The largest database of trusted experimental protocols

C-src Genes

C-src genes encode a non-receptor tyrosine kinase that plays a key role in cell signaling and regulation of cellular processes such as cell growth, differentiation, and motility.
Alterations or dysregulation of c-src genes have been implicated in the development and progression of various cancers, making them an important target for research and therapeutic interventions.

Most cited protocols related to «C-src Genes»

DNA sequences coding for codon-optimized 2A peptides were generated from the oligonucleotides in Table 1, adding 3 amino acids to the N-terminus and 4 amino acids to the C-terminus of the peptide ([7] (link)) (BamHI and BglII sites underlined). 2A peptide sequence variants used are shown in Table 2. 2A sequences were generated by annealing oligos, cutting with BamHI and BglII and cloning into the BglII site of pC5-Kan ([8] ) to produce pC5-Kan 2A. This vector contains 18 commonly used restriction enzyme sites N-terminal to the 2A sequence and 9 additional C-terminal sites that facilitate cloning genes of interest on both sides of the 2A sequence, as well as rare 8-cutter sites for shuttling the gene-2A-gene cassette into the pUAS-C5 ([8] ) expression vector.
MyrGFP was created by PCR using F-myrGFP BclI and R-GFPo StuI and pJFRC19 template ([9] (link)) which contains a fly codon-optimized myristylation sequence derived from the first 85 amino acids of the Drosophila Src homolog (Src64B) followed by a codon-optimized EGFP (F64L, S65T). This PCR product was cut with BclI and StuI and ligated with pC5-Kan 2A cut with BamHI/StuI.
DsRed-nls is also called RedStinger ([10] ), which is a DsRed variant containing a nuclear localization signal from the Drosophila transformer gene. It was amplified by PCR with F-dsRed BamHI and R-dsRed XbaI, and cloned into pCR8GW TOPO, then excised with BamHI/XbaI and cloned into pC5-Kan myrGFP-2A cut with BglII/AvrII to make pC5-Kan myrGFP-2A–RedStinger.
pPac-C5 was generated by modifying the MCS from pPac-PL (C.S. Thummel, unpublished). A BamHI/AvrII fragment from pC5-Kan was inserted into BamHI/XbaI sites of pPacPL to make pPac-C5 with a MCS including the following unique restriction sites: BamHI, XbaI, XhoI, StuI, AgeI, MluI, NheI, KpnI, NotI.
A SpeI/AscI fragment from pC5-Kan myrGFP-2A–RedStinger was then cloned into XbaI/MluI sites of pPac-C5 to generate pPac-C5 myrGFP-2A–RedStinger.
To make pPac-C5 myrGFP-dsRed-nls (no 2A peptide), myrGFP was subcloned from pC5-Kan myrGFP P2A into pBS-C5 ([8] ) using EcoRI/StuI. Then dsRed-nls PCR cut with BamHI/XbaI (as described above) was added into BglII/NheI sites of pBS-C5 myrGFP. myrGFP-dsRed-nls was then cut with SpeI/AscI and ligated with pPac-C5 cut with XbaI/MluI to generate pPac-C5 myrGFP dsRed-nls (no 2A peptide control).
pUAS-C5 attB was made by inserting a 285 bp attB site created from PCR with F-attB NdeI and R-attB NdeI using pattB as template ([11] (link)), then inserted into NdeI site of pUAS-C5 ([8] ).
A SpeI/AscI fragment from pC5-Kan myrGFP-2A–RedStinger was subcloned into pUAS-C5 attB cut with SpeI/AscI and injected into a fly harboring the p{CaryIP}Su(hw)attP1 site at 87B13 on the third chromosome using standard techniques to generate UAS-myrGFP-2A–RedStinger flies.
To make pUAS-C5 attB tdTomato-P2A–GCamP5G, tdTomato was made by PCR with F-tdTomato BamHI and R-tdTomato HpaI using pRSET-B tdTomato template (R. Tsien; [12] (link)), cut with BamHI/HpaI and inserted into the BamHI/StuI sites of pC5-Kan P2A. GCamP5G was amplified from Addgene plasmid #31788 with F-GCaMP5G BamHI and R-GCaMP5G XbaI, then cut with BamHI/XbaI and inserted into BglII/AvrII sites of pC5-Kan tdTomato P2A. The tdTomato-P2A–GCaMP5G fragment was then subcloned into pUAS-C5 attB with PacI/AscI and injected into p{CaryIP}Su(hw)attP1 site at 87B13 on the third chromosome.
Act5c-LexAGADfl flies were generated by amplifying the act5c promoter using F-act5c and R-act5c primers, cloning into pCR8-GW-TOPO (Invitrogen), and performing an L/R reaction with pBPnlsLexAGADflUw ([9] (link)). The resulting construct was integrated into the p{CaryP}attP40 site at 25C6 on the second chromosome.
pC5-Kan P2A has been deposited with Addgene (plasmid #70838) and GenBank (KJ470630). Other plasmids, sequences, and flies are available upon request.
Publication 2014
Chemicals. Tributyltin chloride (TBT; CAS No. 1461-22-9) and Nile red (CAS No. 7385-67-3) were purchased from Sigma-Aldrich, and lipid standards were from Advanti Polar Lipids. All other chemicals were analytical grade and were obtained from Merck.
TBT treatments. TBT was dissolved in acetone; the same amount of acetone (< 0.1 mL/L) was used for a solvent control and in all experimental treatments except in the untreated control (control) to account for any carrier effect. Actual TBT concentrations in test solutions were measured as total tin using a Perkin-Elmer Elan 6000 inductively coupled plasma mass spectrometer (ICP-MS) (Barata et al. 2005 (link)), and were confirmed to be within 10% of nominal concentrations (0.036 and 0.36 μg/L for 0.1 and 1 μg/L doses, respectively).
Experimental animals. All experiments were performed using the well-characterized clone F of D. magna maintained indefinitely as pure parthenogenetic cultures (Barata and Baird 1998 ). Individual cultures were maintained in 100 mL of ASTM hard synthetic water at low and high food-ration levels (Chlorella vulgaris, 1 × 105 and 5 × 105 cells/mL, respectively), as described by Barata and Baird (1998) .
Experimental design. Experiments were initiated with newborn neonates < 4–8 hr old obtained from synchronized females cultured individually at high food-ration levels. Groups of five neonates (F0) were reared in 150 mL of ASTM hard water under high food-ration conditions until the end of the third juvenile instar (about 4–8 hr before molting for the third time). At this point, juveniles were used in three sets of experiments using two TBT treatments, 0.1 μg/L (low; TBT L) and 1 μg/L (high; TBT H). Five to 10 replicates per treatment were used.
The first experiment studied effects of exposure to TBT during the adolescent instar (i.e., 3 days) on the life history of these females (F0) through five consecutive clutches. Their first clutch of neonates, exposed during the egg-provisioning stage (F1) was similarly studied during four consecutive clutches. Following exposure to TBT, F1 females were cultured individually under high food conditions without TBT, and their growth and reproduction performance monitored until the fifth clutch. The tolerance of F1 neonates to starving conditions was studied monitoring the time to death of 10 neonates individually cultured in 50 mL of ASTM hard water alone. The medium was renewed every day. Life-history performance of F1 neonates was studied by culturing them individually in 100 mL of ASTM hard water at high food conditions until the release of the fourth clutch. Measured life-history traits were survival, reproduction, body length of each adult instar (including that of the adolescent instar), age at first reproduction, the size of neonates of each clutch, and the population growth rate (r) estimated from the age-dependent survival and reproduction rates according to the Lotka equation (Barata et al. 2001 ).
