Science topic

Plasmids - Science topic

Plasmids are extrachromosomal, usually CIRCULAR DNA molecules that are self-replicating and transferable from one organism to another. They are found in a variety of bacterial, archaeal, fungal, algal, and plant species. They are used in GENETIC ENGINEERING as CLONING VECTORS.
Questions related to Plasmids
  • asked a question related to Plasmids
Question
1 answer
Hello all,
I have a mini-prepped DNA that I am trying to digest with EagI. After doing the digestion it looks like the reaction with the restriction enzyme maybe linearizing my plasmid as it migrates at its expected weight when next to the DNA ladder. However the undigested sample runs very high, far above the 10kb band of the ladder. I am guessing this is the uncut is showing a nicked plasmid, which is why it's running so high? What do you think?
Thank you
MB
Relevant answer
Answer
dont worry what it is. do another digest that produces several typical fragments. when you see this patternon the gel the plasmid is definitely the one you are looking for.
  • asked a question related to Plasmids
Question
1 answer
I have designed and assembled a 10kb plasmid containing SbfI and NcoI restriction sites flanking a promoter region, which I am attempting to excise and replace with other promoters. When I do a double digest with both enzymes, I do not get my expected 750 bp band. I have already sequenced my plasmid and it definitely contains both of my restriction sites where I am expecting them.
I have digested my plasmid with SbfI and NcoI individually several times and it looks like each enzyme is working as I am seeing a linearised product on an agarose gel. Both enzymes are HF supplied by NEB and follow the same protocol with the same reaction conditions.
I've tried running the reactions for longer doing 30 min, 1 hr, 3 hr digests. I've used DNA from different (although Sbf is unstable after 1 hr) minipreps, different quantities of DNA (0.5 to 5 ug), different volumes of SbfI (0.5 to 3 ul), brand new restriction enzymes and rCutsmart buffers.
I've included a gel image below. Using a 1% agarose gel ran at 90V for 45 min and a Quick Load Plus DNA ladder.
I don't know what else to try or what else could possible be going wrong.
Any help would be greatly appreciated.
Relevant answer
Answer
Indeed, you tried all possible ways and designed excellent experimental setups. Yet, the following suggestions can be helpful:
1. Please check the enzyme units required for per ug DNA and add accordingly.
2. Since you are not getting 750bp band, may be double digestion is not working. You can still get a linear plasmid band in case of single digestion. To ensure, a reaction can be designed with 3 tubes- first one with SbfI only, second one with NcoI only and third one with both enzymes. Tube 1 and 2 can be of smaller reaction volume. If all three tubes show single band, then we can confirm, both enzymes are working.
3. It is recommended to aliquot buffer in small volumes to avoid repeated freezing and thawing.
4. Although it is HF enzyme, you can try a 100uL reaction volume with an overnight reaction time.
Hope it will work.
  • asked a question related to Plasmids
Question
2 answers
Hello
I am currently engaged in research focusing on plant genetic transformation. As part of this endeavor, I have designed a comprehensive in silico plasmid cloning approach. The aim is to enhance the accuracy and efficiency of our genetic engineering efforts in plants.
I welcome and deeply value input and suggestions from all stakeholders involved in this research. Your insights and expertise are crucial as we strive to optimize our methodologies and achieve impactful results in the field of plant genetic transformation.
Together, through collaboration and exchange of ideas, we can ensure the effectiveness and precision of our approaches, ultimately advancing our understanding and application of plant genetic engineering for various beneficial purposes.
Anyone is invited to write me at [email protected]
Thanks in advance everybody
Relevant answer
Answer
If your question is whether this plasmid will work well in plants, the answer is that it will not. The above plasmid is designed as an E. coli expression vector and only for certain strains of E. coli. The plasmid will not replicate in plants nor will the promoters function unless you also provide the T7 RNA polymerase.
  • asked a question related to Plasmids
Question
2 answers
A plasmid of mine has GOI in opposite orientation. I have another plasmid which has exact same RE site but in such a way that it orients my GOI in right direction. However even after multiple attempts of RE digestion, Gel extraction followed by ligation, I didn't got any colonies on my transformation plate. I also checked efficiency of my competent cell with empty vector and found no issue (got 200-300 colonies) . I would be grateful if anyone can suggest me any possible solution.
