Science method
CRISPR - Science method
Explore the latest questions and answers in CRISPR, and find CRISPR experts.
Questions related to CRISPR
hey,
im designing an experiment to optimise prime and base editing epeg/sg RNAs based on their efficiency and off target effects. I understand that using a cell line easy to transfect such as HEKs are ideal for this, and then select the best rna and use in your target cell. But I will need to insert the desired mutations in the HEK cells first… I thought doing the reverse prime/base editing was a bit of a long procedure just to correct the mutation again, so wanted to transduce them with a lenti carrying the gene with the mutation. This gene however does not fit into a lentivirus since it’s quite big, would inserting just a region of the gene that contains the mutations along homology arms (of1kb or how long?) be enough to validate genomic edit? ofc I wouldn’t be able to validate anything at the protein level but just to assess the efficiency at the gene level. has anyone done this or know of a publication which has? Or any reference that might suggest it’s a valid experiment? What do you think? thanks!!!
Dear all, I am studying A GENE influence viral replication and translation OR NOT. After knocking out THAT GENE by CRISPR, I infected the cells (both WT and KO) with virus at MOI of 0.02 and 0.2 . 3 days later, I collected the viral supernatant for TCID50 and extracted total RNA of supernatant and cell pellet (Mixture) by TRIzol. TCID50 showed that virus titer increased in KO groups, however, qPCR showed that virus copies decreased in KO groups. I am so confused which result was convincing, thank you all for kindly help.
Hello everyone,
I'm facing some issues in producing lentivirus from my CRISPR/cas plasmid. Briefly, I used a plasmid encoding for my guide RNA, the Cas9, and puromycin resistance gene.
However, after transfection of the transgene, packaging plasmids, and VSV-G on HEK293T cells no lentivirus seems to be produced. I titrated supernatants from different days post-transfection on the final cell line in which I needed to do my experiments by serial dilution and 1ug/ml puromycin treatment, but they died at every dilution (even when I added pure supernatants).
I also did GFP-expressing lentivirus productions in parallel, which however resulted in a good transduction.
I can't get why I see such a difference in the production of control and transgene-expressing lentivirus, since the protocol was the same. At the very least I should see the same amount of surviving cells after puromycin treatment as the green cells in the control transduction for the same dilution, which is not the case. For transgene lentivirus, I used the backbone plasmid of our collaborator, which they routinely use in the laboratory to produce lentiviruses, and successfully cloned the sgRNA required for my CRISPR editing.
Thank you to anyone who would help me on this matter.
Hi all,
I have been designing my CRISPR Cas9 experiment. I have 2 plasmids one has gRNA and EGFP, the other one has Cas9. I will do co-transfection for these 2 plasmids to get KO. I will sort my GFP positive cells via FACS and I plan to sort cells also for Cas9 protein. Is that a good idea to sort the cells for both cas9 and GFP? (I will use hIPS cells) otherwise how can I select the transfected cells which carry both plasmid in. (I don't use antibiotic selection since my plasmids don't have mammalian antibiotic resistance gene. Also my cells are quite sensitive)
Can you help me?
I am seeking insights into the latest advancements and techniques that can enhance the specificity of CRISPR-Cas9. Specifically, I am interested in understanding the strategies that have been shown to effectively reduce off-target effects while maintaining the efficiency of the gene-editing process.
What is CRISPR? CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats) is a revolutionary gene-editing tool that allows scientists to make precise changes to DNA. It works with a guide RNA that directs the Cas9 enzyme to a specific location in the genome, where it can cut the DNA and enable modifications.
I am currently working on crispr12a with 6x histtaged protein... i am using extraction buffer with with nacl, hepes and protease inhibitor....and in purification iam using nacl, hepesand immidazole
.. but the problem is when i use low conentratuon of immidazole (20-50mM) all the other proteins also come out but when i use highr concentration even crispr is also wasged in the washing step and doesnot bind with the ni-NTa column...can anyone suggest any modification in protocol
CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats) technology has revolutionized the field of genetic engineering by providing a precise, efficient, and relatively easy method for gene editing. Unlike traditional genetic modification methods, which often rely on random insertion of genetic material or less precise techniques, CRISPR allows for targeted modifications at specific locations within the genome.
