saliva samples for ten individuals, with two individuals having two technical blood/saliva replicates (GEO accession number: GSE73745). More specifically, we used these samples and the matched bisulfite pyrosequencing (BPS) data for three probes (cg19754622, cg16106427, cg08899523) to evaluate RELIC and other dye-bias correction methods.
Publication 2017
Biological Assay BLOOD Cell Lines cytidylyl-3'-5'-guanosine DNA Modification Methylases DNMT1 protein, human DNMT3B protein, human Gold HCT116 Cells Homo sapiens hydrogen sulfite Methylation Saliva
Mouse feeder free E14Tg2A ES cells were maintained on gelatin-coated dishes in Glasgow Minimum Essential medium (GMEM; GIBCO), supplemented with 15% heat-inactivated fetal bovine serum, 55 µM β-mercaptoethanol (GIBCO), 2 mM L-glutamine, 0.1 mM MEM nonessential amino acid, 5,000 units/ml penicillin/streptomycin and 1,000 units/ml of ESGRO (Chemicon) under feeder-free conditions. J1 ES cells and Dnmt1−/−Dnmt3a−/−Dnmt3b−/− (DNMT TKO) ES cells were maintained on gelatin-coated dishes with mitotically inactivated mouse embryonic fibroblasts. Alkaline phosphatase staining was performed with Alkaline Phosphatase Detection Kit (Chemicon). For growth curve analysis, control and knockdown ESCs were plated at 1×103 cells/cm2 and counted for 6 consecutive days. For self-renewal analysis cells were plated on 96 well plates at single cell density and the number of colonies on each plate was counted 6 days after plating.
To knockdown Tet proteins, lentiviral transduction was performed in mouse ES cells as described previously 19 (link). Short-hairpin RNA (shRNA) sequences (Supplementary Table 2) were cloned into pTY vetctor under the U6 promoter. To rescue Tet1 knockdown with Nanog, cDNA of Nanog was placed downstream of puromycin-resistant gene and foot-and-mouth disease virus 2A segment (Fig. S12), which enables multicistronic expression of transgenes in ES cells using a single promoter 21 (link).
Total RNA from mouse tissues was isolated using Trizol reagent (Invitrogen) and total RNA from cultured cells was isolated using RNeasy Mini Kit (Qiagen), and cDNA was generated with Improm-IITM Reverse Transcription System (Promega). Real-time quantitative PCR reactions were performed on an ABI PRISM 7700 Sequence Detection System (Applied Biosystems) using SYBR Green reagent (Invitrogen). cDNA levels of target genes were analyzed using comparative Ct methods, where Ct is the cycle threshold number and normalized to GAPDH. RT-qPCR primers are listed in Supplementary table 3.
Publication 2010
2-Mercaptoethanol Alkaline Phosphatase Amino Acids Cells Cultured Cells DNA, Complementary DNA Modification Methylases DNMT1 protein, human DNMT3B protein, human Embryo Embryonic Stem Cells Enhanced S-Cone Syndrome Feeder Cells Fetal Bovine Serum Fibroblasts Foot-and-Mouth Disease Virus GAPDH protein, human Gelatins Genes Glutamine Hyperostosis, Diffuse Idiopathic Skeletal Mus neuronectin Oligonucleotide Primers Penicillins prisma Promega Proteins Puromycin Reverse Transcription Short Hairpin RNA Streptomycin SYBR Green I Tissues Transgenes trizol
Plasmid sequences can be found in Supplementary Table S6. Targeting donor constructs were either synthesized as ssDNA oligonucleotides (Integrated DNA Technologies) or produced by amplifying 300 to 200 bp long homology arms with the respective external and internal primer sets (Supplementary Table S2). These PCR products of the 5′ and 3′ homology arms were pooled and an overlap extension PCR with the external primers was performed to yield the final targeting fragments. The gRNA vector was synthesized at Eurofins MWG Operon based on the sequences described (3 (link)). The subcloning