Relevant answer
Answer
I agree with the gel extraction being a possible issue. Do you use a column kit or a pellet in resin kit? If it's a column, add an extra wash. If it's a resin/slurry kit, add an extra spin and transfer.
You could also try a different ligase and make sure the buffer is fresh (it does expire, especially if it has been frozen/thawed many times or left out).
I like Quick Ligase from New England Biolabs, you can do plasmid prep, digestion, gel, cleanup, ligate and transform all in one reasonable day.
Good luck!
  • asked a question related to Plasmids
Question
2 answers
Hi,
I'm trying to find a pDNA transient transfection carrier for my MDA-MB-231.
I'm using either liposome or lipopolyplex, but the transfection variability between experiments are too great.
I can only suspect that thin film hydration method has high variability (due to water bath sonication) and changed to ultrasonication which gave 0% transfection (positive ctrl worked, so no probs with pDNA).
Here's the protocol.
1) Thin film made in 4-mL vial or RB or e-tube using rotovap (5 mg/mL lipids in chloroform)
2) Hydration tested with DW/opti-mem/PBS/HEPES using water (1 mg/mL)
3) DNA solution added to liposome solution while vortexing
4) Cells treated with 1 ug DNA/well in opti-mem for 4 hrs, then complete media exchanged or added
For lipoplexes,
1) LPEI solution was added to DNA solution (N/P=10), RT incubation, 30 min
2) Liposome mixture thin film made as above
3) Hydration tested with various buffers or polyplex solution
4) Polyplex solution added to liposome
5) Cells treated with 1 ug DNA/well in opti-mem for 4 hrs, then complete media exchanged or added
I'm already on a number of tries and been frustrated with the result because no matter how consistent I am, the results are different.
Please share your wisdom with me!
Relevant answer
Answer
Why don't you use commercial products? lipofectamin, Mirus, jetpei.... there is plenty of them try the one that is best for your cells...
  • asked a question related to Plasmids
Question
2 answers
In particular using a DNA plasmid transient transfection on differentiated skeletal muscle cells?
Relevant answer
Answer
Yes, you can transfect differentiated cells, including skeletal muscle cells. Transfection of differentiated cells can be more challenging than transfecting undifferentiated cells due to factors such as decreased cell proliferation and altered cellular morphology. However, it's still feasible.
When transfecting differentiated cells like skeletal muscle cells, you may need to optimize your transfection protocol to enhance efficiency. This could involve selecting appropriate transfection reagents and optimizing transfection conditions such as cell density, transfection reagent-to-DNA ratio, and incubation time.
DNA plasmid transient transfection is one method commonly used to introduce exogenous DNA into cells, including differentiated cells. In this method, the DNA plasmid containing the gene of interest is introduced into the cells, typically with the help of transfection reagents. The plasmid DNA can then be expressed by the cells, leading to the production of the desired protein.
Keep in mind that the efficiency of transient transfection can vary depending on the cell type and the specific experimental conditions. Additionally, the expression of the transfected gene may not be as stable or long-lasting as with other methods such as stable transfection or viral transduction.
Overall, while transfecting differentiated cells like skeletal muscle cells may present challenges, it is certainly possible with optimization of protocols and careful consideration of experimental conditions.
Sincerely
  • asked a question related to Plasmids
Question
2 answers
I am trying to clone the cowpea gene (3.5Kb) under the control of the 35s promoter (Gateway plasmid pK7WG2D). I used pDONR222 for the BP reaction. I got the positive colonies, and then sequencing confirmed that my gene was inserted in the donor plasmid. After the LR reaction, I got a lot of positive colonies in the antibiotic (Spectinomycin) plates. But when i do the colony PCR or PCR after plasmid extraction, i don't get amplification with my gene specific primers. However, I get amplification while using backbone-specific primer. I tried multiple protocols and used top10 and DH5a for transformation, but I had the same issue. What could be the problem?
Relevant answer
Answer
Have you tried transforming a negative control for the LR reaction (pK7WG2D destination vector + LR clonase mix but no insert) to see if it gives you a lot of colonies?
If I had to guess, you're getting a high amount clones where attR1 and attR2 have recombined between each other, since they're long inverted repeats that with only 3 mismatching nucleotides. This would allow the vector to close in the absence of an insert.