The second set of experiments (experiments 1 and 2) aimed to study lipid droplet changes across food and TBT treatments using the Nile red assay. In experiment 1, animals were exposed to three food regimes: starving (no food added), low food (1 × 105 cells/mL C. vulgaris), and high food (5 × 105 cells/mL C. vulgaris). In experiment 2, animals were exposed to two TBT concentrations (TBT L and TBT H) across low and high food levels. Exposures lasted through all the adolescent instars, and females were sampled just after their fourth molt and having released their eggs into the brood pouch (48 hr), as shown in Supplemental Material, Figure S1.
The third set of experiments aimed to determine effects of TBT L and TBT H on the dynamics of lipids, lipid droplets, and mRNA levels of selected genes across an entire adolescent intermolt cycle. Experiments were conducted only at high food levels and included five samplings: 0 hr (just after the third molt), 8 hr, 16 hr, 24 hr, and just after the fourth molt (48 hr). At each sampling, three and five replicates of 5 individuals were collected and processed for total lipid determination and mRNA gene transcription measurement, respectively, and 10 animals were processed for Nile red determination. At the 48-hr sampling period, females were de-brooded by gently flushing water into the brood pouch. Obtained eggs and de-brooded females were then collected and used for lipid and gene transcription analyses. Because of the large number of synchronized animals needed, three different independent but consecutive experiments were performed and used for lipidomic, gene transcription, and Nile red determinations, respectively.
Nile red determination. The Nile red stock solution was prepared in acetone and stored protected from light following Tingaud-Sequeira et al. (2011) (link). Just before use, the working solution was prepared by diluting the stock solution to 1.5 μM in ASTM. Live individuals were then exposed to Nile red working solution in the dark for 1 hr at 20°C. After incubation, animals were placed in 100 mL ASTM for 1 min to allow clearance of Nile red residuals. Following clearance, animals were placed individually in 1.5-mL centrifuge tubes, the remaining water was removed, and samples were sonicated in 300 μL of isopropanol. The homogenized extract was then centrifuged at 10,000 × g. We used 200 μL of supernatant to measure Nile red fluorescence using an excitation/emission wavelength of 530/590 nm and a microplate fluorescence reader (Synergy 2, BioTek). Each treatment had one animal per sample (10 replicates in total). For each quantification and treatment, 10 blanks (animals not exposed to Nile red) were used to account for background levels of fluorescence. After exposure to Nile red, images were taken in the area surrounding the midgut for visualization of lipid droplets. Fluorescence and bright file images were obtained using a Nikon SMZ1500 microscope and a Nikon Intensilight C-HGFI with a GFP filter (EX 472/30, EM 520/35; Nikon).
Lipidomic analyses. Lipidomic analyses were performed as described by Gorrochategui et al. (2014) (link), with minor modifications. Each replicate consisted of a pool of five animals that were homogenized in 500 μL phosphate-buffered saline (PBS), pH 7.4, with 2,6-di-tert-butyl-4-methylphenol (BHT; 0.01%) as an antioxidant. Lipid extraction was performed using a modification of Folch’s method (Folch et al. 1957 (link)). Briefly, 100 μL of the homogenized sample was mixed with 500 μL of chloroform and 250 μL of methanol. Internal standards (200 pmol) (described in Supplemental Material, Table S1) were also added. Samples were heated at 48°C overnight and dried under N2 the next day. Lipid extracts were solubilized in 150 μL methanol. The liquid chromatograph–mass spectrometer consisted of a Waters Aquity UPLC system connected to a Waters LCT Premier Orthogonal Accelerated Time of Flight Mass Spectrometer (Waters) operated in positive and negative electrospray ionization (ESI) mode. Full-scan spectra from 50 to 1,500 Da were obtained. Mass accuracy and reproducibility were maintained by using an independent reference spray (LockSpray; Waters). A 100-mm × 2.1-mm i.d., 1.7-μm C8 Acquity UPLC BEH (Waters) analytical column was used. Further chromatographic details of mobile phases were described by Gorrochategui et al. (2014) (link).
Quantification was carried out using the ion chromatogram obtained for each compound using 50-mDa windows. The linear dynamic range was determined by injection of standard mixtures. Positive identification of compounds was based on the accurate mass measurement, with an error < 5 mg/L, and its LC retention time compared with that of a standard (± 2%).
A total of 116 lipids were identified and quantified by UPLC-TOF ESI-positive mode that were distributed as follows: five classes of glycerophospholipids [phosphocholine (PC) with 20 lipids, lysophosphatidylcholine (LPC) with 6 lipids, phosphatidylethanolamine (PE) with 9 lipids, phosphatidylserine (PS) with 7 lipids, and phosphatidylinositol (PI) with 3 lipids]; diacylglycerols (DG) with 20 lipids; triacylglycerols (TG) with 39 lipids; cholesterylesters (CE) with 4 lipids; and sphingolipids (SM) with 8 lipids. Glycerophospholipids, diacylglycerol, triacylglycerol, and cholesterylesters were annotated as :. Sphingolipids were annotated as :.
Transcriptomic analyses. Methods of extraction, purification, and quantification of mRNA from the studied genes and their primers follow previous procedures (Campos et al. 2013 (link)). Eight genes were selected for representation of different pathways/gene families: EcRB, HR3, HR38, Neverland, Hb2, RXR, MET, and SRC. The gene glyceraldehyde 3-phosphate dehydrogenase (G3PDH) was used as an internal control. For each of the genes, primers were designed using Primer Quest (IDT Technologies) and are listed in Supplemental Material, Table S2. Aliquots of 10 ng were used to quantify specific transcripts in a LightCycler® 480 real-time PCR system (Roche) using LightCycler 480 SYBR Green I Master® (Roche). Relative abundance values of all genes were calculated from the second derivative of their respective amplification curve (Cp; crossing point) values calculated by technical triplicates. Cp values of target genes were compared with the corresponding reference genes.
Data analyses. The effect of food rations and/or treatment or sampling period or juvenile stage on Nile red fluorescence, lipidomic profiles, mRNA abundance, and life history and physiological responses were analyzed by two-way and/or one-way analysis of variance (ANOVA). Post hoc Dunnett’s or Tukey’s tests were performed to compare exposure treatments with solvent controls. Prior to analyses, all data except survival responses were log transformed to achieve normality and variance homoscedasticity. If not indicated otherwise, significance levels were set at p < 0.05. Survival responses were assessed by Wilcoxon-Gehan tests. Tests were performed with IBM-SPSS statistics software, version 19. Lipidomic data were further analyzed using cluster and K-means analyses in R (R Core Team 2014 ) to identify clusters of lipid families similarly affected by TBT.
Publication 2015

