Science method

CRISPR - Science method

Explore the latest questions and answers in CRISPR, and find CRISPR experts.
Questions related to CRISPR
  • asked a question related to CRISPR
Question
2 answers
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!!!
Relevant answer
Answer
There is no point. Why introduce cell type and that vector situation as new variables? This is not going to tell you anything about what happens in your final intended system. Optimize for what you want, as directly as possible.
  • asked a question related to CRISPR
Question
4 answers
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.
Relevant answer
Answer
Didier Poncet Thanks again for your kindly help, and I have learned a lot from it! So i would take TCDI50 as consideration, for it represents infectious particles and shows the viral infectivity.
  • asked a question related to CRISPR
Question
4 answers
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.
Relevant answer
Answer
As Nicolas mentioned, size matters. Larger transgenes result in lower titers. If you are not experienced at making lenti vectors then your titer may be low for smaller transgenes like GFP. With this larger construct, your titer may fall low enough that you don't detect it at all.
Did you titer the GFP virus? If so, what was the titer? Did you transduce the experimental cells with the GFP virus or did you transduce 293?
Have you tried adding your Cas9 virus to 293 to see if you can detect it? The 293 line will probably be more permissive to lenti transduction. I would titrate both virus stocks on 293 to get a handle on titer and, if you can detect virus from the Cas9 prep, the difference in titer between the two stocks.
  • asked a question related to CRISPR
Question
3 answers
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?
Relevant answer
Answer
I hope you are doing well.
Yes, flow cytometry, commonly known as Fluorescence-Activated Cell Sorting (FACS), has been widely used on CRISPR-Cas9 transfected cells, and it serves as a powerful tool for the analysis and sorting of these modified cells. The application of FACS in this context generally focuses on the identification, quantification, and isolation of cells that have been successfully edited by CRISPR-Cas9.
Here's a detailed explanation of how FACS is utilized with CRISPR-Cas9 transfected cells:
  1. Detection of Transfection Efficiency: After transfecting cells with CRISPR-Cas9 components, such as Cas9 protein and guide RNA (gRNA), a fluorescent reporter (e.g., GFP or RFP) is often co-expressed. FACS is then used to detect the expression of these reporters, providing a reliable measure of transfection efficiency.
  2. Sorting of Edited Cells: For targeted gene editing, cells that exhibit specific phenotypic changes, such as the knockout or knock-in of a gene, can be selectively sorted using FACS. This approach allows for the enrichment of cell populations with the desired genetic modifications, improving the efficiency of subsequent experiments.
  3. Validation of Gene Editing: Beyond sorting, FACS can be employed to validate the functional outcomes of CRISPR-Cas9 editing. For example, if the target gene affects surface markers or other cellular characteristics detectable by flow cytometry, FACS can confirm successful editing at the functional level.
  4. Single-Cell Analysis: FACS allows for the isolation of single cells, which can be particularly valuable when generating clonal populations from edited cells. Single-cell analysis enables researchers to assess the heterogeneity of gene editing outcomes, which is critical for applications requiring precise genetic modifications.
These applications underscore the versatility of FACS in conjunction with CRISPR-Cas9 technology, providing both qualitative and quantitative insights into the gene editing process. Proper optimization of FACS parameters, including antibody selection, gating strategies, and fluorescence compensation, is essential for achieving accurate results.
If you have any specific questions or require further details, please do not hesitate to reach out.l Reviewing the protocols listed here may offer further guidance in addressing this issue
l Reviewing the protocols listed here may offer further guidance in addressing this issue
  • asked a question related to CRISPR
Question
2 answers
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.
Relevant answer
I'm sorry but this is not my area of expertise. Enjoy your research!
  • asked a question related to CRISPR
Question
3 answers
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.
Relevant answer
Answer
I am not an expert in this field, but I am very interested and have researched to find an answer. I received some assistance from tlooto.com for this response. Could you please review the response below to see if it is correct?
CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats) is a revolutionary gene-editing tool that uses a guide RNA to direct the Cas9 enzyme to a specific genome location for precise DNA modifications. Despite its promise, CRISPR faces significant limitations and challenges in agriculture, particularly regarding off-target effects. Off-target effects can introduce unwanted, potentially harmful genome alterations, complicating the development of safe, reliable crop traits [4][5]. Additionally, technical and governance challenges, such as regulatory hurdles and the intricacy of gene drive mechanisms, further hinder its agricultural application [1][3]. Advances in detection methods like digital PCR and improvements in guide RNA synthesis are being developed to mitigate these issues [2][6].
Reference
[1] Chen, K., Wang, Y., Zhang, R., Zhang, H., & Gao, C. (2019). CRISPR/Cas Genome Editing and Precision Plant Breeding in Agriculture.. Annual review of plant biology, 70, 667-697 .
[2] El-Mounadi, K., Morales-Floriano, M. L., & Garcia-Ruiz, H. (2020). Principles, Applications, and Biosafety of Plant Genome Editing Using CRISPR-Cas9. Frontiers in Plant Science, 11.
[3] Son, S., & Park, S. (2022). Challenges Facing CRISPR/Cas9-Based Genome Editing in Plants. Frontiers in Plant Science, 13.
[4] Saini, H., Thakur, R., Gill, R., Tyagi, K., & Goswami, M. (2023). CRISPR/Cas9-gene editing approaches in plant breeding. GM Crops & Food, 14, 1 - 17.
[5] Zhang, Y., Chen, S., Yang, L., & Zhang, Q. (2023). Application progress of CRISPR/Cas9 genome-editing technology in edible fungi. Frontiers in Microbiology, 14.
[6] Sato, G., & Kuroda, K. (2023). Overcoming the Limitations of CRISPR-Cas9 Systems in Saccharomyces cerevisiae: Off-Target Effects, Epigenome, and Mitochondrial Editing. Microorganisms, 11.
  • asked a question related to CRISPR
Question
3 answers
Relevant answer
Answer
Hort Tandem Repeat (STR) testing, often used in forensic science and genetic analysis, is a powerful tool for identifying individuals based on the number of repeating units at specific locations in their DNA. STR testing can still identify someone with a germ-line mutation by detecting changes in the repeat patterns at specific DNA loci. If a germ-line mutation alters an STR region, the individual’s STR profile may show a new allele or a different number of repeats than expected based on their relatives. This can result in mismatches in parent-child STR profiles, which might indicate a mutation. While STR testing is generally reliable, the results must be carefully interpreted.
  • asked a question related to CRISPR
Question
5 answers
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
Relevant answer
Answer
As I said before, even with the tag, the protein could be folded in such a way that the interaction with the nickel ions is not strong enough. If the protein becomes adsorbed at 5-10 mM imidazole and desorbs in 20-50 mM imidazole with washing, wash with the binding buffer, afterward, run an imidazole gradient (very slow) to 50 mM imidazole (or less).
  • asked a question related to CRISPR
Question
1 answer
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.
Relevant answer
Answer
Recent advancements in CRISPR technology have significantly improved its precision, efficiency, and versatility compared to traditional genetic modification methods. Here’s a simplified overview:
  1. Enhanced Precision: New CRISPR tools, like CRISPR/Cas9 variants and base editors, allow for more accurate editing of specific DNA sequences. Base editors can change individual DNA bases without causing double-strand breaks, reducing errors and unintended effects.
  2. Extended Applications: CRISPR has expanded beyond simple gene knockouts to include more complex modifications, such as inserting or correcting genes. Technologies like CRISPR/Cas12 and CRISPR/Cas13 offer new ways to target different types of genetic material and improve the scope of CRISPR applications.
  3. Increased Efficiency: Advances have made CRISPR more efficient in targeting and editing specific genes. Techniques such as CRISPR interference (CRISPRi) and CRISPR activation (CRISPRa) enable precise regulation of gene expression without altering the DNA sequence.
  4. Reduced Off-Target Effects: Improved CRISPR designs and delivery methods reduce the risk of off-target effects, where unintended parts of the genome might be altered.
Comparison to Traditional Methods:
  • Precision: Traditional methods, like random mutagenesis or older gene-editing techniques, often result in less specific changes, potentially affecting multiple genes or regulatory elements. CRISPR provides more targeted edits.
  • Speed and Cost: CRISPR is generally faster and more cost-effective than traditional methods, which can involve time-consuming screening and more complex procedures.
  • Versatility: CRISPR’s ability to edit multiple genes simultaneously and its adaptability to various organisms make it more versatile compared to traditional approaches that might be limited to single-gene modifications.
Overall, CRISPR technology represents a significant advancement over traditional genetic modification methods, offering greater precision, efficiency, and flexibility in genetic research and applications.
  • asked a question related to CRISPR
Question
5 answers
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.
Relevant answer
Answer
Well, creating a semi-permeable transcription bubble can be challenging in the context of the structural stability of DNA as it tends to reanneal to its double-stranded form. Next is the concern of replication, which involves the fact that semi-permanent unwinding can potentially hinder the replication machinery from proceeding with DNA replication. Lastly, to maintain the transcription bubble to its semi-permanent unwound state, RNA polymerase is required to be halted in its activity at the transcription site, which could, in turn, lead to instability and interference in the replication of the plasmid. Considering these aspects, I believe three probable yet theoretical strategies can be adopted in this regard. First is genetically engineering a modified RNA polymerase, which can maintain the plasmid DNA at its single-stranded state by getting associated at a precise plasmid location without hindering the transcription process. Second is implementing genetically engineered single-strand binding (SSB) proteins, which can keep the plasmid DNA at its unwound state without interfering with RNA synthesis. Lastly, chemical molecules such as intercalating agents are introduced, which can develop proximal unwinding by being inserted at the nitrogenous base pairs of plasmid DNA; Molecules that are enhancers or activators of helicases; Hydrogen bond destabilizers like Di-Methyl Sulfoxide (DMSO), Urea or Formamide which can perform denaturation of double-stranded DNA; Cross-linking agents like Psoralens which forms covalent cross-linkages between single-stranded DNA molecules and DNA or RNA polymerases; Ligands which associate with single-stranded DNA such as Peptide Nucleic Acids (PNAs) and nucleic analogs which stabilizes the single-stranded structures of DNA; Alkylating agents such as Nitrogen and Sulfur derivatives of Mustard gas, Ethyl Methanesulfonate (EMS), Methyl Methanesulfonate (MMS), N-Nitrosoureas and Temozolomide. Nevertheless, besides being hypothetical, all these strategies have cons of their own.
  • asked a question related to CRISPR
Question
3 answers
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!
Relevant answer
Answer
Mauro Lago Docampo thank you so much for your response!
Regarding selection of KRAS mutant clones: The authors in the paper I mention above (Clevers, Nature 2015) select for KRAS G12D mutants by removing EGF and adding the antibiotic gefitinib to culture media. My plan is to select for KRAS mutant clones using the selection media described in this paper.
The more research I have done into this experiment, I plan on introducing the triple KO in a second separate step to that described above. Again, the authors of the Clevers paper describe a media specially for selection of triple KO clones (-WNT -Noggin +Nutlin-3).
My plan was to then select and expand a single clone that has successfully been transfected with the G12D mutant/triple KO. I will confirm successful transfection with western blot and Sanger sequencing.
I hope this sufficiently addresses the points you made!
  • asked a question related to CRISPR
Question
2 answers
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?
Relevant answer
Answer
Dear Colleague,
I hope this message finds you well. The stability and half-life of guide RNA (gRNA) are critical factors in the efficacy of CRISPR/Cas9 genome editing systems. Here is a detailed and logical explanation regarding the average half-life of gRNA not bound to Cas9.