of targeting sequences was performed by circular amplification. The surrogate reporter (pSR) was generated by inserting in vitro annealed DNA oligos via AsiSI and NruI into pCAG-mCh (18 (link)). eGFP was amplified using the primers eGFP-F and eGFP-R and sequentially cloned into pCAG-mCh-NruI linker to generate the pSR construct. Reporters were generated by subcloning invitro annealed DNA oligos containing CRISPR target sites into KpnI and NheI digested pSR. The attB-GFP-knockin construct was generated from R6K-NFLAP (19 (link)) by ligation free cloning (20 (link)) rearranging the backbone sequences into the artificial intron and introducing the attB site 5′ of the GFP open reading frame (ORF), removing its start codon. The attB-GFP-Poly(A) and attB-mCh-Poly(A) constructs were created by amplifying the GFP ORF including the stop codon and SV40 Poly(A) signal from pCAG-eGFP-IB and inserted into the attB-LAP-tag backbone by ligation free cloning. The attB-mCh-Poly(A)-mPGK-PuroR construct was generated by subcloning the mPGK-PuroR sequence from pPthc-Oct3/4 (21 (link)) and ligating it into the EcoRV site of the attB-mCh-Poly(A) construct. The attB-GFP-Poly(A)-mPGK-NeoR was produced by first exchanging the PuroR in pPthc-Oct3/4 with NeoR from pEGFP-C1 (22 (link)) using HindIII. The combined mPGK-NeoR was then subcloned into the attB-GFP-Poly(A) vector via the same EcoRV site mentioned previously. The attB-GFP-Dnmt1cDNA-Poly(A), attB-GFP-Tet1cDNA-Poly(A) and attB-GFP-Dnmt3b1cDNA-Poly(A) constructs were generated by inserting the appropriate cDNAs from constructs reported previously (17 (link),23 (link)–24 (link)) via AsiSI/NotI sites into the attB-GFP-Poly(A) and attB-mCh-Poly(A) vectors respectively. The attB-GFP-Dnmt3b6-Poly(A), attB-GFP-Tet1-d1–1363-Poly(A), attB-GFP-Tet1-d833–1053-Poly(A), attB-GFP-Tet1-d833–1363-Poly(A) vectors were produced via circular amplification with overlap extension primers using the above mentioned attB-GFP-Dnmt1/Dnmt3b1/Tet1cDNA-Poly(A) constructs as templates.
The attB-GFP-Dnmt3b6-Poly(A)-mPGK-NeoR and attB-mCh-Dnmt3b1-Poly(A)-mPGK-PuroR integration constructs were created by inserting the Dnmt3b6 and Dnmt3b1 sequences (from attB-GFP-Dnmt3b6-Poly(A) and attB-GFP-Dnmt3b1-Poly(A)) using AsiSI/NotI sites into attB-GFP-Poly(A)-mPGK-NeoR and attB-mCh-Poly(A)-mPGK-PuroR vectors, respectively.
All constructs described in this study are available via Addgene or via http://human.bio.lmu.de/_webtools/MINtool/.
Publication 2015
2',5'-oligoadenylate Arm, Upper Base Sequence Cloning Vectors Clustered Regularly Interspaced Short Palindromic Repeats Codon, Initiator Codon, Terminator DNA, Complementary DNA, Single-Stranded DNMT1 protein, human Homo sapiens Introns Ligation Oligonucleotide Primers Oligonucleotides Operon Plasmids Poly A POU5F1 protein, human Simian virus 40 Tissue Donors Vertebral Column
CD8+ T cells were purified from spleens of OT-I mice by negative selection with magnetic beads (EasySep, Stemcell Technologies). After purification, cells were 97.7±0.5% CD8+ T cell and contained 0.11±0.04% CD11b+ CD11c- monocytes and 0.09±0.05% CD11b+ CD11c+ dendritic cells. In each well of a 24-well plate, 5x105 of the purified CD8+ T cells/ml were cultured in complete media (RPMI 1640, 10% FBS (Gibco), 1% 2mM L-glutamine (Life Technologies), 1% HEPES (Life Technologies), 1% 100nM Sodium Pyruvate (Life Technologies), 1% non-essential amino acides (Life Technologies), 100U/ml penicillin (Gibco) and 100μg/ml Streptomycin-sulfate (Gibco), 0.05mM Betamercaptoethanol (Sigma)) with IL-15 (5ng/ml, Peprotech, Cat 210–15) and IL-7 (5ng/ml, Peprotech, Cat 210–07) with or without 10ng/ml OVA(257–264) peptide (Anaspec Cat AS-60193).