Protocol full text hidden due to copyright restrictions

Open the protocol to access the free full text link

Publication 2009
CTNNB1 protein, human Genes K-ras Genes Oncogenes, myc TCF4 protein, human Transforming Growth Factor beta

Protocol full text hidden due to copyright restrictions

Open the protocol to access the free full text link

Publication 2012
Biopharmaceuticals DNA Replication Gender Gene Expression Genes Genetic Diversity Genome Transcriptome
We collected surgical stomach cancer tissue samples from 9 patients who underwent surgical operations between 2012 and 2015. Frozen tissue from patients who agreed to preoperative tissue collection was stored in a −70°C deep freezer.
We choose 4 SRC-GC and 5 WD-GC cases. The cancer tissue was cut into 5 µm sections, and H&E staining was performed. H&E-stained slides were used to identify the morphology and location of gastric cancer tissue within normal gastric tissue. Afterward, the remaining tissue was cut at 10-20 µm thickness. We performed tissue collection by laser capture microdissection (LCM; Veritas LCM2110, Molecular Device Corporation, CA, USA) and compared these slides with the H&E slides (Figure 1A-C) according to the manufacturer’s recommendations. After tissue collection, we extracted RNA using a Total Purification Kit (Norgene Bioteck Corp, Thorold, Ontario, Canada) according to the manufacturer’s recommendations.
We collected 9 surgical samples in total, 6 WD-GC samples and 3 SRC-GC samples, and we found that 1 WD-GC sample was severely degraded. Ultimately, we compared 5 WD-GC samples with 3 SRC-GC samples (Figure 1D-F). We performed transcriptome sequencing, and we compared expression profiles between clinically comparable samples using transcriptome resequencing data. mRNA sequencing was performed with next-generation sequencing technology by Macrogen, Inc. (Seoul, Republic of Korea). We performed gene enrichment and functional annotation analysis using the Database for Annotation, Visualization and Integrated Discovery (DAVID) bioinformatics resources (https://david.ncifcrf.gov) with the Ingenuity Pathway Analysis system (Qiagen, Hilden, Germany), and we determined the top 10 different genes in terms of the biologic process, cellular component, and molecular function categories and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway differences between SRC-GC and WD-GC (Figure 2).19 (link)
We generated a schematic illustration of the results of the cancer driver variant analysis. We filtered 170,602 variants and 16,460 genes, and we ultimately extracted 52 variants and 30 cancer driver genes. We performed variant analysis and network analysis with Insilicogen Inc. software (Yongin-si, Korea). We found 16,460 genes with 170,602 variants by comparing IGC (A group) and DGC (B group) samples. The genes were filtered by the 1000 Genomes Project, ExAC, NHLBI ESP exomes (with the parameters African American and European American), and Allele Frequency Community (with the parameters common variants and allele frequency below 0.1) resources. Common variants were filtered to include only pathogenic variants (according to the ACMG guidelines classification). We selected more than 2/3 of the samples in the SRC-GCB group and more than 2/5 of the samples in the WD-GC group and filtered the results by biological context (metastasis, signet ring adenocarcinoma or signet ring cell primary gastric adenocarcinoma). Finally, we found cancer driver variants with a frequency greater than 0.01% in the COSMIC and TCGA databases (Figure 3; Table 1).
Publication 2021

Most recents protocols related to «C-src Genes»