If that happened, it would delete both ccdB and the chloramphenicol resistance gene (based on this map: https://gatewayvectors.vib.be/collection/pk7wg2d). This would fortunately be easy to verify, as the spectinomycin resistant colonies that you previously confirmed do not contain your gene should be chloramphenicol sensitive, whereas E. coli with the original pK7WG2D plasmid should be chloramphenicol resistant.
I would also recommend ordering fresh enzyme mix, just in case that's part of the issue.
  • asked a question related to Plasmids
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 Plasmids
Question
3 answers
I would like to track in real time the growth of bacteria using fluorescent miscoscopy. I remember there was at least one plasmid that could be used: transfected bacteria would emit a red or green fluorescent signal that could be used to track their growth and position.
Alas, I don't remember what was the name of these plasmids and I can't find a reference in the literature.
Does somebody know these kind of plasmids for live tracking of bacteria? Where can I buy them?
Thank you.
Relevant answer
Answer
What bacterial species are you working with? There's many GFP/RFP plasmids out there, but which ones you use depend on what species you're talking about. Plasmid replication, promoter recognition and codon usage is generally restricted by taxonomy.
Generally, my favorite place to obtain plasmids is AddGene.org
  • asked a question related to Plasmids
Question
3 answers
Hello all. I purified some plasmids after doing precultures of strains from a 2-month old plate in -4 degrees. The PCR results were fine and I used theses plasmids to transform Agrobacterium cells. Should I be concerned about physiological changes for these strains from the old plate or even mutations concerning their genetic material ? (PS: I have the strains in glycerol too and I know that spread from glycerol stock should be a best practice).
Relevant answer
Answer
I wouldn't use 2 month old plates unless there was no other option, personally.
  • asked a question related to Plasmids
Question
8 answers
Hello Everyone! I use Golden Gate technology to build plasmids in my lab. I have a plasmid that I've built that I'm now trying to maxi prep. I've already done a miniprep on this plasmid and sent it for sequencing and it was verified to be what I expected it to be. So I took this DNA and transformed it again to do a maxi prep. For both the Mini and Maxi Prep I used NEB5a cells. I've done the maxi prep on this plasmid twice and each time I'm left with just the antibiotic resistance and promoter. I have no idea where the rest of my insert is going. The GOI, Terminator, introns etc are all disappearing.
The source DNA that I'm using is sequencing correctly. I have no Idea what is happening during the maxi prep process.
Relevant answer
Answer
If you're observing a disappearance of a portion of your plasmid, there are several potential reasons for this phenomenon:
  1. Degradation: Plasmid DNA can be susceptible to degradation by nucleases, especially if it's not stored properly or if it's exposed to conditions that promote enzymatic activity. Ensure that your plasmid is stored in appropriate buffer and conditions to minimize degradation.
  2. Instability: Some plasmids may contain sequences that are unstable, leading to deletion or loss of certain regions over time. This instability can be exacerbated by factors such as the presence of repetitive sequences or sequences prone to recombination.
  3. Selective Pressure: If your plasmid contains a selectable marker, such as an antibiotic resistance gene, there might be selective pressure against maintaining the entire plasmid if the antibiotic is not continuously applied. This could lead to the loss of parts of the plasmid that are not essential for survival under the given conditions.
  4. Replication Errors: Errors during DNA replication, such as slippage or strand misalignment, can result in the loss of DNA sequences. This is more likely to occur in regions with repetitive sequences or secondary structures that can cause replication stalling.
  5. Contamination: If you're working with bacterial cultures containing the plasmid, contamination with other bacteria or phages could lead to the loss of the plasmid or parts of it.
  • asked a question related to Plasmids
Question
1 answer
Hello everyone, in the gel results image provided, the samples in lanes 2 and 5 represent linearized plasmid DNA from groups A and B respectively. Additionally, the samples in lanes 3 and 6 depict circular plasmid DNA from groups A and B respectively. Nothing was loaded to Lane 4. I am curious as to why there seems to be leakage in the sample of lane 6. Also, do you think this leakage occurred immediately after loading or during the gel run? Moreover, could the anomaly observed in lane 4 be linked to the leakage from the sample in lane 6?
I'd appreciate all your inputs.