I was wondering if it is possible to form a permanent open "ssDNA bubble" similar to a transcription bubble (>13 nucleotides) within E. coli. These criteria are important:
1. Open ssDNA bubble within replicable (in E. coli) genetic element. So no C-Traps under force.
2. No proteins, nucleic acids, or other toxic chemicals supporting the bubble. Can help during nucleation, but bubble has to be accessible for protein interaction.
3. Stable in bioorthogonal conditions. Physiological pH, salt, 37 °C, etc.
I am brand new to CRISPR/Cas9 and would be very appreciative of input on my experimental plan/design!
For context, I want to manipulate 4 cancer-associated genes - APC, TP53, SMAD4, and KRAS - in colonic organoid cells. I will transfect cells when they are in a single-cell suspension during passaging. I wish to knock out APC, TP53 and SMAD4, and introduce a G12D mutation in KRAS. These manipulations have been done previously in colonic organoids via plasmid lipofection (Hans Clevers et. al, Nature 2015). The authors first introduce the G12D mutation and select for KRAS mutants by removing EGF and adding the antibiotic gefitinib to culture media. They then introduce the triple APC, TP53, SMAD4 KO and select mutants using culture media -WNT -Noggin +Nutlin-3.
I, rather, wish to multiplex the G12D mutation and triple KO into one experiment, use electroporation instead of lipofection, and transfect ribonucleoproteins (RNPs) and my oligonucleotide donor DNA sequence to avoid issues associated with plasmid use.
For my experimental design so far:
sgRNA design:
So far, I have used ChopChop and Synthego for design of sgRNAs . I plan on selecting ~3 sgRNAs per gene to ensure that my genes of interest are actually knocked out. The Clevers paper also includes the sequences of the gRNAs they used, so this is another option. As I plan on using Synthego's SpCas9 2NLS Nuclease, sgRNAs suggested by Synthego are compatible with PAMs recognized by this protein, so I am leaning towards using these.
Oligonucleotide design for KRAS G12D mutation:
Clevers et. al target KRAS with a sgRNA and introduce a GGT to GAT mutation in Exon 1 of KRAS using donor DNA. As I wish to make the exact same mutation, I plan on using their same KRAS target sequences: number 1, 5′-GAATATAAACTTGTGGTAGTTGG-3′; number 2, 5′-GTAGTTGGAGCTGGTGGCGTAGG-3′ and donor DNA oligo 5'-CTGAATATAAACTTGTGGTAGTTGGAGCCGATGGAGTAGGCAAGAGTGCCT-3' (Figure 2c).
Cas9 selection:
From what I understand, I will need to select a Cas9 protein that is capable of recognizing PAM sequences in each of my target genes. The KRAS PAM sequence desribed by Clevers et al is AGG, so my plan is to use Synthego's SpCas9 2NLS Nuclease which recognizes 5'-NGG-3' PAMs and design my other sgRNAs so they target sequences ~20 nucleotides upstream of any 5'-NGG-3' PAM sequence.
Protocol:
I plan to use the publicly available protocol from the Corn Lab (Cas9 RNA nucleofection for cell lines using Lonza 4D Nucleofector V.1) and the Lonza nucleofector.
Any feedback on what I have outlined above would be greatly appreciated and I am happy to provide any more info as needed!
I am a member of the iGEM team 2014, Goteborg, Sweden. In our project we are trying to build a "yeast age counter", i.e. a synthetic circuit that expresses a different fluorescent protein according to the replicative lifespan of a yeast cell.
Without going too much into detail in the circuit design, one of our biggest issues is to make sure that the gRNA transcript, produced during the late G1 phase, "survives" through a whole cell cycle until the next G1 phase. In yeast the generation time is around 90 minutes and the above mentioned transcript is without the poly (A) tail so we are confident it will accumulate in the nucleus.
The main question is: will the transcript survive long enough or will it be degraded within one cell cycle?
Currently in our lab, we are trying to knockdown EGFR gene using Crispr-Cas9 system. We purchased ThermoFisher's GeneArtTM Precision gRNA Kit and followed their user's guide. However, when we are performing the very first step of synthesizing DNA template for gRNA using PCR, we got nearly no product for our desired product (around 110bp), which we already figured that out as we made a mistake when designing our primers. However, we have no idea why our control group (primers provided by ThermoFisher) has two bands when there was only supposed to be one band. We repeated the experiment, this time we even got an extra band in control group compared to the first time, which is super weird. I have attached the pictures of what the band is supposed to look like and our first and second PCR results. The lowest ladder on DNA marker is 100 bp. Can someone share their opinions on this issue? I appreciate it!
glucagon biogenesis
glucagon secretion
siRNA
CRISPR
overexpression
Hey guys,
So I am trying to make a fluorescent reporter line in ES cells for a particularly AT rich region of DNA. Since there are no NGG sequences around where I was to cut I cannot use the Cas9 system so I've had to use TALEN pairs instead. Unfortunately I am not getting any colonies. I recently saw publications from the Zhang lab about the new Cpf1 protein and that it cuts in AT rich regions and produces a staggered sticky end cut.