To investigate the effects of UC-MSC-CM on TGF-β1-induced brosis in HEFs, HEFs, originally cultured in medium supplemented with serum, underwent serum deprivation for 24 h in HEF-speci c serum-free medium. Following this, HEFs were exposed to 5 ng/mL of TGF-β1, either independently or in conjunction with UC-MSC-conditioned medium (CM) at concentrations of 100, 250, or 500 µg/mL. This research aimed to explore the in uence of UC-MSC-CM on TGF-β1-induced brosis in HEFs.
Real-time quantitative polymerase chain reaction (RT-qPCR)
Total RNA was extracted from the HEFs using the TRIzol reagent (Ambion, Carlsbad, CA). Then, an equal amount of RNA (1 µg) was reverse-transcribed into cDNA utilizing the ReverTra Ace qPCR RT Master Mix Kit (TOYOBO, Osaka, Japan), in accordance with the manufacturer's protocol. All RT-qPCR reactions were performed using a Roche Light Cycler 96 instrument (Roche) with Faster-Start Essential DNA Probes Master Mix (Roche). The mRNA levels of all genes were normalized to those of GAPDH. The speci c primers for the collagen1A1 (Hs00164004_m1), bronectin (Hs01549976_m1), α-smooth muscle actin (α-SMA) (ACTA2, Hs00426835_g1), MKL1 (MRTF-A, Hs01090249_g1), SRF (Hs01065256_m1), RHOA (Hs00357608_m1), ROCK1 (Hs01127701_m1), ROCK2 (Hs00178154_m1), SRC (Hs01082246_m1), YAP1 (Hs00902712_g1), WWTR1 (Hs00210007_m1), and GAPDH (Hs03929097_g1) genes were purchased from Applied Biosystems (Foster City, CA).
Publication 2024
After the 24-h exposure to 10 nM of VGVAPG and 10 nM of VVGPGA peptides and in co-treatment with the c-SRC inhibitor I, samples of total RNA were extracted from the analyzed cell lines according to the manufacturer's protocol. The RNA quality and quantity were determined spectrophotometrically at 260 nm and 280 nm (ND/1000 UV/ Vis; Thermo Fisher NanoDrop, USA). Two-step real-time RT-PCR was conducted using the CFX Real Time System (BioRad, USA). The reverse transcription (RT) reaction was performed at a final volume of 20 µL with 800 ng of RNA (as a cDNA template) using the cDNA reverse transcription kit according to the manufacturer's protocol. Products from the RT reaction were amplified using the FastStart Universal Probe Master kit with TaqMan probes as primers for specific genes encoding cMYC, KI67, PPARγ, GAPDH, NFKB2, ELANE, and MTOR according to the manufacturer's protocol. Amplification was carried out in a total volume of 20 µL containing 1.0 µL of the RT product, and GAPDH was used as a reference gene.
Publication 2024
Histopathologically, lung cancer divides into non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC). Current research demonstrates significantly elevated CEMIP mRNA and protein levels in NSCLC or SCLC cells compared to adjacent normal tissues. High CEMIP expression correlates with poor survival among NSCLC patients (Tang et al., 2019 (link)). Manipulating CEMIP levels in NSCLC lines—either knocking it out or overexpressing it—led to respective decreases or increases in the expression of epithelial-mesenchymal transition (EMT) marker genes, mediated by PI3K-Akt signaling (Tang et al., 2019 (link)). CEMIP promotes SCLC cell migration, proliferation and invasion (Li et al., 2020 (link); Li et al., 2023 (link); Mo et al., 2023 (link)).
Moreover, CEMIP’s hyaluronidase activity leads to the depolymerization of high molecular weight hyaluronic acid into low molecular weight forms. The accumulation of low molecular weight hyaluronic acid activates its receptor TLR2, recruiting c-Src and activating ERK1/2 signaling, thus promoting F-actin rearrangement and SCLC cell migration and invasion (Li et al., 2023 (link)).
Studies also reveal that CEMIP disrupts the interaction between FBXW7, an E3 ubiquitin ligase, and c-Myc in SCLC cells (Mo et al., 2023 (link)). This interference reduces the ubiquitination level of c-Myc, leading to its stabilization and increased nuclear accumulation. Through its indirect regulation of c-Myc, CEMIP promotes glutamine-dependent proliferation in SCLC cells.
Publication 2024
Primary murine and human AEC were infected with PR/8 at MOI 0.5, as described previously11 (link). A/PR8 was diluted in PBS-/- containing BSA and was added to the cells for 1 h, until the inoculum was removed and changed to infection medium (DMEM supplemented with BSA, pen/strep, L-Glutamine and trypsin) for further incubation. For co-culture experiments, AEC were seeded first, allowed to reach confluence, and were infected with A/PR8 or mock infected. 24 h p.i., BMDM were flow-sorted from the BALF of A/PR8-infected mice (BMDM1 were sorted at D7 p.i., BMDM2 were sorted at D21 p.i.) and added directly to the monolayer for 24 h. Cells were stained with Annexin V for apoptosis and Ki67 for proliferation assays and quantified by FACS. For western blot analysis, murine AEC were cultured in medium DMEM supplemented with pen/strep, L-glutamine and 10% heat-inactivated FCS. Cells were incubated at 37 °C for 5 days and thereafter treated with rPLET1 (40 ng/ml) for 12 h. Cell lysates were prepared in the same manner, as those used for the kinase activity assay described below. For the proliferation assays, murine AEC were cultured in DMEM enriched with pen/strep, L-glutamine and 2% FCS. Cells were incubated at 37 °C for 4 days and thereafter treated with rPLET1 (40 ng/ml) and/or MEK inhibitor U0126 (Promega, #V1121, 10 µM) for 12 h. For qPCR analysis, murine AEC were cultured in DMEM, pen/strep, L-glutamine and 10% FCS, after reaching