For single peptide stimulation, cells were cultured in the presence of OVA(257–264) peptide for 48 hours. The peptide was then removed by washing the cells two times with complete media. For the remaining 3 days, the cells were cultured in the complete media with cytokines. For repeat peptide stimulation, 10ng/ml OVA(257–264) peptide was added daily for five days. The cells were washed also on day 2 to allow for comparable culture conditions. Unstimulated control cells were cultured in media with cytokines but without adding peptide. Cells from all three conditions were checked daily, and when the cells were confluent, they were split and cultured with fresh complete media containing cytokines. After day 5, some of the cell were washed two times with complete media and maintained in the media only with cytokines for another three days. In some experiments cell cultures were treated on day 2 with 20μM DNA methyltransferase (DNMT) inhibitor SGI-1027 (Tocris, Bio-techne) that targets DNA methyltransferases DNMT3B, DNMT3A and DNMT1.
On day five, cells were harvested and counted using an automated counting system (Countess, Life Technologies). Cells were stained with DAPI Viability dye (Beckman Coulter, Cat B30437) and Acridine Orange (Biotium, Cat 40039) to distinguish live and dead cells.
Publication 2020
Acridine Orange CD8-Positive T-Lymphocytes Cell Culture Techniques Cells Culture Media Cytokine DAPI Dendritic Cells DNA Modification Methylases DNMT1 protein, human DNMT3B protein, human Glutamine HEPES Interleukin-15 ITGAM protein, human Monocytes Mus Penicillins Peptides Pyruvate SGI-1027 Sodium Stem Cells Streptomycin Sulfate

Most recents protocols related to «DNMT1 protein, human»

Values are shown as the mean ± standard error of mean (SEM), and error bars for scatter dot plots represent one SEM. Since aerobic capacity and cardiac fibrosis are significant clinical outcomes related to the survival of HF patients [4 (link), 8 (link)], power (1- β) analysis for paired sample t tests used to compare the difference in V O2peak and ECV fractions before and after HIIT. Differences in physical PCS, MCS, and LVWMS were estimated by the chi-square test.
The nonparametric test was used in the study owing to the limited sample size. The Wilcoxon signed rank test was conducted to estimate within-group differences between data before and after HIIT, including exercise capacity function, CMR-LGE results (LV geometry, functions, and ECV fractions), and blood chemistry data. The Mann‒Whitney U test was used to estimate differences in selected protein amounts obtained from LC‒MS results and methylation levels between cells incubated in patient serum before and after HIIT. Relationships between the DNMT1 levels and health-related physical fitness and CMR-LGE findings were assessed by Spearman’s correlation analysis.
Relative protein expression (measurements/baseline) of VLCAD, Cyto C, CASP3, lamin B1, actin and Arp2 in HCFs between the original and knockdown of ACADVL was compared by the Mann‒Whitney U test. This test was also used to assess mitochondrial intensity in HCFs treated with patient serum before and after HIIT and in cells with and without ACADVL knockdown. Kruskall-Wallis test was conducted to assess cell migration speed in three different culture media and with different cell numbers at different times (baseline, 24 h and 48 h after inoculation). Multiple comparisons Dunn’s test was used to estimate differences of cell behaviours between each of the above sampling time. The relationships between normalized changes (  ΔValue=Valuepost-HIIT-Valuepre-HIITValuepre-HIIT ) in exercise performance and CMR-LGE measurements after HIIT were estimated by Spearman correlation and partial correlation analysis after controlling LV mass. All statistical assessments were considered significant at p < 0.05.