Has anyone tried this new protein? I feel like I am running out of options and electroporate cells every week with no success. Any help would be greatly appreciated!
Thanks!
EDIT 6/25/18
In case anyone is following this and interested, I did manage to make a knock-in reporter line using this system. The efficiency of cutting is a bit lower in comparison to cas9 (our highest Cpf1 guide cut 15% in 293T cells using TIDE analysis) but we picked enough clones that we got a line. It also helped that we used a homologous recombination enhancing drug called RS-1 at a concentration of 7.5uM on the day of electroporation for 24hrs. This was done in an iPS line.
I have used ssDNA oligo and plasmids for HDR. I am wondering whether anyone has tried using PCR product directly as HDR template.
Good day! The question is really complex since CRISPR do not have any exact sequence - so the question is the probability of generation of 2 repeat units, each of 23-55 bp and having a short palindromic sequence within and maximum mismatch of 20%, interspersed with a spacer sequence that in 0.6-2.5 of repeat size and that doesn't match to left and right flank of the whole sequence, in a random sequence.
Hi,
I'm creating a knockout for a gene that codes for a nuclear protein. I designed guideRNAs(4 gRNA; two targeting exon 1 and another two targeting exon 2) using software by crispr.mit and uses lentiCRISPR(single vector) plasmid for transfection in HEK293T cells. I harvested the virus and infected HEK293T cells. I manage to get puromycin resistant cells after transfection for all of the gRNA (which is done individually in different plate) with cell death rate not more then 10%. I then continuously grow and passage them in puromycin and uses total cell lysate to extract the protein before proceeding with Western Blot. I failed to gain 100% knockout for all of the gRNA but two of it have slightly fainter bands. The gene have about 3 to 4 copy number in the cells, which might cause non-homozygous deletion for the gene, but I'm not doing single cell cloning since the work is tedious and time costly. The gene is not listed in the essential gene list, and even if the gene is essential, I should see a lot of cell death in puro selection, but I didn't. After gRNA design, I actually checked the gRNA for off targets using Crispr OFF-Finder and checked for secondary structures(hairpin) and self complementarity (5 base pairs) and all of them is fine, no secondary structures and least exonic off targets. So, assuming CRISPR system is very reliable, I'm not sure if the failed knockout is due to non-homozygous deletion (there are other paper that use single gRNA targeting a 4 copy numbered gene in HeLa and is successful), or because of my gRNA is not efficient (I did not use surveyor assay to check for mutation since that assay involves trying it in HEK293 cells too) or the gene is mutated but is repaired very efficiently or etc, I don't know. Any ideas?
I have been running a MAGeCK test command on the terminal for a CRISPR screen to rank sgRNAs and genes based on the read count tables. However, I get only the plot of the top-ranked genes (positively selected) but not the negatively selected ones. On the terminal, I get this error:
INFO  @ Mon, 29 Jul 2024 11:17:57:   Error in plot.window(...) : Logarithmic axis must have positive limits
INFO  @ Mon, 29 Jul 2024 11:17:57:   Calls: plotrankedvalues -> plot -> plot.default -> localWindow -> plot.window
INFO  @ Mon, 29 Jul 2024 11:17:57:   In addition: Warning message:
INFO  @ Mon, 29 Jul 2024 11:17:57:   In xy.coords(x, y, xlabel, ylabel, log) :
INFO  @ Mon, 29 Jul 2024 11:17:57:     1 y value <= 0 omitted from logarithmic plot
INFO  @ Mon, 29 Jul 2024 11:17:57:   Execution halted
I would be very thankful if someone with a similar experience could help solve this issue.