Average Half-Life of Unbound gRNA
  1. General Stability:The stability of gRNA in a cellular environment is influenced by several factors, including the presence of nucleases, the cellular compartment, and the chemical modifications of the RNA.
  2. Unmodified gRNA:Degradation by Nucleases: In the cellular environment, unmodified gRNA is susceptible to degradation by RNases. The half-life of such gRNAs can be relatively short, typically ranging from a few minutes to several hours, depending on the specific conditions and cell type. Literature Estimates: Studies have shown that unmodified gRNA can have a half-life of approximately 30 minutes to 2 hours in various cellular environments.
  3. Modified gRNA:Chemical Modifications: To enhance stability, gRNAs can be chemically modified. Common modifications include 2'-O-methyl and phosphorothioate linkages at the 3' and 5' ends, which can significantly increase their resistance to nuclease degradation. Increased Stability: Modified gRNAs can have extended half-lives, often ranging from several hours to over a day, depending on the extent and type of modifications applied.
  4. Experimental Conditions:In Vitro vs. In Vivo: The half-life of gRNA can differ substantially between in vitro and in vivo conditions. In a controlled in vitro environment, where nucleases are minimized, gRNAs can be more stable compared to the in vivo cellular milieu. Cell Type and Compartment: The type of cells and the intracellular compartment where the gRNA resides also affect its stability. For instance, cytoplasmic RNases can degrade gRNA more rapidly than nuclear RNases.
Practical Implications for CRISPR/Cas9 Experiments
  1. Optimization of gRNA:Chemical Modifications: Using chemically modified gRNAs can be advantageous for experiments requiring prolonged activity or when working in environments with high nuclease activity. In Vitro Transcription: If using in vitro transcribed gRNAs, consider incorporating stabilizing modifications to enhance their half-life.
  2. Delivery Methods:RNP Complexes: Delivering gRNAs as ribonucleoprotein (RNP) complexes with Cas9 can protect the gRNA from degradation, as the binding to Cas9 can shield the gRNA from nucleases. Lipid Nanoparticles and Electroporation: Utilize efficient delivery methods such as lipid nanoparticles or electroporation to ensure rapid delivery of gRNA into cells, minimizing the exposure to extracellular nucleases.
  3. Experimental Design:Time Course Studies: When designing CRISPR/Cas9 experiments, consider the half-life of unbound gRNA in your time course studies to ensure that sufficient active gRNA is present throughout the experiment. Storage and Handling: Store gRNAs at -80°C and minimize freeze-thaw cycles to maintain their stability and activity.
Conclusion
The average half-life of unmodified gRNA not bound to Cas9 can range from 30 minutes to 2 hours, depending on the specific cellular conditions. Chemical modifications can significantly enhance the stability of gRNA, extending its half-life to several hours or more. Understanding these factors is crucial for optimizing CRISPR/Cas9 experiments and ensuring efficient genome editing outcomes.
Should you have any further questions or require additional assistance, please feel free to reach out.
In a Crispr/Cas system, what is the average half life of a guide RNA (gRNA) not bound to Cas9?
  • asked a question related to CRISPR
Question
3 answers
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!
Relevant answer
Answer
Dear Colleague,
I hope this message finds you well. Observing extra bands on gRNA template PCR results can be attributed to several factors related to the PCR process and template quality. Here is a detailed and logical approach to understanding and troubleshooting this issue:
Potential Causes of Extra Bands in gRNA Template PCR
  1. Non-Specific Amplification:Primer Design: Primers may bind to unintended regions of the template DNA, resulting in non-specific amplification. GC Content and Tm: Primers with suboptimal GC content or melting temperatures (Tm) can lead to non-specific binding and amplification.
  2. Template Contamination:Multiple Templates: Contamination with other DNA templates can result in the amplification of unintended products. Degraded Template: Degraded or impure template DNA can contribute to non-specific amplification.
  3. Suboptimal PCR Conditions:Annealing Temperature: Incorrect annealing temperature can lead to non-specific primer binding. Too low an annealing temperature increases non-specific binding. Magnesium Concentration: Suboptimal magnesium ion concentration can affect the specificity of the PCR reaction. Cycle Number: Excessive cycle numbers can amplify non-specific products.
  4. Primer-Dimer Formation:Primer Concentration: High primer concentration can promote the formation of primer-dimers, which appear as extra bands. Complementarity: Partial complementarity between primers can result in the formation of primer-dimers.
Troubleshooting Steps
  1. Review Primer Design:Specificity: Ensure primers are designed to specifically bind to the target region. Use software tools to check for potential off-target binding sites. Tm and GC Content: Optimize the melting temperatures and GC content of the primers to ensure specific binding. Aim for a Tm of 58-60°C and a GC content of 40-60%.
  2. Optimize PCR Conditions:Annealing Temperature: Perform a gradient PCR to determine the optimal annealing temperature. Start with a temperature 2-5°C below the Tm of your primers. Magnesium Concentration: Adjust the magnesium ion concentration in 0.5 mM increments to optimize specificity. Cycle Number: Reduce the number of PCR cycles to the minimum necessary to obtain a visible product.
  3. Improve Template Quality:Purity: Ensure the template DNA is pure and free from contaminants. Use a high-quality DNA extraction method and verify the purity using spectrophotometry (e.g., A260/A280 ratio). Quantity: Use an appropriate amount of template DNA. Too much template can lead to non-specific amplification.
  4. Minimize Primer-Dimer Formation:Primer Concentration: Use primers at a final concentration of 0.1-0.5 µM to minimize primer-dimer formation. Hot-Start PCR: Use a hot-start DNA polymerase to reduce non-specific amplification and primer-dimer formation.
Example Protocol Adjustments
  1. Primer Design:Design primers using a reliable tool like Primer-BLAST to ensure specificity and optimal Tm. Verify the absence of significant secondary structures and primer-dimer formation using a tool like OligoAnalyzer.
  2. Gradient PCR:Set up a gradient PCR to determine the optimal annealing temperature. Example gradient: 55°C to 65°C.
  3. Reaction Mix Optimization: Prepare a master mix with the following components: diff复制代码- Template DNA: 10-100 ng - Forward Primer: 0.2 µM - Reverse Primer: 0.2 µM - dNTPs: 200 µM each - MgCl2: 1.5-2.5 mM - Buffer: 1x PCR buffer - DNA Polymerase: 0.5-1 unit - Nuclease-free water: to 25 µL final volume
  4. PCR Cycling Conditions:Initial denaturation: 95°C for 2 minutes Denaturation: 95°C for 30 seconds Annealing: 55-65°C (determine optimal temperature) for 30 seconds Extension: 72°C for 30 seconds per kb Final extension: 72°C for 5 minutes Number of cycles: 25-35
By following these steps and making the necessary adjustments, you can reduce or eliminate extra bands in your gRNA template PCR results, ensuring more specific and accurate amplification.
Should you have any further questions or require additional assistance, please feel free to reach out.
This list of protocols might help us better address the issue.
  • asked a question related to CRISPR
Question
9 answers
glucagon biogenesis
glucagon secretion 
siRNA
CRISPR
overexpression
Relevant answer
Answer
Dear Colleague,
I hope this message finds you well. When studying glucagon secretion in pancreatic alpha cells, it is crucial to use a reliable and well-characterized cell line. One of the most commonly used and reliable pancreatic alpha cell lines for this purpose is the αTC1-6 cell line.
αTC1-6 Cell Line Overview
  1. Origin:The αTC1-6 cell line is derived from a transgenic mouse model and is widely used in research focusing on glucagon secretion and alpha cell function.
  2. Characteristics:Glucagon Secretion: This cell line is known for its robust and reproducible glucagon secretion, making it suitable for studying the regulation of glucagon release. Alpha Cell Markers: αTC1-6 cells express key alpha cell markers, including glucagon, which confirms their identity and relevance for alpha cell research. Culture Conditions: These cells can be cultured under standard conditions using DMEM supplemented with 10% fetal bovine serum (FBS), 1% penicillin-streptomycin, and 15 mM HEPES.
  3. Advantages:Reproducibility: αTC1-6 cells provide consistent and reproducible results, essential for experimental reliability. Ease of Use: They are relatively easy to culture and maintain, which simplifies experimental procedures.
Protocol for Studying Glucagon Secretion in αTC1-6 Cells
  1. Cell Culture:Medium Preparation: Prepare DMEM supplemented with 10% FBS, 1% penicillin-streptomycin, and 15 mM HEPES. Cell Maintenance: Culture αTC1-6 cells in a humidified incubator at 37°C with 5% CO2. Passage the cells when they reach 70-80% confluency.
  2. Glucagon Secretion Assay:Cell Seeding: Seed αTC1-6 cells in a 24-well plate at an appropriate density (e.g., 1 x 10^5 cells/well) and allow them to adhere overnight. Glucose Starvation: Incubate the cells in glucose-free DMEM for 1-2 hours to enhance glucagon secretion responsiveness. Stimulation: Treat the cells with different glucose concentrations or other secretagogues (e.g., arginine) to stimulate glucagon secretion. Include appropriate controls. Collection of Supernatants: Collect the culture supernatants at various time points for glucagon measurement.
  3. Glucagon Measurement:ELISA: Use a glucagon-specific ELISA kit to quantify glucagon levels in the collected supernatants according to the manufacturer's instructions. Ensure that the kit is sensitive and specific for glucagon detection. Normalization: Normalize the glucagon secretion data to the cell number or total protein content to account for variations in cell density.
Data Analysis and Interpretation
  1. Quantification: Analyze the glucagon concentrations from the ELISA and compare the secretion levels under different conditions.
  2. Statistical Analysis: Perform appropriate statistical tests (e.g., ANOVA, t-test) to determine the significance of the differences observed between experimental groups.
  3. Validation: Validate your findings by repeating the experiments and ensuring consistency across multiple independent replicates.
Alternative Cell Lines
While αTC1-6 cells are widely used and reliable, you may also consider other cell lines such as InR1-G9 cells, which are another pancreatic alpha cell line. However, αTC1-6 cells are generally preferred due to their extensive characterization and reproducible glucagon secretion.
By using the αTC1-6 cell line and following the outlined protocol, you can effectively study glucagon secretion and gain insights into the regulation of pancreatic alpha cell function.
Should you have any further questions or require additional assistance, please feel free to reach out.
This protocol list might provide further insights to address this issue.
  • asked a question related to CRISPR
Question
9 answers
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.
Relevant answer
Answer
researchers have used the CRISPR/Cpf1 (now often referred to as CRISPR/Cas12a) system for generating knock-in lines, similar to how CRISPR/Cas9 is used. The CRISPR/Cpf1 system has some distinct advantages and differences compared to CRISPR/Cas9, which can be beneficial in certain applications.
Overview of CRISPR/Cpf1 for Knock-In
**1. System Characteristics
**1.1 Cpf1 (Cas12a):
Different Mechanism: Cpf1/Cas12a has a different cleavage mechanism compared to Cas9. It generates sticky ends with 5' overhangs, which can be advantageous for certain types of genetic modifications.
Simpler Design: Cpf1 requires a simpler guide RNA (gRNA) design, with only one RNA molecule (as opposed to the separate crRNA and tracrRNA required for Cas9).
**1.2 Knock-In Strategy:
Targeting Vector: A donor DNA vector with the desired insert and homology arms flanks the target site. The homology arms are sequences homologous to regions flanking the double-strand break (DSB) induced by Cpf1.