Publication 2023
Actins Acyl-Coa Dehydrogenase Very Long Chain Deficiency Blood Chemical Analysis Caspase 3 Cells Culture Media DNMT1 protein, human Exercise, Aerobic Fibrosis Heart lamin B1 Long-Chain-Acyl-CoA Dehydrogenase Methylation Migration, Cell Mitochondria Patients Physical Examination Proteins Serum Vaccination
The ACADVL gene encodes for very long-chain acyl-CoA dehydrogenase (VLCAD), which functions within mitochondria and is essential for fatty acid oxidation. HCFs were prepared for western blot analysis of VLCAD, caspase-3 (CASP3), cytochrome c (Cyto C), lamin B1, β-actin, and Arp2 with the internal reference protein glyceraldehyde 3-phosphate dehydrogenase (GAPDH) before knockdown of the ACADVL gene. The above proteins were quantified again after knockdown of ACADVL. Detailed methods of the western blotting and knockdown procedure are provided in Additional file 2.
Knockdown of DNMT1 leads to generally decreased DNA methylation and activates cascades of genotoxic stress [31 (link)] in cells, resulting in signal transduction unrelated to cardiac fibrosis. Thus, we preferred to downregulate the ACADVL gene expression to simulate the HIIT-associated inhibition of human cardiac fibroblast activities.
Publication 2023
Actin-Related Protein 2 Actins Acyl-Coa Dehydrogenase Very Long Chain Deficiency Caspase 3 Cells Cytochromes c DNA Methylation DNMT1 protein, human Fatty Acids, Essential Fibroblasts Fibrosis Gene Expression Gene Knockdown Techniques Genes Genotoxic Stress Glyceraldehyde-3-Phosphate Dehydrogenases Heart lamin B1 Long-Chain-Acyl-CoA Dehydrogenase Mitochondria Proteins Psychological Inhibition Signal Transduction Western Blot
To test for an association between overall piRNA or KRAB-ZFP pathway activity and genome size, we first compiled male and female gonad RNA-Seq datasets for vertebrates of diverse genome sizes, including P. ornatum (ornate burrowing frog), Gallus gallus (chicken), D. rerio (zebrafish), Xenopus tropicalis (Western clawed frog), A. carolinensis (green anole), Mus musculus (mouse), Geotrypetes seraphini (Gaboon caecilian), Rhinatrema bivittatum (two-lined caecilian), and Caecilia tentaculata (bearded caecilian) spanning genomes sizes from 1.0—5.5 Gb, and P. waltl (the Iberian ribbed newt), A. mexicanum (the Mexican axolotl), C. orientalis (the fire-bellied newt), P. annectens, and P. aethiopicus (African and marbled lungfishes) spanning genome sizes from 20—∼130 Gb (Supplementary Files S8,S9). We performed de novo assemblies using the same pipeline as for R. sibiricus on all obtained datasets.