the confluence they were infected with PR/8 at MOI 0.5, as described previously11 (link) and treated with rPlet1 (20–40 ng/ml) and/or Src activator (Santa Cruz Biotechnology, Cat No #sc-3052, 10 µM) for 12 h. Cells were stored RLT buffer and stored in –80 °C for further qPCR analyses. For qPCR analyses of Src family-related genes, murine AEC were cultured as described above and treated with rPlet1 for 1 and 2 h. Cells were taken into RLT buffer and stored in –80 °C for qPCR analysis. In the stimulation assay of TR-AM with the AEC supernatant, AEC were cultured as mentioned above. When the cells reached confluence, they were infected with PR/8 at MOI 2.0 or mock infected, as described above and incubated at 37 °C for 12 h/24 h. The supernatant of these cells was used to stimulate the TR-AM (300,000 cells/cm2 in a 24-well plate, seeded in RPMI medium supplemented with pen/strep, L-glutamine, 2% FCS and 2.5% HEPES, 6 h prior to the treatment) for 12 h. The supernatant of TR-AM cultures was used for soluble Plet1 quantification by ELISA.
Publication 2024
Full-length human PANX1 in the pEGC Bacmam vector from our previous study was used(14 (link)). Mouse PANX1 (UniprotID: Q9JIP4) was synthesized by GenScript and subcloned into the pEGC Bacmam vector(45 (link)). The translated product contains the human or mouse PANX1 protein, a thrombin digestion site (LVPRGS), an enhanced GFP protein, and an 8x His tag. pCMV5 mouse Src was a gift from Joan Brugge & Peter Howley (Addgene plasmid # 13663; http://n2t.net/addgene:13663; RRID:Addgene_13663). The mouse Src gene was subcloned into the pEGN Bacmam vector(45 (link)). The translated product contains a mCherry protein, a thrombin digestion site, and the Src protein. Primers for site-directed mutagenesis were designed using QuikChange Primer Design website (https://www.agilent.com/store/primerDesignProgram.jsp) and synthesized by Eurofins Genomics. The QuikChange mutagenesis protocol was used to generate all the mutants of the study. Sanger sequencing was performed to identify positive clones.
Adherent HEK293T (ECACC, Catalogue Number: 96121229) cells were grown in DMEM media supplemented with 10% fetal bovine serum. Transient transfection was conducted using lipofectamine-2000 by following the manufacturer’s protocol. Specifically, the cells were cultured in 60mm Petri dishes until 80% confluency. Transfection solution was made by mixing 500ng of plasmid DNA, 4uL of lipofectamine-2000 reagent, and 100uL Opti-MEM media. After 10min incubation at room temperature, the DNA-lipid complexes were added to the cell culture and incubated at 37°C. The next day, 10mM sodium butyrate was added to the cells to boost protein expression. The cell culture was then grown at 30°C for another day before harvesting. The cell pellet was flash-frozen with liquid nitrogen and stored at −80°C. For co-transfection experiments, equal amounts of hPANX1 (250 ng) and mSrc (250 ng) plasmids were used in the transfection mixture. For Neuro2A cell culture (ATCC, Catalogue Number: CCL-131), EMEM media is used instead of DMEM. All the other procedures described above are the same for expressing proteins in Neuro2A cells.
For the western blot experiment, the cell pellet was lysed in TBS buffer (20 mM Tris pH 8.0, 150 mM NaCl) with protease an inhibitor cocktail (1 mM phenylmethylsulfonyl fluoride, 2 mM pepstatin, 0.8 μM aprotinin and 2 μg/ml leupeptin), 1mM sodium orthovanadate and 1% glycol-diosgenin detergent for 30 min on ice. The lysate was clarified by centrifugation at 21,000rpm for 20 min and the soluble portions were mixed with 2x SDS loading buffer supplemented with 5% 2-Mercaptoethanol. The samples were resolved in precast gradient SDS gel (4–20%) or 7.5% PhosTag gel. A Chemidoc instrument was used to directly image the in-gel fluorescence signal after electrophoresis. Subsequently, the protein in the gel was transferred to the nitrocellulose membrane using the semi-dry transfer buffer (48 mM Tris base, 39 mM glycine, 20% methanol). The membrane is blocked in the TBST buffer (20 mM Tris pH8.0, 150 mM NaCl, 0.1% Tween 80) with 4% non-fat milk for 1 h at room temperature. Afterward, primary antibodies (1:2000 dilution) were incubated with the membrane overnight at 4°C. The next day, the membrane was washed with TBST buffer for 4 times, 10 min each before goat anti-rabbit IgG secondary antibodies were added (1:25000 dilution). After 1h, the secondary antibodies were discarded and the membrane was washed again with TBST for 4 times, 10 min each. The western blot signal was detected using ECL Pierce substrate and imaged using a Chemidoc instrument. A brightfield image was overlaid with the illuminance signal to visualize the position of protein markers in the membrane. The anti-PANX1 antibody is obtained from abcam (Catalogue number: ab124131). The anti-Src (Catalogue number: 36D10) and anti-pY100 (Catalogue number: 9411) antibodies are obtained from Cell Signaling. The anti-PANX1-pY198 (Catalogue number: ABN1681) and anti-PANX1-pY308 (Catalogue number: ABN1680) antibodies were obtained from Millipore Sigma.