Repair Mechanism: The DSB created by Cpf1 is repaired by homology-directed repair (HDR) using the donor DNA vector to incorporate the knock-in sequence.
**2. Experimental Steps
**2.1 Designing Guides:
Select Target Sites: Identify suitable target sites for Cpf1, usually requiring PAM sequences (5'-TTTV-3' for Cpf1).
Design gRNA: Design the single gRNA specific to your target site. This guide will direct the Cpf1 enzyme to the correct location in the genome.
**2.2 Preparing Components:
Cpf1 Protein: Obtain or produce the Cpf1 protein.
gRNA: Synthesize the gRNA or use a vector to express it.
Donor DNA: Clone the knock-in sequence into a donor vector with homology arms flanking the insertion site.
**2.3 Transfection:
Transfect Cells: Introduce Cpf1, gRNA, and donor DNA into your target cells via transfection methods suitable for your cell type (e.g., electroporation, lipofection, or viral transduction).
**2.4 Selection and Screening:
Selection: Use selection markers if included in the donor vector to identify successfully modified cells.
Screening: Screen for successful knock-ins using methods like PCR, sequencing, or Southern blotting to verify the incorporation of the knock-in sequence.
**3. Advantages and Considerations
**3.1 Advantages:
Sticky Ends: The sticky ends generated by Cpf1 can improve the efficiency of HDR by creating more compatible ends with the donor DNA.
Fewer Off-Target Effects: Some studies suggest Cpf1 may have fewer off-target effects compared to Cas9, although this can depend on the specific context and guide RNA design.
**3.2 Considerations:
Efficiency: The efficiency of knock-in can vary depending on the cell type and the specific CRISPR/Cpf1 system used.
Optimization: Experimental conditions, such as the ratio of Cpf1, gRNA, and donor DNA, need to be optimized for each system and cell type.
Summary
Using CRISPR/Cpf1 for generating knock-in lines is a viable and potentially advantageous alternative to CRISPR/Cas9. The system's distinct properties, such as its ability to generate sticky ends, can be leveraged to improve knock-in efficiency. As with any gene editing system, careful design, optimization, and validation are key to successful outcomes.
l With this protocol list, we might find more ways to solve this problem.
  • asked a question related to CRISPR
Question
4 answers
I have used ssDNA oligo and plasmids for HDR. I am wondering whether anyone has tried using PCR product directly as HDR template. 
Relevant answer
Answer
you can use a PCR product as a CRISPR HDR (Homology-Directed Repair) donor, but there are several considerations to ensure its effectiveness:
**1. Quality of the PCR Product
**1.1 Purity:
Ensure Purity: The PCR product should be pure and free from contaminants, such as residual primers, enzymes, and other by-products. Use a purification step like gel extraction or commercial PCR clean-up kits.
**1.2 Length and Integrity:
Check Integrity: Verify that the PCR product is of the correct size and does not have unintended bands or degradation products. This can be assessed by running an aliquot on an agarose gel.
**2. Design Considerations for the HDR Donor
**2.1 Homology Arms:
Include Homology Arms: The PCR product should contain homology arms flanking the desired insertion or modification site. The homology arms should be sufficiently long (typically 500-1000 bp) to promote efficient HDR.
**2.2 Donor Sequence:
Incorporate the Desired Sequence: Ensure that the sequence you want to insert or modify is correctly included in the PCR product. Verify the sequence by sequencing if necessary.
**2.3 Avoiding Repeats:
Minimize Repeats: Avoid long repeats or sequences that might cause unwanted recombination or instability.
**3. Transformation and Transfection
**3.1 Transformation Efficiency:
Assess Efficiency: If using bacteria, check the efficiency of transformation. High-quality DNA is essential for efficient transformation or transfection.
**3.2 Transfection:
Optimize Conditions: When using the PCR product in mammalian cells, optimize transfection conditions to ensure efficient delivery of the donor DNA. Methods include lipofection, electroporation, or viral delivery systems.
**4. Validation
**4.1 Confirm Insertion:
Validation: After introducing the HDR donor into the cells, validate successful insertion or modification by PCR, sequencing, or other molecular assays.
**4.2 Screening:
Screen for Correct Integration: Use appropriate screening methods to confirm that the HDR process occurred correctly and that the donor DNA has been properly integrated.
**5. Considerations for Using PCR Products
**5.1 Length Limitations:
Product Length: PCR products are typically short (e.g., 200-1000 bp). For larger insertions or modifications, consider using plasmid-based donors with larger homology arms.
**5.2 End Modification:
Blunt Ends vs. Overhangs: Ensure that the ends of the PCR product are compatible with the CRISPR-generated DSB (Double-Strand Break) ends. Depending on the CRISPR system used, you might need to modify the ends (e.g., to be blunt or to have overhangs).
**5.3 PCR Artifacts:
Check for Artifacts: Ensure that the PCR product does not contain artifacts such as nonspecific bands or residual primers that could affect the HDR process.
Alternative Approaches
**1. Plasmid-Based Donors:
Consider Using Plasmids: For larger modifications or more complex constructs, using plasmid-based HDR donors with appropriate homology arms may be more effective.
**2. Commercial Donors:
Pre-made Donors: Commercially available HDR donor plasmids often come with optimized designs for efficient integration and can save time and effort.
In summary, while PCR products can be used as HDR donors, their effectiveness depends on proper design, purification, and delivery. For larger or more complex modifications, plasmid-based donors or commercially available options might be preferable.
l Reviewing the protocols listed here may offer further guidance in addressing this issue.
  • asked a question related to CRISPR
Question
2 answers
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.
Relevant answer
Answer
Estimating the probability of forming a short CRISPR with a single spacer in a random sequence involves several steps. This calculation depends on the specific sequence characteristics and the CRISPR system's requirements. Here’s a structured approach to estimate this probability:
**1. Define the Parameters
**1.1 CRISPR System Characteristics:
Spacers: Typically, spacers in CRISPR systems are around 20 nucleotides long.
Protospacer Adjacent Motif (PAM): CRISPR systems require a PAM sequence adjacent to the target site. For example, the Streptococcus pyogenes Cas9 requires the PAM sequence "NGG."
**1.2 Random Sequence Properties:
Length: Determine the length of the random sequence where you are searching for the spacer.
Nucleotide Composition: For a truly random sequence, assume equal probabilities for each nucleotide (A, T, C, G).
**2. Calculate the Probability of a Specific Spacer Sequence
**2.1 Probability of Matching a Specific Spacer:
Calculate for PAM: If the PAM sequence is required, first calculate the probability of finding this PAM sequence in the random sequence.
Probability of Spacer Sequence: For a spacer of length L nucleotides, the probability of finding a specific sequence of length L in a random sequence is:
𝑃
(
spacer
)
=
(
1
4
)
𝐿
P(spacer)=(
4
1
)
L
where
1
4
4
1
is the probability of each nucleotide occurring at a specific position, and
𝐿
L is the length of the spacer.
**2.2 Consider PAM Sequence:
Probability of PAM: For a PAM sequence of length k nucleotides, assuming equal probability for each nucleotide, the probability of finding the PAM is:
𝑃
(
PAM
)
=
(
1
4
)
𝑘
P(PAM)=(
4
1
)
k
**3. Calculate the Probability of Spacer and PAM Co-occurrence
**3.1 Independent Events:
Assuming Independence: If the presence of the spacer and PAM are independent, the combined probability of finding both in the random sequence is:
𝑃
(
spacer and PAM
)
=
𝑃
(
spacer
)
×
𝑃
(
PAM
)
P(spacer and PAM)=P(spacer)×P(PAM)
**3.2 Search Space:
Length of Random Sequence: If you are searching within a sequence of length N, the number of potential positions for the spacer and PAM is N - (L + k - 1).
**4. Estimate the Expected Number of Hits
**4.1 Expected Hits:
Calculate Expected Number: Multiply the probability of finding the spacer and PAM by the number of potential positions:
Expected Number of Hits
=
𝑃
(
spacer and PAM
)
×
(
𝑁
(
𝐿
+
𝑘
1
)
)
Expected Number of Hits=P(spacer and PAM)×(N−(L+k−1))
**4.2 Adjust for Overlaps:
Overlap: Adjust calculations if the spacer and PAM are not independent or if there are constraints on their positioning relative to each other.
Example Calculation
Assuming:
Spacer length (L) = 20 nucleotides
PAM length (k) = 3 nucleotides
Random sequence length (N) = 1000 nucleotides
Probability of Spacer:
𝑃
(
spacer
)
=
(
1
4
)
20
=
9.09
×
1
0
13
P(spacer)=(
4
1
)
20
=9.09×10
−13
Probability of PAM:
𝑃
(
PAM
)
=
(
1
4
)
3
=
0.0156
P(PAM)=(
4
1
)
3
=0.0156
Combined Probability:
𝑃
(
spacer and PAM
)
=
9.09
×
1
0
13
×
0.0156
=
1.42
×
1
0
14
P(spacer and PAM)=9.09×10
−13
×0.0156=1.42×10
−14
Expected Hits:
Expected Number of Hits
=
1.42
×
1
0
14
×
(
1000
(
20
+
3
1
)
)
1.42
×
1
0
14
×
977
1.39
×
1
0
11
Expected Number of Hits=1.42×10
−14
×(1000−(20+3−1))≈1.42×10
−14
×977≈1.39×10
−11
Conclusion
In this example, the probability of finding a specific 20-nucleotide spacer and a 3-nucleotide PAM sequence in a random 1000-nucleotide sequence is extremely low, reflecting the challenge of finding specific CRISPR target sites. Adjust parameters accordingly based on your specific requirements and sequence characteristics.
l This protocol list might provide further insights to address this issue.
  • asked a question related to CRISPR
Question
4 answers
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?
Relevant answer
Answer
If you're experiencing inefficient CRISPR knockout results, several factors could be contributing to the suboptimal outcome. Here’s a troubleshooting guide to help identify and address common issues:
1. Design Issues
1.1 Guide RNA (gRNA) Design:
Target Sequence: Ensure that the gRNA specifically targets a unique and suitable sequence in the gene of interest. Use tools like CRISPR design tools (e.g., CRISPR design tools by Broad Institute) to assess off-target effects and efficiency.
gRNA Efficiency: Verify that the gRNA has high predicted efficiency. Tools like CRISPR-Cas9 target site predictors can help evaluate this.
1.2 PAM Sequence: Check if the protospacer adjacent motif (PAM) sequence is present and optimal for the Cas9 protein you are using (e.g., NGG for SpCas9).
2. Delivery Method
2.1 Transfection Efficiency:
Transfection Method: Ensure that your transfection method (e.g., lipofection, electroporation) is efficient. Low transfection efficiency can lead to inadequate expression of the CRISPR components.
Optimization: Optimize the transfection conditions (e.g., reagent concentrations, cell density) for your specific cell type.
2.2 Expression of CRISPR Components:
Plasmid Quality: Verify the quality and quantity of your CRISPR plasmids. Ensure they are properly prepared and free from contaminants.
Protein Expression: Confirm that the Cas9 protein and gRNA are being expressed at sufficient levels. Check for expression using methods like Western blotting or PCR.
3. Cell Line Issues
3.1 Cell Type: Some cell types are more challenging to transfect or less responsive to CRISPR. Verify that your cell line is amenable to CRISPR editing.
3.2 Cell Health: Ensure that the cells are healthy and in the appropriate growth phase. Poor cell health can affect CRISPR efficiency.
4. Validation and Detection
4.1 Off-Target Effects: Perform off-target analysis to ensure that the observed effects are due to the intended CRISPR activity and not unintended modifications.