Thank you,
Hi All,
I'm currently trying to perform a CRISPR-mediated knock-in of a fluorophore in the middle of a receptor channel. I'm basing the protocol off of Siedman's paper:
My first question would be: Has anyone had any experience in using circular donor plasmids for HR? We have always used ssODNs, but since the segment we will be making is too long (2kb), we will be unable to order an ssODN. Other papers I've looked have often use other gRNAs to cut on either side of the gBlock, liberating the gene to be inserted along with homology arms flanking either side. However, it seems that due to the blunt-ended nature of the dsDNA cut out, there is a high off-target incorporation. Siedman's paper seems to have just left the homology arms flanking the gene of interest in the actual plasmid itself. Has anyone else done this before?
My second question: How important is homology arm symmetry. Currently, the two homology arms flanking my KI gene is asymmetrical. Downstream of the insertion site, there is a CT-right tandem repeat segment, that cannot be incorporated into the gBlock, leaving the right homology arm at 170bp, vs 500bp for the left homology arm. What do you guys think? It seems that asymmetrical homology arms are better for ssODNs, but I'm not sure about in a circular donor plasmid?
Would be great if someone could help me out! Thanks so much!
I am planning to do gene knockout for human cell line using all in nonviral plasmid which has both the GFP and the Puromycin resistance gene. I am wondering which one is better to start with, the cell sorting with GFP and then The Puromycin selection, or the opposite?
CRISPR/Cas
#Pancreatic Cancer
#Biotechnological Engineering
#Biotechnology
#Molecular Cloning
#Gene Knockout
#Genetic Engineering
#CRISPR/CAS9
#CRISPR
I am looking for a CRISPR protocol for application in leishmania strains
I'm working on a specific tissue knock in by CRISPR.
Hello everyone
I'm using CRISPR technology to edit a mutated gene in human cells. I used 2 gRNAs, which resulted in the deletion of about 300bp between the two guide RNAs. This was confirmed by gel analysis and Sanger sequencing. To confirm my results further, I sent them for ONP sequencing to get the percentage of deleted sequences. I counted the number of deletions and divided it by the total number of reads. Is this correct? I used the alignment and amplicon tools in epi2me, and I did not get any useful results. Any help is appreciated.
I am currently investigating the CRISPR 9 case to target my gene of interest for knockout. The vector being used is lentiCRISPR v2puro. I digested and dephosphorylated 5ug of the lentiviral CRISPR plasmid with BsmBI,Subsequently, I ran a gel electrophoresis as shown in the attached image.
Following this, I excised the larger band and stored it at -20°C overnight. The next step involved purifying the gel using a Promega kit. During the purification process, I added 500ul of membrane binding solution, briefly vortexed the mixture, and then incubated it on a heating block at 88°C for approximately 25 minutes until the gel slice completely dissolved.
The DNA concentration for 5ug was measured at 30.144 ng/μl, with a 260/280 ratio of 1.70 and a 260/230 ratio of 1.26.
Next, I performed phosphorylation and annealing of each pair of oligos, which were then diluted to a 1:200 ratio. The two target sequences are as follows:
- gCTATGTGGTCGGAGAAACGT (I added "g" as it did not start with "g"). For the forward primer, CACC was added to the 5' end, and for the reverse primer, AAAC was added to the 5' end.Forward: CACCGCTATGTGGTCGGAGAAACGT Reverse: AAACACGTTTCTCCGACCACATAGC
- GTTTTGGTTCAGACTCGAGG (I did not add "g" as it already starts with "g").Forward: CACCGTTTTGGTTCAGACTCGAGG Reverse: AAACCCTCGAGTCTGAACCAAAAC
I have heard that it is recommended to add "g" to target sequences even if they already start with "g", as omitting it could result in a lack of colonies after transformation. is that correct ?
The third step involved ligating the sequences using Quick Ligase. In the ligation reaction, a 1:3 ratio was used, so 2.2 ul of vector and 6.8 ul of insert DNA were added, along with 10ul of ligation reaction buffer and 1ul of Quick Ligase, making the total reaction volume 20ul. The mixture was then incubated at room temperature for 1 hour before adding 50 ul of DH5a cells. Heat shock was performed by incubating on ice for 30 minutes, followed by a 30-second incubation in a 42°C water bath and then back on ice for 2 minutes. Subsequently, 250 ul of prewarmed LB media was added, and the mixture was incubated on a shaker at 37°C for 1 hour. The resulting solution was then spread onto LB agar plates containing Ampicillin (100 μg/mL) and incubated in an incubator at 37°C overnight. Colonies were obtained for the positive control using pUC19, but not for the sample.
During this this procedure If anyone has any insights into why colonies were not obtained your input would be greatly appreciated.