For de-glycosylation experiment, Neuro2A/HEK293T cells expressing designated genes were solubilized in TBS buffer with protease inhibitor cocktail (1 mM phenylmethylsulfonyl fluoride, 2 mM pepstatin, 0.8 μM aprotinin and 2 μg/ml leupeptin), 1mM sodium orthovanadate and 1% glyco-diosgenin detergent for 30min on ice. The samples were clarified by centrifugation at 20,000g for 30 min. The de-glycosylation reaction was made by mixing 16 uL of the supernatant with 2uL of PNGase F enzyme and 2 uL of GlycoBuffer 2 (10X). The control reaction replaced the PNGase F enzyme with water. The reaction was allowed to occur at room temperature overnight. The next day, the samples were mixed with 20 uL 2× SDS sample-loading buffer (Sigma) supplemented with 5% βME and resolved by SDS-PAGE. The gel was imaged in the ChemiDoc system by probing the GFP and mCherry fluorescence signal.
For the dephosphorylation experiment, mSrc-mCherry WT, Y529F, and K297M were expressed in HEK293T cells. The cells were lysed in TBS buffer (20mM Tris pH8, 150mM NaCl) with 1% glyco-diosgenin detergent for 30min on ice. After clarifying the non-soluble debris by centrifugation at 21,000rpm for 20min, 20 μL supernatant was mixed with 2.5 μL of 10X NEBuffer for Protein MetalloPhosphatases, 2.5 μL 10 mM MnCl2 and 2 μL Lambda Protein Phosphatase (NEB). The dephosphorylation reaction was allowed to occur at 30 °C overnight. Control samples without adding Lambda Protein Phosphatase are placed on ice. The next day, the samples were mixed with 2× SDS sample-loading buffer (Sigma) supplemented with 5% βME and resolved by SDS-PAGE or Phos-tag gel. The gel was imaged in the ChemiDoc system by probing the mCherry fluorescence signal or western blot analysis.
For in vitro phosphorylation and mass-spectrometry analysis, the hPANX1 WT or mutants were expressed alone or co-expressed with mSrc Y529F mutant in HEK293T cells. Specifically, 50 μg of plasmid DNA (in the case of the co-expressing experiment, 25 μg of human PANX1 plasmid and 25 μg of mouse Src Y529F mutant plasmid was used) was mixed with 150ug of PEI 25K (Polysciences) in a and incubated at room temperature for 30 min. The DNA-PEI complex was then added to the suspension cell culture of HEK293T cells at a density of 2×106 cells/ml. After growing at 37 °C for 8 h, 10 mM sodium butyrate was added, and the temperature was changed to 30°C for 40 h. The cells were then harvested and stored at −80 °C until purification. The protein purification procedure was described in our previous study(14 (link)).
The in vitro phosphorylation is conducted using the human Src-GST protein (Sigma-Aldrich, Catalogue number: S1076) according to the manufacturer’s protocol. Specifically, 1.5 μg of purified PANX1 protein is mixed with kinase assay buffer (25 mm MOPS, pH 7.2, 20 mM MgCl2, 12.5 mM MnCl2, 5 mM EGTA, 2 mM EDTA, 0.25 mM DTT), diluted 1:5 with 50 ng/μl bovine serum albumin (BSA). The reaction was supplemented with 0.25 mM ATP, and 0.3 μg recombinant human (active) Src-GST kinase. Samples were incubated for 1 h at 30 °C using a PCR thermocycler. After 1 h, 2x SDS buffer is mixed with the sample to stop the reaction. The sample is then resolved by an SDS-PAGE gel for subsequent western blot analysis.
Purified hPANX1 protein w- or w/o- co-expressing mSrc Y529F mutant was resolved in SDS-PAGE gel and the band corresponding to hPANX1 protein was cut and subjected to in-gel digestion with trypsin. Half of each digested sample was analyzed by nano LC-MS/MS with a Waters M-Class HPLC system interfaced with a ThermoFisher Fusion Lumos mass spectrometer. Peptides were loaded on a trapping column and eluted over a 75μm analytical column at 350nL/min; both columns were packed with Luna C18 resin (Phenomenex). The mass spectrometer was operated in data-dependent mode, with the Orbitrap operating at 60,000 FWHM and 15,000 FWHM for MS and MS/MS respectively. The instrument was run with a 3 s cycle for MS and MS/MS.
Data were searched using a local copy of Mascot (Matrix Science) with the following parameters. Enzyme: Trypsin/P; Database: SwissProt Human (concatenated forward and reverse plus common contaminants); Fixed modification: Carbamidomethyl (C) Variable modifications: Oxidation (M), Acetyl (N-term), Pyro-Glu (N-term Q), Deamidation (N/Q); Mass values: Monoisotopic; Peptide Mass Tolerance: 10 ppm; Fragment Mass Tolerance: 0.02 Da; Max Missed Cleavages: 2. Mascot DAT files were parsed into Scaffold (Proteome Software) for validation, filtering and to create a non-redundant list per sample. Data were filtered using 1% protein and peptide FDR and requiring at least two unique peptides per protein.
TsA201 cells were transfected using Lipofectamine 2000 (Thermo Fisher) according to the manufacturer’s protocol. The transfected cells were incubated at 37°C for 18–24 hours before electrophysiological measurements. Whole-cell recordings were performed using a Multiclamp 700B (Axon Instruments) and Clampex software, with pipettes of 3–5 MOhm resistance filled with an internal solution containing 145 mM NaCl, 10 mM Hepes, 10 mM EGTA, pH adjusted to 7.4. The external bath solution contained 160 mM NaCl, 10 mM Hepes, 3 mM KCl, 2 mM CaCl2, and 1 mM MgCl2, also adjusted to pH 7.4. During the recordings, voltage steps ranging from −100 mV to +140 mV were applied, each lasting 100 ms with a 20 mV increment between steps, and membrane currents were digitally recorded at 10 kHz and filtered at 2 kHz. To precisely measure the PANX1 channel's current, carbenoxolone disodium salt (CBX), a blocker of the channel, was added to the bath solution at a final concentration of 0.1 mM, and CBX-sensitive currents were subsequently calculated by comparing the difference in current amplitude in a cell with and without the presence of CBX.
Publication Preprint 2024