We identified transcripts of 21 genes receiving a direct annotation of piRNA processing in vertebrates in the Gene Ontology knowledgebase that were present in the majority of our target species: ASZ1, BTBD18 (BTBDI), DDX4, EXD1, FKBP6, GPAT2, HENMT1 (HENMT), MAEL, MOV10l1 (M10L1), PIWIL1, PIWIL2, PIWIL4, PLD6, TDRD1, TDRD5, TDRD6, TDRD7, TDRD9, TDRD12 (TDR12), TDRD15 (TDR15), and TDRKH. In addition, we identified transcripts of 14 genes encoding proteins that create a transcriptionally repressive chromatin environment in response to recruitment by PIWI proteins or KRAB-ZFP proteins, 12 of which received a direct annotation of NuRD complex in the Gene Ontology knowledgebase and 2 of which were taken from the literature: CBX5, CHD3, CHD4, CSNK2A1 (CSK21), DNMT1, GATAD2A (P66A), MBD3, MTA1, MTA2, RBBP4, RBBP7, SALL1, SETDB1 (SETB1), and ZBTB7A (ZBT7A) (Ecco et al., 2017 (link); Wang et al., 2023 (link)). Finally, we identified TRIM28, which bridges this repressive complex to TE-bound KRAB-ZFP proteins in tetrapods, lungfishes, and coelacanths (Ecco et al., 2017 (link)). For comparison, we identified transcripts of 14 protein-coding genes receiving a direct annotation of miRNA processing in vertebrates in the Gene Ontology knowledgebase, which we did not predict to differ in expression based on genome size: ADAR (DSRAD), AGO1, AGO2, AGO3, AGO4, DICER1, NUP155 (NU155), PUM1, PUM2, SNIP1, SPOUT1 (CI114), TARBP2 (TRBP2), TRIM71 (LIN41), and ZC3H7B. Expression levels for each transcript in each individual were measured with Salmon (Patro et al., 2017 (link)) (Supplementary File S10).
As a proxy for overall piRNA silencing activity, for each individual, we calculated the ratio of total piRNA pathway expression (summed TPM of 21 genes) to total miRNA pathway expression (summed TPM of 14 genes). As a proxy for transcriptional repression driven by both the piRNA pathway and KRAB-ZFP binding activity, we calculated the ratio of total transcriptional repression machinery expression (summed TPM of 14 genes) to total miRNA pathway expression. Finally, we calculated the ratio of TRIM28 expression to total miRNA pathway expression for each individual. We also calculated these ratios with a more conservative dataset allowing for no missing genes; this yielded 15 piRNA pathway genes, 9 KRAB-ZFP genes, and 13 miRNA genes. We plotted these ratios to reveal any relationship between TE silencing pathway expression and genome size.
Publication 2023
Ambystoma mexicanum BRAF protein, human CHD4 protein, human Chickens Chromatin CSNK2A1 protein, human DICER1 protein, human DNMT1 protein, human EIF2C2 protein, human Gene Products, Protein Genes Genome Males methyl-CpG binding domain protein 3, human Mi-2 Nucleosome Remodeling and Deacetylase Complex Mice, House MicroRNAs Mta1 protein, human Mus Negroid Races Newts Ovary Piwi-Interacting RNA Proteins PUM2 protein, human Rana RBBP7 protein, human Repression, Psychology RNA-Seq Salmon SETDB1 protein, human Transcription, Genetic TRIM28 protein, human Vertebrates Xenopus laevis ZBTB7A protein, human ZC3H7B protein, human Zebrafish
Full details of the methodology have been described previously15 (link). In brief, DNA libraries were prepared using either TruSeq Nano HT Sample Prep Kit (Illumina) or KAPA PCR-Free v2.1 (Roche) and sequenced on the Illumina HiSeq X Ten platform. Germline samples were sequenced to an average depth of 30X and tumour samples 90X. RNA libraries used the TruSeq Stranded mRNA Preparation Kit and sequenced on either the HiSeq 4000 or NextSeq 500 platform to a targeted paired-end read depth of 80 M reads.
WGS germline variants were subtracted from the tumour sequencing data to identify somatic only variants. Somatic variants were individually analysed with prioritisation of variants in cancer related genes and were manually mined for any mutations containing K27M, K27I, G34R, G34V or G34W changes in the following histone genes: H3F3A, H3F3B, H3F3C, HIST1H1A, HIST1H1B, HIST1H2AA, HIST1H2BA, HIST1H3A, HIST1H4A, HIST1H3C, HIST2H3C, HIST3H2BB and HIST3H3. Genes involved in the PRC2 complex and common epigenetic regulators were also mined for molecular aberrations in DNMT1, DNMT3A, DNMT3B, DNMT3L, TET1, TET2, TET3, EED, EZH2, SUZ12, SET, RBAP, KDM6A, BCOR, BCORL1, CREBBP, LZTR1 and ASZL1.