Does anyone have a working protocol or suggestion for CRISPR knockout gene in THP-1 cell. I worked for more than half year to knock CFB gene in THP-1 cells. When I transduced the CAS9 into THP-1 at the beginning, after I applied blasticidin 99% cells died, very small portion of cells didn't proliferate, and finally all cells died. I got CAS9 expressing THP-1 cell at the third try. But I still cannot get single cell clone by limited dilution. Because the single cell didn't proliferate, all the cells died later.
It was even exceedingly difficult to transduce the guide RNA into the CAS9 expressing THP-1 cells. I tried twice, all the cells died after apply the selection antibiotics hygromycin. The transduced cells even died faster than the untransduced cells.
Any suggest and help are welcome.
Strain: LBA4404
Growing media: YEB (pH 7)
Temperature: 28°C, 200 rpm (overnight culture)
Antibiotic use: Kanamycin and Streptomycin
Discoveries, Promises, and Perils of the application of CRISPR technologies to insects
CRISPR gene editing technologies have opened the door to disabling, modifying, and replacing genes in a wide range of organisms including insects. Since its first applications in Drosophila more than a decade ago, CRISPR has been used in diverse insect species to study questions that range from gene function and physiology to development and behavior, as well as to explore its utility for pest control. The time is ripe to evaluate the successes, the failures, and the perils associated with the application of these technologies to insects. We seek submissions related to all aspects of gene editing. These include, but are not limited to: insights gained regarding fundamental questions of biology (e.g., physiology, metabolism, development, behavior); application of these technologies to pest control and their ethical and ecological implications; and technical advances (e.g., for efficient gene editing, gene replacement and gene insertion; for identifying and minimizing off-target effects; for spatiotemporal-specific gene editing; for high-throughput functional genomics screen, etc.). We encourage researchers to share their findings, insights, expertise, and precautionary principles on these game-changing technologies, gained from their application to any insect species.
Guest editors:
John J. Ewer - Valparaiso University, Valparaiso, Chile.
David D. Dolezel - Institute of Entomology Biology Centre Czech Academy of Sciences, Prague, Czechia.
Huidong Wang - Henan University, Kaifeng, China. wanghd@henu.edu.cn
Manuscript submission information:
Submission Deadline: 1st October 2024
The journal’s submission platform Editorial Manager® is now available for receiving submissions to this Special Issue. Please refer to the Guide for Authors to prepare your manuscript, and select the 'VSI:The application of CRISPR technologies to insects' article type when submitting your manuscript online. Both the Guide for Authors and the submission portal could be found on the Journal Homepage here: https://www.sciencedirect.com/journal/insect-biochemistry-and-molecular-biology/special-issues
Keywords:
CRIPR-Cas9; gene editing; non-model insects; reverse genetics; genetically-modified organisms; gene-drive systems
Please send us an email (john.ewer@uv.cl, dolezel@entu.cas.cz, wanghd@henu.edu.cn) if you would like to contribute a manuscript
I have transfected neurons using electroporation before playing plasmid with Cas9 which makes a single double stranded cut. But I am not able to see any results. Is there a way I can delete the whole gene using CRISPR in primary neuron culture?
Can anyone share their experience regarding the acquisition or gifting of cell lines mentioned in a Nature article? Specifically, I am interested in knowing if there are any journal-specific policies involved.
The cell line in question contains a CRISPR-mediated stably expressing protein labeled with GFP, and its published in the Nature journal. I intend to use this cell line for my own experiment. Naturally, I am willing to acknowledge or provide authorship as appropriate. However, I would like to know if it is possible to obtain this cell line directly from the PIs lab and what the relevant policies are of nature journals if I get them from PI lab (I understand I will refer the article).
Has anyone ever received or gifted cell lines before? I am aware that exchanging plasmids is a common occurrence, but I have never personally obtained cell lines in this manner.
I appreciate any information you can provide.
Thank you in advance.
Hi,
I recently made a mutant cell line by excising an exon from the gene, DNMT1, using CRISPR-CAS9. I isolated a single cell population that has this mutation and confirmed the mutation using PCR and Sanger sequencing. My PI also wants to use RT-qPCR to show that the sequence is missing in the mRNA. I made 3 sets of primers targetting the exon, so I would only expect amplification in the negative control cell line and not in the mutant line. However, when I ran the qPCR, I got normal amplification of this DNMT1 exon in both the negative control and the mutant line (~ct values around 23 for both).