4.2 Knockout Efficiency: Validate knockout efficiency using various methods:
Genotyping: PCR and sequencing to confirm insertions or deletions (indels) at the target site.
Western Blotting: To check the absence or reduced levels of the target protein.
Flow Cytometry: If applicable, to assess changes in protein expression at the cellular level.
4.3 Functional Assays: Assess the functional impact of the knockout to confirm that it affects the biological function or pathway you are targeting.
5. Technical Issues
5.1 Cas9 Activity: Ensure that the Cas9 protein is active and properly localized within the cell.
5.2 gRNA Stability: Check the stability of the gRNA. It should be adequately expressed and stable within the cell.
5.3 Plasmid Integration: If using plasmid-based CRISPR, ensure that the plasmids are integrated into the genome or stably expressed in the cells.
6. Alternative Approaches
6.1 Use of Different gRNAs: Try designing and using different gRNAs targeting other regions of the gene.
6.2 Cas9 Variants: Consider using different Cas9 variants or other CRISPR-associated proteins (e.g., Cpf1) that might be more effective in your system.
6.3 Experimental Conditions: Test different experimental conditions, such as different cell densities or culture conditions, to improve efficiency.
By systematically addressing these factors, you can identify and correct issues that may be causing inefficient CRISPR knockout results.
l Check out this protocol list; it might provide additional insights for resolving the issue.
  • asked a question related to CRISPR
Question
4 answers
Relevant answer
Answer
Which epistemology do you associate with biology? Why?”
- epistemology absolutely directly is associated with biology, since all points/steps in the utmost fundamental result in epistemology – “Scientific method” , see https://en.wikipedia.org/wiki/Scientific_method
“…An iterative,[43] pragmatic[12] scheme of the four points above is sometimes offered as a guideline for proceeding:[47]
Define a question
Gather information and resources (observe)
Form an explanatory hypothesis
Test the hypothesis by performing an experiment and collecting data in a reproducible manner
Analyze the data
Interpret the data and draw conclusions that serve as a starting point for a new hypothesis
Publish results
Retest (frequently done by other scientists)
The iterative cycle inherent in this step-by-step method goes from point 3 to 6 and back to 3 again. ……”
- all/every living being, even bacteria, use and perform in their lives at their behavior.
Cheers
  • asked a question related to CRISPR
Question
1 answer
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,
Relevant answer
Answer
I found the solution to the issue. The problem was caused by a small formatting error in the gRNA list file. Specifically, there was a space between the gene name "septin" and the number "5" in the entry for "septin 5". It should be "septin5" without a space. This minor formatting error prevented the data from being processed correctly, leading to the plotting error. It took a long time to identify this issue, but it's resolved now. I'm sharing this in case someone else runs into the same error.
Best,
  • asked a question related to CRISPR
Question
2 answers
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!
Relevant answer
Answer
Hi...did you find the solution!?
  • asked a question related to CRISPR
Question
4 answers
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
Relevant answer
Answer
I usually select the cells before carrying out FACS.
  • asked a question related to CRISPR
Question
1 answer
I am looking for a CRISPR protocol for application in leishmania strains
Relevant answer
Answer
Hi Catherine, this might be a good place to start: http://www.leishgedit.net/Home.html
  • asked a question related to CRISPR
Question
4 answers
I'm working on a specific tissue knock in by CRISPR.
Relevant answer
Answer
CCTOP
  • asked a question related to CRISPR
Question
1 answer
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.
Relevant answer
Answer
Yeah I found the same thing. The nanopore tools are terrible and customer support completely unresponsive. You'll be better off aligning it yourself in ClustalW.
The high random error rate between reads is still a problem. We use pacbio for this now, which has very high fidelity. Costly, but highly effective.
  • asked a question related to CRISPR
Question
7 answers
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:
  1. 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
  2. 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.
Relevant answer
Answer
I think it's usually sold as a 10000X stock in DMSO, so 3.5 µL. If you have trouble seeing the band then double it to 7 µL.
  • asked a question related to CRISPR
Question
3 answers
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.
Relevant answer
Answer
Hi Zeyu;
Did you solve this problem?
  • asked a question related to CRISPR
Question
3 answers
Strain: LBA4404
Growing media: YEB (pH 7)
Temperature: 28°C, 200 rpm (overnight culture)
Antibiotic use: Kanamycin and Streptomycin
Relevant answer
Answer
It looks good to me too. I use AGL1 and GV3101 and those both have a pink coloration when you get a large pellet. They don't look very pink as colonies on plates as the color is faint.
  • asked a question related to CRISPR
Question
1 answer
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
Relevant answer
  • asked a question related to CRISPR
Question
3 answers
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?
Relevant answer
Answer
Thanks for the advice. The knock out mouse is not available but I will try the lentivirus .
Tripti
  • asked a question related to CRISPR
Question
4 answers
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.
Relevant answer
Answer
Basically, if you're publishing in any Nature journal you're obligated to make cell lines available to other qualified researchers (either via public repository or by sending them yourself), either for free or for a "reasonable" handling cost. There might be some extra hoops to jump through if the PI is from a non-profit institution / university and you're trying to acquire them at a for profit company. Also, a lot of fluorescent proteins are patented by private companies, so any use of them by for-profit entities will probably require extra licensing agreements with those companies, even if the cell line itself was created by a non-profit institution.
I think your experience with acquiring said cell line is going to be more related to the specific parties and individual PI involved than the journal publisher.
  • asked a question related to CRISPR
Question
4 answers
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!
Relevant answer
Answer
Using RT-qPCR to validate CRISPR-based knockout (KO) experiments can be highly effective but may also encounter several challenges that need careful consideration and troubleshooting. Here’s a structured approach to effectively use RT-qPCR for confirming gene knockouts and addressing potential issues:
1. Design of RT-qPCR Assays
  • Primer Design: Ensure that the primers for RT-qPCR are designed to specifically amplify the target region affected by the CRISPR KO. Primers should flank the CRISPR target site or span exon junctions to avoid amplification of potential non-targeted isoforms or residual non-edited transcripts.
  • Control Genes: Select appropriate housekeeping genes as controls for normalization. These should be genes known to have stable expression across your experimental conditions.
2. RNA Extraction and Quality Control
  • RNA Quality: Extract high-quality RNA using a reliable method. The integrity of RNA can be assessed using gel electrophoresis or a bioanalyzer. Degraded RNA can lead to unreliable RT-qPCR results.
  • Contamination Check: Ensure that the RNA is free from genomic DNA contamination. Treat the RNA samples with DNase I to remove any potential contaminating DNA that could affect the RT-qPCR results.
3. cDNA Synthesis
  • Reverse Transcription: Use a high-quality reverse transcription kit for cDNA synthesis. The choice between random hexamers and oligo(dT) primers depends on the location of your qPCR primers and the RNA regions you aim to detect.
4. qPCR Reaction Setup
  • Efficiency Validation: Before performing RT-qPCR on your samples, validate the efficiency of your primers. Perform a standard curve analysis using a serial dilution of cDNA to ensure that primer efficiency is between 90% and 110%.
  • Technical Replicates: Include technical replicates for each sample to ensure reproducibility and reliability of the RT-qPCR data.
5. Data Analysis
  • Relative Quantification: Use the ΔΔCt method for relative quantification of gene expression. This method involves comparing the Ct values of the target gene normalized to a reference gene and then comparing the normalized value to a control sample.
  • Statistical Analysis: Perform statistical analysis to compare gene expression levels between CRISPR-edited and control samples. This can help confirm whether the gene knockdown or knockout has been successful.
6. Troubleshooting Common Issues
  • Inconsistent Results: If results are inconsistent, check for primer-dimer formation, non-specific amplification (check melt curves), or issues with RNA quality.
  • Partial Knockdown Observed: Sometimes, CRISPR may not completely knock out a gene but reduce its expression. Consider the possibility of incomplete editing or existence of differentially edited cell populations.
  • No Change in Expression: If no change in expression is detected, verify the CRISPR target site and mutation by sequencing the genomic DNA. It's possible that the CRISPR system did not induce the expected edits, or that compensatory mechanisms in the cell have maintained the expression levels.
Conclusion
Using RT-qPCR to validate CRISPR knockouts involves careful experimental design, precise execution, and thorough data analysis. By addressing each step with meticulous detail—from RNA extraction to data analysis—you can robustly validate the success of CRISPR-mediated gene knockouts and gain insights into the genetic manipulations within your experimental system.
Perhaps this protocol list can give us more information to help solve the problem.
  • asked a question related to CRISPR
Question
6 answers
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
Relevant answer
Answer
The process of single cell clonal expansion of C2C12 cells, a murine myoblast cell line frequently used in muscle biology research, involves isolating and expanding individual cells to form genetically homogeneous colonies. This technique is pivotal for studies requiring uniform cell populations to ensure consistency and reproducibility of experimental results. Here’s a detailed guide on how to perform single cell clonal expansion of C2C12 cells:
Step 1: Preparation
  • Cell Culture Maintenance: Begin with a well-maintained stock of C2C12 cells, ensuring they are healthy and not over-confluent. Cells should be in the logarithmic phase of growth to maximize viability.
  • Reagents and Equipment: Prepare all necessary culture media, plates, and equipment. Ensure that all materials are sterile and the workspace is clean to prevent contamination.
Step 2: Single Cell Isolation
  • Detachment: Harvest C2C12 cells from the culture flask using trypsin-EDTA to detach them. Neutralize trypsin with complete growth medium (e.g., DMEM supplemented with 10% fetal bovine serum) and perform a cell count.
  • Dilution for Single Cell Sorting: Dilute the cell suspension to a concentration where the probability of having a single cell per well is maximized. Typically, this involves diluting to approximately 1-3 cells per 100 µL.
  • Plating: Plate the cells in a 96-well plate, depositing 100 µL per well. It's advisable to use an automated cell sorter or flow cytometer if available, as it can improve the accuracy of depositing single cells into each well.
Step 3: Clonal Expansion
  • Incubation: Incubate the plated cells at 37°C in a humidified atmosphere with 5% CO2. Regularly monitor the wells under a microscope to identify those containing a single cell and to check for signs of cell division and colony formation.
  • Feeding: Change the medium every 2-3 days to provide fresh nutrients and remove metabolic waste. Take care to avoid disturbing the cells, especially in the early stages of colony formation.
Step 4: Subculturing Colonies
  • Colony Selection: Once the colonies have reached a sufficient size (typically visible clusters of cells), they can be gently trypsinized and expanded into larger culture vessels. Select colonies that appear morphologically homogeneous.
  • Expansion: Transfer the expanded cells to larger plates (e.g., 24-well, then 6-well, and eventually T25 flasks) as the colony size increases, maintaining them under standard culture conditions.
Step 5: Validation and Characterization
  • Genetic and Phenotypic Characterization: Perform necessary validation tests to confirm the clonality and homogeneity of the expanded cells. This might include genetic profiling, myogenic differentiation assays, and karyotyping to ensure the cells maintain their characteristic traits.
Conclusion
Single cell clonal expansion of C2C12 cells is a meticulous process that requires precise technique and careful handling to ensure that colonies derived from a single cell grow and proliferate successfully. This method is essential for experiments requiring uniform cellular responses, particularly in genetic studies, drug testing, and muscle physiology research.
With this protocol list, we might find more ways to solve this problem.
  • asked a question related to CRISPR
Question
2 answers
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!