Publication 2023
BCORL1 protein, human Diploid Cell DNA Library DNMT1 protein, human DNMT3B protein, human EZH2 protein, human Gene, Cancer Genes Germ-Line Mutation Germ Line Histones KDM6A protein, human Mutation Neoplasms Polycomb Repressive Complex 2 RNA, Messenger
From Superbiotek in Shanghai, China (#REC1601), we acquired a TMA of 80 paired rectal cancers and corresponding normal tissues. Surgical samples from the patients were taken between May 2008 and December 2012 through operations. The patients’ median survival duration was 81.5 months, ranging from 14 to 130 months. For every case, clinicopathological information including overall survival time, survival status, age, gender, tumor size, pathological T, N, and M stage, and grade was accessible. Based on this commercial TMA, we conducted a retrospective analysis.
For immunohistochemistry (IHC) process, the TMA slides were deparaffinized, rehydrated, and incubated by 3% hydrogen peroxide to block the endogenous peroxidase activity for 10 min at room temperature. Antigens were restored by boiling in a pressure cooker containing sodium citrate buffer for 90 s. The slides were incubated in bovine serum albumin (BSA) for 30 min to reduce nonspecific background. Then, they were incubated with rabbit monoclonal NSUN4 antibody (HPA028489, Sigma), NSUN7 antibody (HPA020653, Sigma), and DNMT1 antibody (HPA002694, Sigma) at 4°C overnight. Next, secondary antibody was incubated with the slides for 1 h at 37°C. Finally, the slides were developed in 3, 3’-diaminobenzidine (DAB) and stained with hematoxylin.
The slides were assessed digitally with the APERIO ScanScope (Leica Biosystems, Germany) and the APERIO ImageScope (Leica Biosystems, Germany) using the positive pixel counting algorithm. The IHC staining results were interpreted by both the intensity of staining and the staining positive area. Each sample was assigned a score according to the intensity of the staining (0 = no staining; 1 = weak staining; 2 = moderate staining; and 3 = strong staining) and the proportion of stained cells (0 = 0%; 1 = 1%–25%; 2 = 25%–50%; 3 = 50%–75%; 4 = 75%–100%). The final score was calculated as the staining intensity multiplying positive area score, ranging from 0 to 12. The IHC results of TMA-rectal cancer were independently reviewed by two experienced pathologists who were blinded to the clinical parameters.
Publication 2023
Antigens Buffers Cardiac Arrest Debility Division Phase, Cell DNMT1 protein, human Gender Immunoglobulins Immunohistochemistry Monoclonal Antibodies Neoplasms Operative Surgical Procedures Pathologists Patients Peroxidase Peroxide, Hydrogen Pressure Rabbits Rectal Cancer Serum Albumin, Bovine Sodium Citrate Tissues

Top products related to «DNMT1 protein, human»

Sourced in United States, United Kingdom
DNMT1 is an enzyme that catalyzes the transfer of methyl groups to DNA molecules, playing a crucial role in the maintenance of DNA methylation patterns. It is involved in the epigenetic regulation of gene expression.
Sourced in United States, China, Germany, United Kingdom, Canada, Japan, France, Italy, Switzerland, Australia, Spain, Belgium, Denmark, Singapore, India, Netherlands, Sweden, New Zealand, Portugal, Poland, Israel, Lithuania, Hong Kong, Argentina, Ireland, Austria, Czechia, Cameroon, Taiwan, Province of China, Morocco
Lipofectamine 2000 is a cationic lipid-based transfection reagent designed for efficient and reliable delivery of nucleic acids, such as plasmid DNA and small interfering RNA (siRNA), into a wide range of eukaryotic cell types. It facilitates the formation of complexes between the nucleic acid and the lipid components, which can then be introduced into cells to enable gene expression or gene silencing studies.