I've extracted RNA three separate times to make sure I didnt have RNA contamination the times prior, but I still get the same result.
If anyone has experience with this or may have solutions, any help is appreciated!
I want to generate knockout cell lines (of C2C12) with the crispr/cas method. Then use FACS to get single cells in a 96-wells plate. After cell sorting I want to expand and clone the single cells, but I don't have a protocol for single cell clonal expansion. Especially, because C2C12 cells are hard to culture as single cells (from what I know from literature). Can anyone help me or please give me some advice?
Kind regards,
Floris
We are genome editing Clotridioides difficile via CRISPR system. However, we met some difficulties generating a clean knock out. The colonies after editing showed both WT and KO PCR bands as the attached gel shows. We tried to re-streak and isolate single colonies multiple times, but we always got WT colonies instead (couldn't isolate KO colonies). Would you please give us some suggestions about this issue? Thanks a lot!
Im using CRISPR edited GM12878 cells to grow the single cell clones.
It was digested by using FastDigest BsmBI. I just found out that there are 2 cutting site of BsmBI. So, should I design 2 sets of primers to check for the gRNA insert?
Quite a naive question
I am looking for the most optimal way to transfect different cell lines with the same construct. Some of them are notoriously difficult to transfect, like RWPE1
I am satisfied with the quality of transient DNA transfection, which I did on simple lines like HEK293 or HeLa. But maybe it’s time for me to somehow optimize the process? Please advise, maybe it’s time for me to learn CRISPR? pLenti? Something else?
talk to me please
Hello,
I am trying to generate the CRISPR Knock In of about 1.7kb in HEK293T Cell line my HDR(linearized) is having 500bp Right Homology Arm left, 500bp Right Homology Arm and 1.8kb transgene how much concentration of my HDR should I use for knock in? is it same as gRNA concentration?
I'm using the Chen&Wente protocol: https://www.addgene.org/static/cms/filer_public/02/12/0212c99c-6937-4884-8fb0-a097b965f1c3/sgrna-plasmid-construction-protocol.pdf
This is supposed to be extremely efficient, but somehow, I've never gotten it to work. I ordered completely new enzymes because I thought my enzymes may be thawing (I re-ordered BsmBI, BlgIII, and Sal1HF), and my negative control had a greatly decreased number of colonies compared to my guide+plasmid plates for the first time; hence, I thought my enzymes were the problem. However, upon sequencing, I still got empty vector. Should the next step be to order new ligase? I'm not sure what's going wrong!
Hello ! I m a master just starting work in CRISPR genome editing ,
and trying to knock-in a reporter in my interesting gene
my method is transfect RNP and dsODN by lipo2000 to 293T cell line ,there is variety RNP and donor DNA concentration found in paper (few nM to 60nM RNP , about 500ng or even 18nM ODN ect)
In my experiment , I have a donor DNA about 4kb ,
so I perform a set of test using 60nM RNP (1:1 Cas9/gRNA) with 50 to 400 ng dsODN , but get no successful
at next condition test , I perform 0,15,30 nM of RNP and positive plasmid control to test transfection efficiency and get the result EGFP may get lower with higer RNP concentration.
whether the too giant size of molecular and negative charge give rice to barrier when liposome formation in my condition?
is my donor DNA too big or it must should be plasmid or ssODN?
or just this method working in even low efficiency?
please any good condition and advice !
sincere thanks.
and sorry for too much question and typo
I am doing crispr knockout in T cells. After transfection, I am trying to validate knockout by western blot and sequencing. with western blot, I can see truncated protein in knockout cells whereas in scrambled cells, band is at the expected position of the target protein.
With agarose gel, I can see 3 bands for knockout protein and scrambled is one band. My question is how do I sequence three bands. Shall i cut individual bands extract and purify DNA and then send for sequncing ?
what should be my approach? I am attaching pic of gel for reference.
pls help
I am doing crispr knockout in T cells. After transfection, I am trying to validate knockout by western blot and sequencing. with western blot, I can see truncated protein in knockout cells whereas in scrambled cells, band is at the expected position of the target protein.
With agarose gel, I can see 3 bands for knockout protein and scrambled is one band. My question is how do I sequence three bands. Shall i cut individual bands extract and purify DNA and then send for sequncing ?
what should be my approach? I am attaching pic of gel for reference
I have the sgRNA vector from addgene(#75112). It has the BsmBI site. I've designed the sgRNA already.