Relevant answer
Answer
Using CRISPR genome editing to generate mixed colonies of Clostridioides difficile (formerly known as Clostridium difficile) could be a challenging task due to the complexity of the organism and the intricacies of CRISPR technology. However, it's theoretically possible with careful design and execution.
Here's a general outline of how you might attempt this:
  1. Design CRISPR Constructs: Design CRISPR guide RNAs (gRNAs) that target specific loci in the C. difficile genome. These gRNAs should be designed to introduce desired mutations or modifications. Consider targeting genes involved in antibiotic resistance, virulence, or other traits of interest.
  2. CRISPR Delivery: Deliver the CRISPR system components (Cas9 protein and gRNA) into C. difficile cells. This can be achieved using methods such as electroporation, conjugation, or phage-mediated delivery.
  3. Selection and Screening: After CRISPR delivery, select for transformed C. difficile cells using appropriate antibiotic selection markers or other selection methods. Screen for colonies that exhibit the desired mutations or modifications using techniques such as PCR, sequencing, or phenotypic assays.
  4. Mixed Colony Formation: To generate mixed colonies, you may need to employ methods that promote genetic heterogeneity within the bacterial population. This could involve introducing CRISPR components at different time points or using CRISPR systems that allow for multiplex editing of multiple loci simultaneously.
  5. Characterization: Characterize the mixed colonies to assess the diversity and stability of the introduced mutations or modifications. This may involve analyzing the genotype and phenotype of individual colonies as well as assessing their competitive fitness in vitro or in animal models.
It's important to note that generating mixed colonies of C. difficile using CRISPR genome editing would require careful optimization of experimental conditions and rigorous validation of the resulting colonies.
  • asked a question related to CRISPR
Question
1 answer
Im using CRISPR edited GM12878 cells to grow the single cell clones.
Relevant answer
Answer
Implementing single-cell cloning by limiting dilution in CRISPR-edited lymphoblastoid cell lines (LCLs) presents challenges and considerations, including variations in CRISPR editing efficiency, cell viability, genetic heterogeneity, selection strategies, and clone validation. While specific literature on this topic might be limited, similar techniques have been successful in other cell types. Successful implementation requires optimizing CRISPR editing protocols for LCLs, thorough validation of clones, and consultation with experts in CRISPR editing and cell biology.
  • asked a question related to CRISPR
Question
3 answers
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? 
Relevant answer
Answer
When conducting experiments involving the lentiCRISPR v2 system for CRISPR-Cas9-mediated gene editing, verifying the correct insertion of the guide RNA (gRNA) sequence is a critical step. This ensures the specificity and efficacy of the gene editing process. To facilitate this, PCR amplification of the gRNA cassette followed by sequencing is commonly performed. Utilizing specific primers that flank the gRNA cloning site within the lentiCRISPR v2 vector is essential for this verification process.
Primer Design for gRNA Insert Verification:
The lentiCRISPR v2 vector, a widely used tool in gene editing, incorporates a U6 promoter driving the expression of the gRNA, followed by a cloning site where the gRNA is inserted. To amplify this region and confirm the presence and correct orientation of the gRNA insert, primers that anneal to sequences flanking the cloning site are used.
Suggested Primers:
  1. U6 Promoter Forward Primer: This primer is designed to anneal to the U6 promoter, which is upstream of the gRNA cloning site. An example sequence for the U6 forward primer is 5’-GACTATCATATGCTTACCGTAACTTGAA-3’. This primer initiates PCR amplification from the promoter region towards the inserted gRNA sequence.
  2. gRNA Cloning Site Reverse Primer: For the reverse primer, it is recommended to use a sequence that anneals to a region downstream of the gRNA cloning site within the vector. A commonly used reverse primer sequence is 5’-AAAAGCACCGACTCGGTGCC-3’. This primer is designed to bind just outside the typical gRNA insert site, ensuring that any PCR product generated includes the entire gRNA sequence.
PCR Amplification and Verification:
  • PCR Conditions: Set up PCR reactions using the above primers, with the lentiCRISPR v2 vector containing the gRNA insert as the template. Optimize the PCR conditions, including annealing temperature and extension time, based on the primer melting temperatures and expected product size.
  • Gel Electrophoresis: Following PCR, run the products on an agarose gel to verify the size of the amplified fragment. The expected size should correlate with the length of the U6 promoter, gRNA insert, and the sequence between the gRNA insert site and the reverse primer binding site.
  • Sequencing: To confirm the sequence of the inserted gRNA, purify the PCR product and subject it to Sanger sequencing. Analyze the sequencing results to ensure the gRNA sequence matches the intended target sequence without mutations or errors.
Conclusion:
The use of well-designed primers specific to the U6 promoter and the region downstream of the gRNA cloning site is essential for verifying the correct insertion of the gRNA in the lentiCRISPR v2 vector. This verification step is crucial for the success of CRISPR-Cas9 gene editing experiments, as it directly impacts the specificity and efficacy of the editing process. By following these guidelines for primer selection, PCR amplification, and sequencing, researchers can confidently validate the presence and accuracy of their gRNA inserts within the lentiCRISPR v2 system.
Take a look at this protocol list; it could assist in understanding and solving the problem.
  • asked a question related to CRISPR
Question
3 answers
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
Relevant answer
Answer
It is difficult to have an single optimal way for every cell lines, each cells have their own strength driving multiple signaling pathway, upper limit of the plasmid copy received and interaction between GOI and its own proteins, hence affecting gene expression facility. We had tried at least four different transfection method with more than ten different reagents just to make a cell population to express 0.1% of GOI, while in 293T the expression was almost hitting the upper limit of detection in flow cytometry. Therefore, to generate an optimized protocol for each cell lines or primary cells are considered easier then to generate a single protocol for universal use. The cloning process or to achieve a single double KI/KO clone by CRISPR is very time consuming and troublesome, if you want to go through CRISPR, try look at the newest technique such as base/prime editing by Dr. David Liu. In the other hand, lentivirus is not, in our lab, lentivirus is an useful tool for large scale cell line production, a single production process may grant us enough amount of lentiviral particle for more then 100 times of usage. However, if a gene is already being difficult to express in such cell lines, I would rather look into an optimization of DNA construct, a small twist such as codon optimization or driving promoter sometimes grant you more then 50-fold of difference, that will be far practical then transfection process.
Best
  • asked a question related to CRISPR
Question
5 answers
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?
Relevant answer
Answer
@Maria I used that ratio for mass
  • asked a question related to CRISPR
Question
4 answers
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!
Relevant answer
Answer
Dear [Recipient],
I hope this message finds you well. It's clear that you are encountering challenges with your CRISPR/Cas9 cloning protocol, specifically in the insertion of your guide RNA (gRNA) sequence into the vector. This step is crucial for the successful application of CRISPR/Cas9 technology, and encountering difficulties can be frustrating. Below, I outline a series of considerations and troubleshooting tips to assist you in resolving this issue.
Assessing the Cloning Strategy:
  1. Vector and Insert Compatibility:Ensure that the vector and insert have compatible ends for ligation. This could be cohesive ends generated by restriction enzymes or blunt ends for blunt-end cloning. Verify that the enzymes used do not cut within your insert sequence.
  2. Restriction Enzyme Efficiency:Confirm the complete digestion of the vector by the restriction enzymes. Incomplete digestion can lead to low cloning efficiency. It might be helpful to use a dephosphorylation step to prevent vector self-ligation.
  3. Insert Preparation:Verify the purity and concentration of the insert. Contaminants from PCR amplification or previous cloning steps can inhibit ligation. Consider running a gel extraction to purify the insert.
Ligation and Transformation Efficiency:
  1. Ligation Conditions:Optimize the molar ratio of vector to insert. A common starting point is a 1:3 vector-to-insert ratio, but this can be adjusted depending on the sizes of the vector and insert and the specifics of your ligation kit. Ensure that the ligation reaction is incubated under optimal conditions recommended by the enzyme supplier.
  2. Competent Cells:The efficiency of transformation can greatly affect cloning success. Use high-efficiency competent cells and ensure they are properly thawed and kept on ice before transformation.
  3. Recovery Phase:After transformation, a proper recovery phase in rich medium allows for the expression of antibiotic resistance genes before plating on selective media.
Verification of Cloning Success:
  1. Colony PCR:Screen transformants by colony PCR to quickly identify colonies that may contain the insert. Design primers that anneal to vector sequences flanking the cloning site.
  2. Restriction Analysis:Perform a restriction digest of plasmid DNA from positive colonies to confirm the presence and orientation of the insert.
  3. Sequencing:Sanger sequencing of plasmid DNA from potential positive clones can confirm the correct insertion and sequence integrity of the insert.
Additional Considerations:
  • Gel Purification: If your insert and vector are of similar sizes and difficult to separate by gel electrophoresis, consider using an alternative method for purification or a different strategy for cloning.
  • Alternative Cloning Methods: If traditional cloning continues to fail, consider using a site-specific recombination system (e.g., Gateway cloning) or a seamless cloning kit (e.g., Gibson Assembly or Golden Gate cloning) that might offer more flexibility and efficiency.
Conclusion:
Troubleshooting cloning protocols requires patience and systematic optimization of each step. By carefully reviewing your protocol and considering each of the points mentioned above, you can identify and rectify the issue hindering your cloning success.
Should you require further assistance or have specific questions at any step of your protocol, please do not hesitate to reach out. I am here to support you in advancing your research projects.
Best regards,
With this protocol list, we might find more ways to solve this problem.
  • asked a question related to CRISPR
Question
7 answers
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
Relevant answer
Answer
Dear [Recipient],
I hope this message finds you well. Your inquiry regarding the use of liposomes for the delivery of Cas9/gRNA ribonucleoprotein (RNP) complexes and double-stranded oligodeoxynucleotides (dsODNs) is both timely and relevant in the context of genome editing technologies. This delivery strategy is increasingly recognized for its potential to facilitate efficient, targeted genome editing by reducing off-target effects and enhancing delivery efficiency. Below, I provide detailed advice and considerations for employing liposomes in this cutting-edge application.
Understanding the Delivery System:
Liposomes are versatile, lipid-based vesicles capable of encapsulating nucleic acids and proteins, facilitating their delivery into cells. Their biocompatibility, low immunogenicity, and ability to merge with cell membranes make them an attractive delivery system for Cas9/gRNA RNP and dsODNs.
Preparing Cas9/gRNA RNP and dsODNs:
  1. Cas9/gRNA RNP Complex Formation:Prioritize the use of high-purity, recombinant Cas9 protein and synthetically derived gRNA. Mix them in a stoichiometric ratio to form the RNP complex. Incubation conditions for complex formation can vary, so optimization may be required.
  2. dsODN Preparation:Ensure that the dsODNs, used as repair templates for homology-directed repair (HDR), are synthesized with high fidelity and purified to remove any contaminants that might affect liposome encapsulation and cell uptake.
Liposome Preparation and Complex Loading:
  1. Liposome Selection:Choose liposomes with a composition and size suitable for RNP and dsODN delivery. Cationic liposomes are often preferred due to their ability to form complexes with the negatively charged nucleic acids and proteins.
  2. Encapsulation Efficiency:Optimizing the encapsulation efficiency is crucial. This may involve adjusting the ratio of liposomes to the RNP/dsODN complexes, the concentration of components, and the incubation conditions. Utilize assays to quantify encapsulation efficiency and adjust protocols accordingly.
  3. Protective Measures:Incorporate strategies to protect the integrity of the RNP and dsODN during the encapsulation process. This might include the use of buffers that stabilize the RNP complex and prevent dsODN degradation.