Relevant answer
Answer
This does not look like leakage but rather that a fiber or something similar is embedded in your agarose gel and/or that you did not properly mix in the gel staining dye before pouring the gel. Make sure that your buffers are not contaminated with anything and that you properly stir in the dye before casting the gel. Also make sure that the running buffer is not contaminated with anything of unknown origin.
  • asked a question related to Plasmids
Question
3 answers
Should I use multiple gRNA transfections (Donor) mixed as well as one by one along with a Cas9 plasmid to see which gRNA is working, or start with only 1 gRNA at a time?
Options:
  1. 1 gRNA + Cas9 (1 gRNA + Cas9 set only)
  2. Multiple gRNAs + Cas9 (2-3 gRNA plasmids + Cas9 plasmid transfection; all combined)
  3. 1 gRNA + Cas9 (1 gRNA + Cas9; 4-5 gRNA and Cas9 each set separate transfection, and one of them may work)
  • or I can use all in one Addgene plasmids and follow 2-3 gRNA-Cas9 plasmids transfection in one go as well as one by one, as per above strategy.
Addgene options I found for all in one plasmids are pSpCas9(BB)-2A-GFP (PX458) and pX330-U6-Chimeric_BB-CBh-hSpCas9.
Which Addgene plasmids for the Cas9 would be ideal for any gene? I can clone only the gRNA sequence in the donor plasmid (Addgene) or order from a supplier. For gRNA, can I use any commercial or addgene cloned plasmids (please share a link)? What website do you believe would be the best to get the gRNA for the gene of interest?
Please help with the making this decision if you have experience with these experiments and what would be the best path to go with.
Relevant answer
Answer
CRISPR-Cas9-mediated gene deletion is a powerful tool for genome editing, allowing precise removal of specific genomic regions. The best approach for CRISPR-Cas9-mediated gene deletion may vary depending on the specific experimental requirements, cell type, and desired outcome. However, careful design of gRNAs, efficient delivery of CRISPR components, thorough screening and validation, and rigorous off-target analysis are key considerations for successful gene deletion experiments. Additionally, optimization and iterative refinement of experimental procedures can help improve the efficiency and reliability of CRISPR-Cas9 editing.
  • asked a question related to Plasmids
Question
5 answers
Next step is doing qPCR.
Thanks
Relevant answer
Answer
Separating genomic DNA and plasmid DNA from algae can be achieved through a process called DNA extraction. Here's a general guide on how you can do it manually:
Materials Needed:
  1. Algal cells
  2. Lysis buffer (containing Tris-HCl, EDTA, SDS, and sometimes proteinase K)
  3. Phenol:chloroform:isoamyl alcohol (25:24:1)
  4. Chloroform:isoamyl alcohol (24:1)
  5. Ethanol
  6. Isopropanol
  7. RNase A (optional, if RNA contamination is a concern)
  8. TE buffer (Tris-EDTA buffer)
Procedure:
1. Cell Lysis:
  1. Harvest the algae cells by centrifugation and wash them with PBS buffer to remove any contaminants.
  2. Resuspend the algal cells in lysis buffer and incubate at an appropriate temperature for cell lysis. This buffer should break open the cells and release the DNA.
  3. Optionally, you can add RNase A to the lysis buffer to degrade any RNA that might be present.
2. DNA Extraction:
  1. After cell lysis, add an equal volume of phenol:chloroform:isoamyl alcohol to the lysate.
  2. Mix the solution thoroughly by inverting the tube gently.
  3. Centrifuge the mixture at high speed to separate the aqueous phase (containing DNA) from the organic phase.
  4. Carefully transfer the aqueous phase (top layer) to a new tube, avoiding the interface.
  5. Repeat the phenol:chloroform:isoamyl alcohol extraction step to ensure complete removal of contaminants.
  6. Precipitate the DNA by adding 2-2.5 volumes of cold ethanol to the aqueous phase.
  7. Incubate the mixture at -20°C or -80°C for about 30 minutes to allow DNA precipitation.
  8. Centrifuge the mixture at high speed to pellet the DNA.
  9. Carefully remove the supernatant and wash the DNA pellet with 70% ethanol to remove any residual salts and contaminants.