Sourced in United States, China, Japan, Germany, United Kingdom, Canada, France, Italy, Australia, Spain, Switzerland, Netherlands, Belgium, Lithuania, Denmark, Singapore, New Zealand, India, Brazil, Argentina, Sweden, Norway, Austria, Poland, Finland, Israel, Hong Kong, Cameroon, Sao Tome and Principe, Macao, Taiwan, Province of China, Thailand
TRIzol reagent is a monophasic solution of phenol, guanidine isothiocyanate, and other proprietary components designed for the isolation of total RNA, DNA, and proteins from a variety of biological samples. The reagent maintains the integrity of the RNA while disrupting cells and dissolving cell components.
Sourced in United States, China
DNMT1 is a protein involved in the maintenance of DNA methylation patterns during cell division. It functions to preserve the existing methylation marks on the newly replicated DNA strand.
Sourced in United States, United Kingdom
Ab13537 is a primary antibody produced in rabbit that targets the human CCL5 protein. It is intended for use in immunohistochemistry, western blotting, and ELISA applications.
Sourced in United Kingdom, United States
Anti-DNMT1 is a laboratory reagent that detects and quantifies the expression of DNA (cytosine-5)-methyltransferase 1 (DNMT1) protein. DNMT1 is a key enzyme involved in the maintenance of DNA methylation patterns during cell division.
Sourced in United States, Germany, China, United Kingdom, Morocco, Ireland, France, Italy, Japan, Canada, Spain, Switzerland, New Zealand, India, Hong Kong, Sao Tome and Principe, Sweden, Netherlands, Australia, Belgium, Austria
PVDF membranes are a type of laboratory equipment used for a variety of applications. They are made from polyvinylidene fluoride (PVDF), a durable and chemically resistant material. PVDF membranes are known for their high mechanical strength, thermal stability, and resistance to a wide range of chemicals. They are commonly used in various filtration, separation, and analysis processes in scientific and research settings.
Sourced in United States, United Kingdom, China
DNMT3B is a DNA methyltransferase enzyme that catalyzes the addition of methyl groups to cytosine residues in DNA, playing a critical role in the regulation of gene expression and genomic imprinting.
Sourced in United States, China
DNMT1 is a DNA methyltransferase enzyme that catalyzes the transfer of methyl groups to cytosine residues in DNA. It plays a crucial role in the maintenance of DNA methylation patterns during DNA replication and is essential for normal cellular function.
Sourced in United States, United Kingdom
DNMT3A is a DNA methyltransferase enzyme that catalyzes the transfer of methyl groups to cytosine residues in DNA, playing a key role in the establishment and maintenance of DNA methylation patterns. This enzyme is essential for de novo DNA methylation during embryogenesis and embryonic development.

More about "DNMT1 protein, human"

Explore the crucial role of DNA methyltransferase enzyme DNMT1 in maintaining DNA methylation patterns during cell division.
This epigenetic regulator is essential for normal development, genomic imprinting, and gene expression control.
Disruption of DNMT1 function has been linked to various diseases, including cancer, neurological disorders, and immune system dysfunctions.
Leverage PubCompare.ai, an AI-driven platform, to optimize your DNMT1 protein research by locating relevant protocols from literature, preprints, and patents, and using AI-driven comparisons to identify the best protocols and products for enhanced reproducibility and accuracy.
Discover how DNMT1, Lipofectamine 2000, TRIzol reagent, Ab13537, Anti-DNMT1, PVDF membranes, DNMT3B, and DNMT3A can be utilized in your DNMT1 protein research to maximize results and unlock new insights.