Cell Targeting and Uptake:
  1. Cell Type Considerations:The efficacy of liposome-mediated delivery can vary significantly between cell types. Characterize the uptake efficiency and cellular toxicity in your target cells, optimizing liposome size and surface charge as necessary.
  2. Enhancing Cellular Uptake:Explore strategies to enhance cellular uptake, such as incorporating targeting ligands into the liposome formulation, which can facilitate receptor-mediated endocytosis in specific cell types.
Monitoring Editing Efficiency and Off-target Effects:
  1. Assessment of Genome Editing:Employ sensitive and specific assays to evaluate the efficiency of genome editing and HDR-mediated insertions, such as T7 endonuclease I assay, PCR-based assays, or next-generation sequencing.
  2. Evaluation of Off-target Activity:Utilize computational tools and experimental assays to predict and assess off-target effects, ensuring that the delivery method does not exacerbate unwanted genomic modifications.
Conclusion:
Liposome-mediated delivery of Cas9/gRNA RNP and dsODNs represents a promising avenue for enhancing the specificity and efficiency of genome editing applications. Success in this approach requires careful consideration of liposome composition, optimization of encapsulation processes, and thorough evaluation of delivery outcomes in target cells. Continued innovation and optimization in this area are expected to further refine these techniques, broadening their applicability and effectiveness in both research and therapeutic contexts.
Should you require further assistance or wish to explore additional aspects of this delivery method, please do not hesitate to reach out. I am here to support your research endeavors.
Best regards,
Take a look at this protocol list; it could assist in understanding and solving the problem.
  • asked a question related to CRISPR
Question
1 answer
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
Relevant answer
Answer
You have two options:
1) As you suggested, to cut out the bands, purify them and directly sequence them with the PCR primers, but the sequence will probably be rather “dirty” because of carry-over contamination with other amplification products.
2) Cut out the bands, purify and clone them, dependent on whether you are using a Taq polymerase with proofreading activity or not in a blunt end or TA vector and sequence a couple of positive clones. Either you conduct colony PCRs with your specific primers or vector primers and directly sequence the correct amplification products with the respective primers; or make plasmid minipreps and sequence them.
  • asked a question related to CRISPR
Question
2 answers
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
Relevant answer
Answer
Ajay Keot : Thanks. Sure will try sequencing PCR product.
  • asked a question related to CRISPR
Question
4 answers
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??
Relevant answer
Answer
Dear Esteemed Colleague,
Greetings. I trust this message finds you well and making significant progress in your research endeavors, particularly those involving the CRISPR-Cas9 system, a powerful tool for genome editing. Your inquiry regarding the selection of a control single-guide RNA (sgRNA) for CRISPR experiments is both critical and insightful, as appropriate controls are essential for validating the specificity and efficacy of your gene editing efforts. Below, I provide a detailed overview of control sgRNA selection and its importance in CRISPR studies.
Understanding Control sgRNAs in CRISPR Experiments
In CRISPR-Cas9 experiments, control sgRNAs serve as benchmarks to assess the baseline activity of the system and to distinguish between specific editing effects and nonspecific cellular responses. There are primarily two types of control sgRNAs commonly employed in CRISPR studies:
  1. Non-targeting Control sgRNA:Definition: A non-targeting control sgRNA is designed to have no sequence homology to any genomic DNA sequence in the model organism being studied. Its purpose is to assess the background level of Cas9 activity and nonspecific cellular responses to CRISPR-Cas9 delivery without inducing specific gene editing. Application: Use this control to monitor potential off-target effects and to differentiate between specific gene editing outcomes and general cellular responses to CRISPR-Cas9 components.
  2. Scrambled sgRNA:Definition: A scrambled sgRNA contains a guide sequence that is randomized in such a way that it does not target any genomic loci in the organism. It serves a similar purpose as the non-targeting control by providing a baseline for Cas9 activity without specific genomic targeting. Application: Scrambled sgRNAs are particularly useful for ruling out sequence-dependent off-target effects and for evaluating the specificity of the phenotypic changes observed in CRISPR experiments.
Selection Criteria for Control sgRNAs
  • Sequence Considerations: Ensure that the control sgRNA sequence does not match any region within the genome of the organism being studied. Bioinformatics tools and databases can be utilized to verify the absence of complementary sequences.
  • Efficiency: The control sgRNA should be efficiently processed by the Cas9 nuclease and the cellular machinery, mirroring the conditions of the experimental sgRNAs.
Best Practices in Using Control sgRNAs
  • Experimental Design: Incorporate control sgRNAs in all CRISPR experiments to serve as references for evaluating gene editing efficiency, specificity, and potential cytotoxicity.
  • Data Interpretation: Compare the outcomes of experiments using target-specific sgRNAs with those employing control sgRNAs to discern specific effects from background noise.
  • Validation: Employ multiple control sgRNAs, including both non-targeting and scrambled variants, to robustly validate the specificity and efficacy of the CRISPR-Cas9 editing.
Conclusion
The use of control sgRNAs in CRISPR-Cas9 experiments is paramount for ensuring the reliability and specificity of gene editing studies. By carefully selecting and incorporating appropriate control sgRNAs, researchers can accurately interpret the outcomes of CRISPR experiments and advance our understanding of gene function and regulation.
Should you require further assistance in designing your CRISPR experiments or have additional questions regarding control sgRNA selection, please do not hesitate to reach out. I am here to support your scientific exploration and contribute to the success of your research endeavors.
Warm regards.
This list of protocols might help us better address the issue.
  • asked a question related to CRISPR
Question
4 answers
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?
Relevant answer
Answer
Dear Esteemed Colleague,
Greetings. I hope this message finds you well and immersed in the dynamic field of genetic engineering and regulation. Your inquiry regarding the differences between the CRISPR-dCas9 system and traditional transcriptional regulatory methods highlights a pivotal area of interest in contemporary research. Below, I provide a detailed comparison that elucidates the distinct features, applications, and implications of these two approaches to gene regulation.
Overview of CRISPR-dCas9 System
The CRISPR-dCas9 (dead Cas9) system is an innovative adaptation of the CRISPR-Cas9 genome editing technology. In this variant, the Cas9 nuclease is catalytically inactivated (dCas9), rendering it incapable of inducing double-strand breaks. Instead, dCas9 is harnessed as a precise targeting mechanism to modulate gene expression when fused to transcriptional activators or repressors, or when employed to physically block transcription machinery access.
Traditional Transcriptional Regulatory Methods
Traditional methods of transcriptional regulation encompass a broad spectrum of techniques, including the use of small molecules, RNA interference (RNAi), and overexpression or knockdown of transcription factors or co-regulators. These methods typically act by modulating the cellular concentration of regulatory molecules or by directly interfering with the transcriptional machinery.
Key Differences
  1. Specificity and Precision:CRISPR-dCas9: Offers unparalleled specificity in targeting gene promoters or regulatory elements due to the programmable nature of the guide RNA (gRNA). This allows for precise modulation of gene expression without altering the genomic sequence. Traditional Methods: While effective, they generally lack the same level of target specificity. For example, small molecules and RNAi can have off-target effects, influencing genes beyond the intended target.
  2. Flexibility and Scalability:CRISPR-dCas9: The system's versatility is evident in its ability to be repurposed for either activation or repression of gene expression, multiplexing to target multiple genes simultaneously, and facile adaptation to diverse organisms. Traditional Methods: Typically more limited in scope for simultaneous targeting of multiple genes. Each target often requires a unique set of molecules or vectors, complicating multiplex applications.
  3. Mechanism of Action:CRISPR-dCas9: Acts by physically targeting the DNA sequence, either blocking transcriptional machinery access or recruiting transcriptional modulators to the site of interest. Traditional Methods: Act through various mechanisms, such as degrading mRNA (RNAi), altering transcription factor activity, or modulating chromatin structure indirectly.
  4. Temporal Control:CRISPR-dCas9: Recent advancements have enabled inducible CRISPR-dCas9 systems, providing temporal control over gene regulation. Traditional Methods: While inducible systems exist (e.g., inducible promoters), achieving precise temporal control can be more challenging and less efficient.
  5. Potential for Off-target Effects:CRISPR-dCas9: Despite its specificity, there remains a potential for off-target binding and regulation, necessitating thorough validation. Traditional Methods: Off-target effects are a well-documented concern, particularly with techniques like RNAi, which can lead to the downregulation of unintended transcripts.
Conclusion
The CRISPR-dCas9 system represents a significant advancement in the field of genetic engineering, offering specificity, flexibility, and precise control over gene expression that surpasses many traditional transcriptional regulatory methods. However, the choice between CRISPR-dCas9 and traditional methods should be informed by the specific requirements of the research project, including the desired level of control, target specificity, and potential for off-target effects.
Should you have further inquiries or wish to delve deeper into the applications of CRISPR-dCas9 in transcriptional regulation, please do not hesitate to reach out. I am here to support your scientific exploration and contribute to the advancement of genetic research.
Warm regards.
Check out this protocol list; it might provide additional insights for resolving the issue.
  • asked a question related to CRISPR
Question
5 answers
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.
Relevant answer
Answer
Dear Esteemed Colleague,
Greetings. I trust this message finds you well and engrossed in the cutting-edge realm of genome editing, particularly employing CRISPR-Cas9 technology. Your inquiry about separating homozygous from heterozygous mutations post-CRISPR is both crucial and timely for ensuring the precision and efficacy of genetic modifications. Below, I provide a comprehensive guide outlining methodologies for distinguishing between these genetic variants, thereby enhancing the resolution of your CRISPR-based experiments.
Overview
The CRISPR-Cas9 system enables targeted genomic alterations with high specificity. Following the introduction of CRISPR-Cas9 components into cells, the repair of the induced double-strand break can result in modifications leading to homozygous or heterozygous alleles. Determining the zygosity of these modifications is essential for characterizing the functional consequences of the edits.
Methodologies for Zygosity Determination
  1. PCR and Sanger Sequencing:Procedure: Amplify the target region surrounding the CRISPR-induced modification using PCR, followed by Sanger sequencing of the PCR products. Analysis: Compare the sequencing chromatogram of the edited sample to that of a wild-type control. Homozygous mutations will show a single, clean peak at the modification site, whereas heterozygous mutations will display overlapping peaks, indicating the presence of both the wild-type and edited alleles.
  2. Restriction Fragment Length Polymorphism (RFLP) Analysis:Procedure: If the CRISPR edit introduces or abolishes a restriction enzyme site, perform a PCR to amplify the target region, followed by digestion with the appropriate restriction enzyme. Analysis: Resolve the digested PCR products on an agarose gel. The pattern of bands will differ between homozygous and heterozygous alleles based on the presence or absence of the restriction site.
  3. T7 Endonuclease I (T7EI) Assay:Procedure: Amplify the target region via PCR and denature and reanneal the PCR products to form mismatched duplexes if both edited and wild-type alleles are present. Treat the reannealed products with T7 Endonuclease I, which cleaves mismatched DNA. Analysis: Homozygous edits will not generate cleavage products, whereas heterozygous edits will result in a cleavage pattern visible on an agarose gel, indicating the presence of mismatches.
  4. Digital Droplet PCR (ddPCR):Procedure: Design allele-specific probes for ddPCR that can distinguish between the wild-type and edited alleles. Analysis: Quantify the absolute number of edited and wild-type alleles. The ratio of these alleles can precisely determine zygosity, with a 1:1 ratio indicating heterozygosity and a predominance of one allele type indicating homozygosity.