Top products related to «C-src Genes»

Sourced in United States, China, United Kingdom, Germany, Australia, Japan, Canada, Italy, France, Switzerland, New Zealand, Brazil, Belgium, India, Spain, Israel, Austria, Poland, Ireland, Sweden, Macao, Netherlands, Denmark, Cameroon, Singapore, Portugal, Argentina, Holy See (Vatican City State), Morocco, Uruguay, Mexico, Thailand, Sao Tome and Principe, Hungary, Panama, Hong Kong, Norway, United Arab Emirates, Czechia, Russian Federation, Chile, Moldova, Republic of, Gabon, Palestine, State of, Saudi Arabia, Senegal
Fetal Bovine Serum (FBS) is a cell culture supplement derived from the blood of bovine fetuses. FBS provides a source of proteins, growth factors, and other components that support the growth and maintenance of various cell types in in vitro cell culture applications.
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, 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, Germany, China, Japan, United Kingdom, Canada, France, Italy, Australia, Spain, Switzerland, Belgium, Denmark, Netherlands, India, Ireland, Lithuania, Singapore, Sweden, Norway, Austria, Brazil, Argentina, Hungary, Sao Tome and Principe, New Zealand, Hong Kong, Cameroon, Philippines
TRIzol is a monophasic solution of phenol and guanidine isothiocyanate that is used for the isolation of total RNA from various biological samples. It is a reagent designed to facilitate the disruption of cells and the subsequent isolation of RNA.
Sourced in United States, Germany, United Kingdom, Japan, Lithuania, France, Italy, China, Spain, Canada, Switzerland, Poland, Australia, Belgium, Denmark, Sweden, Hungary, Austria, Ireland, Netherlands, Brazil, Macao, Israel, Singapore, Egypt, Morocco, Palestine, State of, Slovakia
The High-Capacity cDNA Reverse Transcription Kit is a laboratory tool used to convert RNA into complementary DNA (cDNA) molecules. It provides a reliable and efficient method for performing reverse transcription, a fundamental step in various molecular biology applications.
Hexadimethrene bromide is a cationic polymer compound commonly used as a laboratory reagent. It functions as a flocculating agent, facilitating the aggregation and sedimentation of suspended particles in aqueous solutions.
Sourced in Germany, United States, United Kingdom, Netherlands, Spain, Japan, Canada, France, China, Australia, Italy, Switzerland, Sweden, Belgium, Denmark, India, Jamaica, Singapore, Poland, Lithuania, Brazil, New Zealand, Austria, Hong Kong, Portugal, Romania, Cameroon, Norway
The RNeasy Mini Kit is a laboratory equipment designed for the purification of total RNA from a variety of sample types, including animal cells, tissues, and other biological materials. The kit utilizes a silica-based membrane technology to selectively bind and isolate RNA molecules, allowing for efficient extraction and recovery of high-quality RNA.
Sourced in United States, China, United Kingdom, Germany, France, Australia, Canada, Japan, Italy, Switzerland, Belgium, Austria, Spain, Israel, New Zealand, Ireland, Denmark, India, Poland, Sweden, Argentina, Netherlands, Brazil, Macao, Singapore, Sao Tome and Principe, Cameroon, Hong Kong, Portugal, Morocco, Hungary, Finland, Puerto Rico, Holy See (Vatican City State), Gabon, Bulgaria, Norway, Jamaica
DMEM (Dulbecco's Modified Eagle's Medium) is a cell culture medium formulated to support the growth and maintenance of a variety of cell types, including mammalian cells. It provides essential nutrients, amino acids, vitamins, and other components necessary for cell proliferation and survival in an in vitro environment.
Sourced in Japan, China, United States, Germany, Canada, Switzerland
The SYBR Green kit is a reagent used for the quantification of DNA and RNA in real-time PCR (polymerase chain reaction) experiments. It is a fluorescent dye that binds to double-stranded DNA, enabling the detection and measurement of the amplified target sequence during the PCR process.