  10. Air dry the DNA pellet or use a vacuum concentrator to remove the ethanol completely.
  11. Resuspend the DNA pellet in TE buffer or nuclease-free water. This will be your total genomic DNA extract.
3. Plasmid DNA Separation (Optional):
  1. The plasmid DNA, being smaller in size, remains in the supernatant during the ethanol precipitation step. You can perform an additional precipitation step using isopropanol to selectively precipitate the plasmid DNA.
  2. Precipitate the plasmid DNA by adding 0.6 volumes of isopropanol to the supernatant.
  3. Incubate the mixture at -20°C or -80°C for about 30 minutes.
  4. Centrifuge the mixture at high speed to pellet the plasmid DNA.
  5. Wash the DNA pellet with 70% ethanol and air dry or vacuum concentrate as before.
  6. Resuspend the plasmid DNA pellet in TE buffer or nuclease-free water.
Notes:
  • Ensure proper handling of hazardous chemicals and biological materials.
  • Maintain sterility during the procedure to avoid contamination.
  • Adjust the protocol based on the specific characteristics of your algae species and the intended downstream applications of the extracted DNA.
  • asked a question related to Plasmids
Question
1 answer
I found a kit that they separated plasmid and genomic DNA form mammalian cell, but I want to extract both plasmid and genomic DNA manually.
I also want to use the extracted DNA for qPCR.
Relevant answer
Answer
Any process you use to separate plasmid from genomic DNA is only going to be an enrichment and not an absolute purification. In other words there will always be some level of cross contamination. For some applications this won't matter but PCR will always pick up the background contamination.
Depending upon wha you are trying to do, you may not actually need to separate the plasmid from the genomic.
  • asked a question related to Plasmids
Question
2 answers
Anyone has the pAd∆F6 (or pAd Delta F6) helper vector plasmid map?
I did miniprep, and want to check this helper vector by restriction enzyme digestion.
Thanks,
Gang
Relevant answer
Answer
I do not have direct access to specific plasmid maps or databases, but I can guide you on how to find the pAdΔF6 (or pAd Delta F6) helper vector plasmid map. The pAdΔF6 vector is commonly used in adenoviral vector production and is critical for providing necessary adenoviral genes that are deleted in the main adenoviral backbone plasmid.
Here are steps you can follow to obtain the plasmid map:
  1. Contact the Source Laboratory: If the plasmid was mentioned in a research paper or you know which lab developed it, contacting the laboratory directly is often the best way to obtain detailed information, including the plasmid map. Researchers generally share these materials upon request, sometimes with a material transfer agreement (MTA).
  2. Search Plasmid Repositories: Repositories like Addgene, a nonprofit organization that distributes plasmids for researchers, often have detailed plasmid maps and sequence information. You can search the repository by plasmid name or use keywords related to your plasmid.
  3. University or Institutional Biorepositories: Some universities and research institutions maintain their own collections of plasmids and other genetic tools. If the vector was developed within an academic setting, it might be available through such a repository.
  4. Literature Search: Look for publications or supplementary materials where the plasmid might have been used or described. Authors often provide detailed methods, and plasmid maps may be included in the supplementary files.
  5. Online Forums and Networks: Researcher networks or online forums such as ResearchGate or protocols.io can be helpful. Researchers often discuss their methods and share tips on vector construction and application. You can post inquiries or search for posts related to the pAdΔF6 vector.
  6. Commercial Suppliers: If the plasmid is used widely, commercial vendors of research reagents might offer the vector or could provide information on where to obtain it. Checking their websites or contacting customer service might yield useful information.
By following these strategies, you should be able to obtain the plasmid map for the pAdΔF6 helper vector. If you find that the information is not readily available or if the search seems too complex, consider reaching out to colleagues who may have experience with adenoviral vector systems, as they might offer additional insights or resources.
Perhaps this protocol list can give us more information to help solve the problem.
  • asked a question related to Plasmids
Question
6 answers
Hi
I'm using qiagene kit to extract my plasmids DNA and recently I encountered problems to purify a lentivirus backbone plasmid (pMDLg/pRRE 8895 bp , Addgene=12251) from maxiprep.