  5. Next-Generation Sequencing (NGS):Procedure: Perform deep sequencing of the target region in a population of cells. Analysis: Bioinformatic analysis can quantify the proportion of reads representing the wild-type versus edited alleles, providing a detailed assessment of zygosity at the population level. For clonal populations, NGS can definitively characterize the zygosity of individual clones.
Considerations and Best Practices
  • Clonal Isolation: For precise zygosity determination, especially in heterogeneous populations, isolating single clones and expanding them for individual analysis is recommended.
  • Validation: Confirm the editing and zygosity results using multiple, complementary methods when possible to ensure accuracy.
  • Bioinformatic Support: Leverage bioinformatics tools for analyzing complex sequencing data, particularly when using high-throughput methods like NGS.
By employing these strategies, you can accurately determine the zygosity of CRISPR-induced genetic modifications, a critical step in validating the success of your genome editing endeavors and understanding the functional implications of your edits.
Should you require further assistance or wish to delve deeper into any of these methodologies, please do not hesitate to reach out. I am here to support your research journey and contribute to the advancement of genome editing technologies.
Warm regards.
Take a look at this protocol list; it could assist in understanding and solving the problem.
  • asked a question related to CRISPR
Question
4 answers
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.
Relevant answer
Answer
Dear Esteemed Colleague,
Greetings. I trust this message finds you well and deeply engrossed in the exciting field of genome editing, particularly in the application of CRISPR/Cas9 technology. Your inquiry regarding the significance of the T7 Endonuclease I (T7E1) assay and the role of the HindIII site in the context of CRISPR/Cas9-mediated gene editing is both insightful and timely. These elements play pivotal roles in the optimization and evaluation of genome editing outcomes. Below, I elucidate the contributions and implications of the T7E1 assay and HindIII site in this revolutionary genetic engineering approach.
T7 Endonuclease I (T7E1) Assay in CRISPR/Cas9
The T7E1 assay is a widely used method for detecting mismatches, insertions, or deletions in DNA that result from the non-homologous end joining (NHEJ) repair mechanism following a double-strand break (DSB) induced by CRISPR/Cas9. This assay is instrumental in assessing the efficiency of CRISPR/Cas9-mediated gene editing by identifying the presence of edit-induced mutations.
  • Mechanism: The T7E1 enzyme recognizes and cleaves mismatched heteroduplex DNA structures formed during the annealing of DNA strands that contain mismatches. After CRISPR/Cas9-induced cleavage and imprecise repair, re-annealed DNA molecules at the target site may contain mismatches that are substrates for T7E1.
  • Application: The assay involves PCR amplification of the target region, denaturation and re-annealing of the PCR products to form heteroduplexes, followed by T7E1 digestion and gel electrophoresis. The presence of cleavage products indicates gene editing events.
  • Significance: The T7E1 assay provides a relatively simple, cost-effective, and rapid means to estimate the editing efficiency and to screen for successful CRISPR/Cas9-mediated gene targeting, facilitating the selection of optimal guide RNAs (gRNAs) and conditions for specific genome editing projects.
HindIII Site in CRISPR/Cas9-mediated Gene Editing
The HindIII site, a recognition sequence for the HindIII restriction enzyme, is often used in molecular cloning and can also play a role in CRISPR/Cas9 genome editing, particularly in the design and validation of editing strategies.
  • Cloning Vector Design: In the context of CRISPR/Cas9, plasmids used to express Cas9 and the guide RNA might incorporate HindIII sites for cloning purposes, allowing for the insertion of specific gRNAs or the addition of other genetic elements.
  • Validation of Gene Editing: Introduction or removal of a HindIII site at the target locus can serve as a strategy for validating CRISPR/Cas9 editing. For example, designing a gRNA that leads to the disruption or creation of a HindIII site enables the use of HindIII digestion followed by gel electrophoresis to confirm editing events.
  • Significance: The strategic use of HindIII sites provides a straightforward and efficient method for the molecular confirmation of CRISPR/Cas9-induced alterations, offering a complementary approach to the T7E1 assay for verifying genome editing specificity and efficiency.
In conclusion, both the T7E1 assay and the utilization of the HindIII site are integral components of the CRISPR/Cas9 toolkit, enhancing the precision, verification, and overall success of genome editing endeavors. These methodologies underscore the sophistication and adaptability of current genetic engineering techniques, paving the way for advanced research and therapeutic applications.
Should you seek further elucidation on these topics or wish to explore additional aspects of CRISPR/Cas9 technology, please do not hesitate to reach out. Your engagement in advancing the frontiers of genetic research is highly valued.
Warm regards.
Reviewing the protocols listed here may offer further guidance in addressing this issue
  • asked a question related to CRISPR
Question
5 answers
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.
Relevant answer
Answer
Dear Esteemed Colleague,
Greetings. I hope this message finds you well and thriving in your research endeavors. Your inquiry regarding the cultivation of HEK293T cells following single-cell sorting is of paramount importance for ensuring the success of your experiments. Single-cell sorting and subsequent culture of such cells require meticulous care to ensure viability and promote growth. Below, I provide a detailed guide designed to optimize the conditions for growing HEK293T cells after single-cell sorting.
1. Preparation Before Sorting
  • Cell Condition: Ensure that the HEK293T cells are in optimal condition prior to sorting. This means cells should be healthy, actively growing, and at a low passage number to ensure the best viability and performance post-sorting.
  • Media Preparation: Use fresh, complete growth medium tailored for HEK293T cells, typically DMEM supplemented with 10% fetal bovine serum (FBS) and 1% penicillin-streptomycin. Pre-warm the medium to 37°C to promote immediate cell recovery.
2. Sorting Parameters
  • Gentle Conditions: Opt for the gentlest sorting parameters possible to preserve cell viability. High pressures and narrow nozzles can damage cells.
  • Single-Cell Purity: Adjust the sorter for maximum purity to ensure that sorted droplets contain a single cell. This may require calibrating the instrument before sorting.
3. Post-Sorting Culture
  • Immediate Transfer: Quickly transfer sorted cells into pre-warmed medium to minimize stress and temperature shock.
  • Low Attachment Plates: Consider using plates treated for low attachment or coated with poly-D-lysine to prevent cells from attaching too rapidly, which can cause stress in some sorted cells.
  • High-Humidity Incubation: Culture the sorted cells in a high-humidity incubator set to 37°C with 5% CO2. This environment helps prevent evaporation and maintains stable conditions conducive to cell recovery and growth.
4. Optimal Culture Conditions
  • Reduced Serum Conditions Initially: Some protocols suggest using media with reduced serum (around 5% FBS) immediately after sorting to minimize stress on the cells. After 24-48 hours, when cells have attached and show signs of recovery, the medium can be switched back to the standard 10% FBS formulation.
  • Use of Rock Inhibitor: Including a Rho-associated protein kinase (ROCK) inhibitor in the culture medium immediately post-sorting can enhance survival of single cells by preventing apoptosis.
  • Monitoring and Subculturing: Carefully monitor cell growth and morphology. When cells reach an appropriate density, typically within 7-14 days post-sorting, they can be gently trypsinized and expanded under normal culture conditions.
5. Troubleshooting
  • Low Viability: If viability is low post-sorting, consider optimizing the sorting parameters, the composition of the sorting buffer, and the immediate post-sort recovery conditions.
  • Slow Growth: Single cells, especially after sorting, may take longer to divide and establish a colony. Patience is key, but if growth is exceptionally slow, verify the incubator settings and the freshness and composition of the media and supplements.
Adhering to these detailed tips and maintaining a meticulous approach to the post-sorting care of HEK293T cells will significantly enhance the likelihood of successful culture from single-cell sorts. It is crucial to adapt these guidelines as necessary to fit the specific requirements of your experimental setup and the unique characteristics of your cell line.
Should you have any further questions or require additional insights into cell culture techniques, please do not hesitate to reach out. I am here to support your scientific exploration and contribute to the advancement of your research.
Warm regards.
Perhaps this protocol list can give us more information to help solve the problem.
  • asked a question related to CRISPR
Question
1 answer
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.
Relevant answer
Answer
It depends. If you just want to separate your pool of GFP-positive cells and if fluorescence is always being expressed, flow is sufficient.
Single clones are when you need a specific population. The result is much more accurate, but the work is greater.
  • asked a question related to CRISPR
Question
9 answers
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? 
Relevant answer
Answer
Confirming a knockout generated by the CRISPR/Cas9 system involves several steps to validate the disruption or deletion of the target gene. Here are some common methods used to confirm CRISPR/Cas9-mediated knockouts:
  1. Genomic DNA Sequencing:Sequence the genomic region surrounding the CRISPR/Cas9 target site to identify any insertions, deletions, or mutations (indels) induced by the CRISPR/Cas9 system. Analyze the sequencing data to determine the presence of frameshift mutations or other disruptions that may result in loss of gene function.
  2. PCR Amplification and Gel Electrophoresis:Design PCR primers flanking the CRISPR/Cas9 target site to amplify the genomic region containing the targeted sequence. Perform PCR amplification using genomic DNA from CRISPR/Cas9-treated cells or organisms as a template. Analyze the PCR products by agarose gel electrophoresis to detect any differences in fragment size between wild-type and knockout alleles, such as the absence of the expected PCR product in knockout samples.
  3. Restriction Fragment Length Polymorphism (RFLP) Analysis:Design PCR primers flanking the CRISPR/Cas9 target site to amplify a genomic region containing a restriction enzyme recognition site. Perform PCR amplification using genomic DNA from CRISPR/Cas9-treated cells or organisms as a template. Digest the PCR products with a restriction enzyme that recognizes and cleaves the wild-type allele but not the CRISPR/Cas9-induced mutant allele. Analyze the digested PCR products by agarose gel electrophoresis to detect changes in fragment size indicative of CRISPR/Cas9-mediated mutations.
  4. Mismatch Cleavage Assay (T7 Endonuclease I Assay):Design PCR primers flanking the CRISPR/Cas9 target site to amplify a genomic region containing potential CRISPR/Cas9-induced mutations. Perform PCR amplification using genomic DNA from CRISPR/Cas9-treated cells or organisms as a template. Denature and reanneal the PCR products to create heteroduplex DNA containing mismatches between wild-type and mutant alleles. Digest the heteroduplex DNA with T7 endonuclease I, which specifically cleaves mismatched DNA. Analyze the digested PCR products by agarose gel electrophoresis to detect the presence of cleavage products indicative of CRISPR/Cas9-induced mutations.
  5. Functional Assays:Perform functional assays, such as cell proliferation assays, reporter gene assays, or phenotypic screening, to assess the impact of CRISPR/Cas9-mediated knockout on gene function. Compare the functional characteristics of CRISPR/Cas9-treated cells or organisms with wild-type controls to identify phenotypic changes associated with gene disruption.
By using one or more of these methods, researchers can confirm the knockout generated by the CRISPR/Cas9 system and validate the loss of gene function in the targeted cells or organisms.
l Check out this protocol list; it might provide additional insights for resolving the issue.
  • asked a question related to CRISPR
Question
4 answers
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?
Relevant answer
Answer
The timing for starting selection after transfection of CRISPR vectors can depend on various factors, including the specific experimental setup, the type of CRISPR system used, and the characteristics of the target cells. However, here are some general considerations to help you determine when to initiate selection after transfection:
  1. Transfection Efficiency: Allow sufficient time for the CRISPR vectors to be transfected into the target cells and express the components necessary for genome editing (e.g., Cas9 enzyme, sgRNA). Transfection efficiency can vary depending on the transfection method and cell type, but it typically ranges from a few hours to a day.