More about "C-src Genes"

C-src genes, also known as cellular-src or proto-oncogene Src, are a family of genes that encode non-receptor tyrosine kinases.
These kinases play a crucial role in cellular signaling pathways, regulating processes such as cell growth, differentiation, and motility.
Alterations or dysregulation of c-src genes have been implicated in the development and progression of various cancers, making them an important target for research and therapeutic interventions.
Understanding the function of c-src genes is essential for advancing cancer research and developing effective treatments.
Researchers often utilize techniques like cell culture, transfection, and gene expression analysis to study the role of c-src in cellular processes.
Common reagents used in these studies include FBS (Fetal Bovine Serum) for cell culture, TRIzol or the RNeasy Mini Kit for RNA extraction, and Lipofectamine 2000 or Hexadimethrene bromide for transfection. qRT-PCR (Quantitative Reverse Transcription Polymerase Chain Reaction) is a widely used method for analyzing c-src gene expression levels.
Researchers may employ the High-Capacity cDNA Reverse Transcription Kit and SYBR Green kits to quantify c-src mRNA levels in various cell lines or tissue samples.
DMEM (Dulbecco's Modified Eagle Medium) is a commonly used cell culture medium for maintaining and propagating cells in these studies.
By exploring the complexity of c-src genes and their involvement in cellular signaling pathways, researchers can uncover new insights into the mechanisms of cancer development and progression.
This knowlege can then be leveraged to develop innovative therapeutic strategies targeting c-src, ultimately improving patient outcomes and advancing the field of oncology.