The problem is, should I also design a control sgRNA???
It seems the vector itself, (also a lot of other common sgRNA vector), already has some sequences there flanked by BsmBI site. Can I directly use the vector without substitution as control??
What are the key differences and benefits over using CRISPR dCas9 system /CRISPR interference (CRISPRi) for transcriptional regulation over more traditional methods such as Structure-based combinatorial protein engineering (SCOPE) or others?
I am performing a KO of my gene by frameshift in zebrafish. I have been screening this by the disruption of a restriction enzyme site in a PCR fragment. I now have an incross of 2 mutant fish and am hoping raise the progeny and get some homozygous fish. I will use DNA from tissue sample as the PCR template but I am wondering how I can separate homozygous from heterozygous. In my experience so far the F1 generation from an outcross with wild type showed an extremely faint undigested band (wt DNA) compared to the really bright digested band (indicating mut DNA) for some reason even though an outcross should automatically result in heterozygous fish containing both wt & mut (digested & undigested) DNA. I presumed that the concentration of the mut & wt allele would be similar. Has anyone come across this difficulty before. Could there be a reason a mutant would amplify more in a PCR reaction (it is only 7 bp shorter)? Any suggestions for a way to tell the difference? I am concerned since the band is so faint on heterozygotes I may misidentify heterozygotes as homozygotes.
My group mates and I are making a report on the journal article "CRISPR/Cas9-mediated gene editing in human zygotes using Cas9 protein" by Tang et al. (2017) and we are having difficulty understanding the purpose of the T7E1 and HindIII site. Initially, we thought the T7E1 indicated gene editing made by CRISPR or indicated non-homologous end joining repair, and that the presence of the HindIII site indicated homology-directed repair; however, as we continue to research on this, we think it may not be as simple initially thought.
Hi all,
I am doing CRISPR on Hek293T cells. After FACS, the one cell wells do not survive/grow only the wells were 50/100 cells were plated. Any tips to improve the survival rate for the one cell cultures? I use DMEM high glucose with 10 % FBS. The cells are 11-15 µm, so I can not use a 22 µm filter to filter medium from other cells.
Thanks a lot.
I am doing a CRISPR targeting to knock out one intron of my gene of interest in a cancer cell line and replace that intron with GFP. I am wondering if I should pick single cell clone with possible targeting events or if I should do flow to select positive cells. Any suggestion? Thank you for any help.
We are interested in generating knockout in cell lines using the CRISPER/CAS9 system and replacing our gene with GFP. After performing a double transfection using the guide RNA and the donor vector, we selected the cells against purmycin and selected a number of colonies. How would you go about confirming the knockout?
Hi All, I am transfecting two vectors (one gRNA vector and a HDR vector) to KO a gene. I have puromycin resistant in HDR vector. I am using Fugene for transfection purpose. I was wondering to know how long should I wait to start selection after transfection of the crispr vectors?
Is there any protocol of knocking out one gene in THP1 cells using CRISPR/Cas9 system ?
Here, I used LentiCRISPR-v2 system to harvest virus carring guide RNA, but when I added the virus into THP1 cells, these cells were going to be activated and differenatiated, and they would grow together.
Also, THP1 cells are a little hard to be transfected with lipo2000, so pX459 or pX458 plasmids system may be not availiable.
Hi everyone. Has anyone worked with Raji cell transfection? I recently used AMAXA technology to transfect Raji cells with a CRISPR construct that resulted in a very poor efficiency (~1%). When I tried the same construct in HEK293T cells with Xtreme gene9 I got more than 80% transfected cells plus very high levels of GFP expression that was part of the cassette. Any alternative suggestions for RAJI transfection? I know that Xtreme gene9 probably won't work well on them...
Has anyone had any success with Crispr and MDA-MB-231 or MCF-7 cell lines?
Limitation:
1) target specific gene:
2) do epigenetic modification:
I'm doing CRISPRi with dead spCas9 on mouse cells, and normally we do the following for gRNA design:
1. 20 nt spacer
2. Extra G on the 5' to enhance expression from hU6 promoter.
Final product: 5'-G-spacer-3'
But I accidentally included two Gs instead of 1 G.
Final product: 5'-GG-spacer-3'
Does this matter? I did *a lot* of cloning and cell line generation this way, so I don't know if I will have time to go back to redo it before my contract is up.