  2. Expression of CRISPR Components: The timing for initiating selection should ensure that the CRISPR components have had enough time to be expressed and become functional within the cells. This can range from several hours to a day or more, depending on the kinetics of gene expression and protein production in the specific cell type.
  3. Off-Target Effects: Initiating selection too soon after transfection may increase the risk of off-target effects, where the CRISPR system induces unintended mutations at genomic loci with partial sequence homology to the target site. Allowing sufficient time for specific on-target editing to occur before applying selection pressure can help minimize off-target effects.
  4. Cell Proliferation Rate: Consider the proliferation rate of the target cells when determining the timing of selection. Cells with faster proliferation rates may require shorter selection times compared to cells with slower proliferation rates to achieve the desired level of genome editing.
  5. Optimization: Perform pilot experiments or optimization studies to determine the optimal timing for selection in your specific experimental system. Test different time points for initiating selection and assess the efficiency and specificity of genome editing under each condition.
  6. Balancing Selection Pressure and Cell Viability: Initiate selection early enough to exert adequate selection pressure on cells containing the CRISPR-mediated edits while ensuring that cell viability is not compromised. Too long a delay in starting selection may allow unedited or less efficiently edited cells to outgrow edited cells.
Based on these considerations, it's common to start selection for edited cells within 24-48 hours after transfection of CRISPR vectors. However, the optimal timing may vary depending on the specific experimental setup and cell type. It's essential to empirically determine the best timing for selection in your particular experimental context through pilot experiments and optimization studies.
l This list of protocols might help us better address the issue.
  • asked a question related to CRISPR
Question
3 answers
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. 
Relevant answer
Answer
there is a protocol for knocking out genes in THP1 cells using the CRISPR/Cas9 system. Here's a general outline of the protocol:
  1. Designing sgRNAs: Select target sites within the gene of interest using bioinformatics tools. Aim for regions near the start codon for efficient knockout. Design single guide RNAs (sgRNAs) using online tools like Benchling or CRISPOR.
  2. Constructing sgRNA expression plasmids: Clone the designed sgRNAs into a plasmid vector containing a Cas9 expression cassette. This plasmid is often called a CRISPR/Cas9 vector. Verify the integrity of the constructs by sequencing.
  3. Transfecting THP1 cells: Transfect the CRISPR/Cas9 vector into THP1 cells using a suitable transfection method such as electroporation or lipid-based transfection reagents. Optimize transfection conditions for THP1 cells to achieve high efficiency.
  4. Selecting for edited cells: After transfection, select for cells containing the CRISPR/Cas9 vector. This can be done by including a selectable marker such as antibiotic resistance or fluorescent protein expression in the vector.
  5. Screening for knockout clones: Use assays like PCR followed by sequencing, T7 endonuclease I assay, or Sanger sequencing to identify clones with the desired gene knockout. Verify the frameshift mutations or deletions in the target gene.
  6. Functional validation: Perform functional assays to confirm the loss of gene function in the knockout clones. This may involve phenotypic analysis, such as assessing changes in cell behavior or molecular assays like western blotting or qPCR to quantify gene expression changes.
  7. Single-cell cloning: If needed, perform single-cell cloning to isolate clonal populations of THP1 cells with the desired gene knockout.
  8. Characterization: Characterize the knockout clones thoroughly to ensure that the desired genetic modification has been achieved and to understand any off-target effects.
  9. Maintenance and expansion: Maintain and expand the knockout clones under suitable culture conditions for further experimentation.
Remember to adhere to appropriate safety guidelines and institutional regulations when working with CRISPR/Cas9 technology and genetically modified cells. Additionally, it's essential to consider potential off-target effects and validate the specificity of CRISPR/Cas9 editing in your system.
l Perhaps this protocol list can give us more information to help solve the problem.
  • asked a question related to CRISPR
Question
5 answers
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...
Relevant answer
Answer
Transfecting Raji cells efficiently can be achieved by optimizing several factors, including the choice of transfection reagent, cell density, and transfection conditions. Here's a general protocol along with some suggestions to enhance the efficiency of transfection in Raji cells:
Materials:
  1. Raji cells
  2. Plasmid DNA or siRNA/RNAi oligonucleotides
  3. Transfection reagent (e.g., Lipofectamine 3000, TransIT-X2, or other suitable reagents)
  4. Opti-MEM or RPMI medium
  5. Fetal bovine serum (FBS)
  6. Antibiotics (optional)
  7. Culture dishes or plates
  8. Pipettes and tips
Protocol:
  1. Cell Preparation:Culture Raji cells in RPMI medium supplemented with 10% FBS and antibiotics (if necessary) in a humidified incubator at 37°C with 5% CO2. Ensure that cells are in log-phase growth and have a high viability (>90%).
  2. Transfection Complex Preparation:Dilute the plasmid DNA or siRNA/RNAi oligonucleotides in Opti-MEM or RPMI medium without serum according to the manufacturer's instructions for the chosen transfection reagent. Prepare the transfection complexes by adding the diluted nucleic acids to the diluted transfection reagent. Incubate the mixture at room temperature for 15-30 minutes to allow complex formation.
  3. Cell Transfection:Seed Raji cells in culture plates or dishes at the desired density the day before transfection to ensure that they reach 60-80% confluency on the day of transfection. Replace the growth medium with Opti-MEM or RPMI medium without serum just before transfection. Add the transfection complexes dropwise to the cells and gently swirl the plate to ensure uniform distribution of the complexes. Incubate the cells with the transfection complexes at 37°C in a CO2 incubator for the recommended duration according to the transfection reagent's protocol.
  4. Post-Transfection:After the transfection period, replace the transfection medium with complete growth medium containing serum to support cell recovery and expression of the transfected genes. Incubate the cells for an additional 24-48 hours to allow for gene expression or knockdown to occur.
  5. Analysis of Transfection Efficiency:Analyze the efficiency of transfection by monitoring the expression of a fluorescent reporter gene if using a reporter plasmid or by assessing the knockdown/knockout efficiency of the target gene if using siRNA/RNAi. Perform downstream experiments or analyses to investigate the functional consequences of gene expression modulation.
Optimization Tips:
  • Test different transfection reagents and concentrations to find the most efficient one for Raji cells.
  • Optimize the ratio of DNA or RNA to transfection reagent for maximum efficiency and minimal cytotoxicity.
  • Consider using a selection marker (e.g., antibiotic resistance gene) in plasmid transfections to enrich for transfected cells and improve efficiency.
  • Try different cell densities and seeding conditions to find the optimal conditions for transfection efficiency.
By carefully optimizing these parameters and following the protocol, you can achieve efficient transfection of Raji cells for your experiments.
l With this protocol list, we might find more ways to solve this problem.
  • asked a question related to CRISPR
Question
6 answers
Has anyone had any success with Crispr and MDA-MB-231 or MCF-7 cell lines?
Relevant answer
Answer
Dear Colleague,
Thank you for your inquiry about the application of CRISPR/Cas9 technology in MDA-MB-231 and MCF-7 cell lines. The use of CRISPR/Cas9 for genome editing has indeed been widely adopted across various cell lines, including those relevant to breast cancer research, such as MDA-MB-231 and MCF-7. The success of CRISPR/Cas9 in these cell lines is well documented in scientific literature, underscoring its potential for understanding cancer biology and developing therapeutic strategies.
MDA-MB-231, a triple-negative breast cancer cell line, and MCF-7, an estrogen receptor-positive line, have been utilized in numerous studies to investigate the role of specific genes in cancer progression, metastasis, drug resistance, and other crucial aspects of cancer biology. CRISPR/Cas9 has been employed in these contexts to knock out genes of interest, introduce specific mutations, or modulate gene expression, thereby elucidating their functions in cancer development and progression.
Several key factors contribute to the success of CRISPR/Cas9 in these cell lines:
  1. Efficient Delivery: Achieving high efficiency of CRISPR/Cas9 delivery into MDA-MB-231 and MCF-7 cells is crucial. Techniques such as electroporation, lentiviral transduction, and lipid-mediated transfection have been successfully used. The choice of delivery method can significantly impact the editing efficiency and cell viability.
  2. Guide RNA Design: Designing highly specific guide RNAs (gRNAs) targeting the gene of interest is essential to minimize off-target effects and enhance editing specificity. Online tools and databases can aid in the design and selection of optimal gRNAs.
  3. Selection and Clonal Expansion: After introducing CRISPR/Cas9 components into the cells, selecting successfully edited cells is crucial. This often involves antibiotic selection (for plasmid vectors carrying resistance genes), followed by clonal expansion and validation of gene editing through PCR, sequencing, or functional assays.
  4. Functional Validation: Post-editing, it's imperative to validate the functional consequences of the genetic modifications. This includes assessing changes in cell phenotype, growth, migration, drug sensitivity, and other relevant cancer cell behaviors.
Literature reports demonstrate successful applications of CRISPR/Cas9 in these cell lines, ranging from basic research on gene function to more applied studies on drug resistance mechanisms and potential therapeutic targets. These studies underscore the adaptability and effectiveness of CRISPR/Cas9 in manipulating the genomes of MDA-MB-231 and MCF-7 cells for diverse research purposes.
In conclusion, CRISPR/Cas9 genome editing has been successfully applied in MDA-MB-231 and MCF-7 cell lines, contributing significantly to breast cancer research. The continued refinement of CRISPR/Cas9 methodologies and an increasing understanding of cell line-specific responses to genome editing will further enhance the utility of these cell models in cancer research.
Best regards,
Reviewing the protocols listed here may offer further guidance in addressing this issue
  • asked a question related to CRISPR
Question
6 answers
Limitation:
1) target specific gene:
2) do epigenetic modification:
Relevant answer
Answer
CRISPR/Cas and dCas (dead Cas) systems have revolutionized genome engineering, allowing researchers to manipulate specific DNA and RNA sequences in living cells. However, they come with certain limitations:
  1. Off-Target Activity:CRISPR/Cas systems can sometimes unintentionally edit other genomic regions similar to the target site. These off-target effects may lead to unintended consequences. Researchers continually work on improving specificity to minimize off-target activity.
  2. Insufficient Indel or Low Homology-Directed Repair (HDR) Efficiency:Indels (insertions or deletions) are common outcomes of CRISPR/Cas editing. However, achieving precise edits with high efficiency remains challenging. Homology-directed repair (HDR), which allows precise DNA replacement, is less efficient than indel formation. Enhancing HDR efficiency is an ongoing goal.
  3. In Vivo Delivery of CRISPR Components:Administering CRISPR/Cas components directly into living organisms (in vivo) poses challenges. Efficient delivery methods are crucial for therapeutic applications. Ensuring that the Cas protein and guide RNA reach the target tissue or cell type is essential.
  4. Immune Responses:Introducing foreign CRISPR components can trigger immune reactions. The immune system may recognize and neutralize them. Researchers need to develop strategies to evade immune responses and ensure long-term safety.
  5. Epigenetic Modifications:dCas9, a modified version of Cas9, lacks nuclease activity but retains DNA-binding capability. It can be fused with epigenetic modifiers (e.g., methyltransferases). However, achieving precise and robust epigenetic modifications using dCas9 remains challenging. Improving efficiency and specificity is an active area of research.
  6. Structural Changes and Stability:Large insertions or deletions using CRISPR can disrupt genomic stability. Structural changes may affect gene regulation and overall cellular health.
In summary, while CRISPR/Cas and dCas hold immense promise, addressing these limitations is crucial for their successful application in gene function studies and epigenetic modifications.
  • asked a question related to CRISPR
Question
3 answers
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.
Relevant answer