I had no problems with miniprep. After I inoculated miniprep bacteria into big flask for large prep, bacteria grew well, every step looked fine but when I run DNA gel I found that there was only one band for uncut plasmid (looked like being linearized, but i did not add in any enzyme!) and there was smear in digested plasmid lane. To test whether my maxiprep kit is working or not, I have done maxiprep for another plasmid (pRSV-Rev 4174bp,pMD2.G 5824 bp) concurrently and it was alright for digestion of this plasmid after maxiprep, which means nothing went wrong in my maxiprep. I cannot figure out what the
problem is. I used same antibiotics (ampicilin), same LB, same maxiprep kit to prepare both constructs but I failed to produce the larger, lower yield plasmid. It is important to highlight that I have ever used the same kit to produce the lentivirus backbone plasmid for 3-4 times, I just encountered the problem of maxiprep in recent one month. So what makes my large prep plasmid being linearized/degraded during maxiprep? I have tried to pick more colonies from my amp plate and cultured with LB in small volume for miniprep and all clones are positive. I cannot understand why it cannot be purified with maxiprep kit? Anyway I hope I can solve this mystery asap with helps from you !!!
Thanks in advance :D
Relevant answer
Answer
Certainly, purifying a lentivirus backbone plasmid using a Maxi Prep kit can present challenges, typically related to yield and purity of the plasmid DNA. Here are some steps and tips to help improve the purification process:
  1. Starting Culture Conditions: Begin with a fresh, well-isolated colony of bacteria that contains your plasmid. Use a bacterial strain optimized for plasmid production, such as Stbl3 or Endura, which are specifically designed to stabilize toxic plasmids and those with repetitive sequences.
  2. Culture Volume and Growth: Ensure that the culture volume and bacterial density are adequate. For a Maxi Prep, typically 100-500 mL of culture is recommended. Allow the culture to grow to an optimal density (OD600 of 0.6-0.8) before harvesting the cells.
  3. Cell Lysis Optimization: The lysis step is crucial. Ensure complete resuspension of the bacterial pellet in the provided P1 buffer before adding the lysis buffer (P2). After adding the lysis buffer, mix gently by inverting the tube 4-6 times. Do not vortex, as this can shear the DNA. Also, strictly adhere to the recommended time for the lysis step; over-lysis can lead to genomic DNA contamination and reduced plasmid purity.
  4. Neutralization and Clearing: Use the provided neutralization buffer (P3) and mix immediately and thoroughly after addition to avoid localized precipitation. Centrifuge at a high speed as recommended by the kit to pellet cell debris effectively. Ensure the supernatant is clear before proceeding to the purification column to prevent clogging.
  5. Binding Conditions: The efficiency of DNA binding to the column can be affected by salt concentration and pH. Make sure that the binding buffer is well mixed and that the pH is within the recommended range.
  6. Washing Steps: Perform all washing steps as indicated in the protocol to remove impurities effectively. This typically involves multiple washes with a high-salt buffer followed by a wash with a low-salt buffer.
  7. Elution Efficiency: Elute the plasmid DNA in a smaller volume than suggested to increase the final DNA concentration. Ensure that the elution buffer is pre-warmed to 50-60°C to increase the yield.
  8. Verification: After purification, assess the quantity and quality of the plasmid using UV spectrophotometry to measure the A260/A280 ratio (ideal ratio is 1.8-2.0 for pure DNA) and running an aliquot on an agarose gel to check for the presence of supercoiled plasmid without degradation.
If problems persist, consider troubleshooting elements such as the health of the bacterial culture, the age of the reagents, or even the type of plasmid backbone, as some sequences may be more prone to degradation or recombination in bacteria.
Take a look at this protocol list; it could assist in understanding and solving the problem.
  • asked a question related to Plasmids
Question
3 answers
Here's the situation: I am currently using 6-Well plates & HEK293t cells with DMEM +10% FBS + 1% P/S and OptiMEM w/ Lipofectamine 2000 for my transfection. Before transfection when cells are 60-75% confluent, I usually change media by adding 2 mL fresh, warm media via 1 mL pipet (therefore 2x). But when doing so, cells detach very easily from the edges of the wells. Probably about 75% stay attached, but these are more localized to the center. I am transfecting in a specific plasmid at low concentration, so I am worried that this detachment will cause lower transfection efficiency in my cells and this plasmid won't get expressed due to low input. Should I redo these replicates?
Relevant answer
Answer
Regarding assessing the expression of specific mutants, I typically refer to primers used in several papers that describe the design of specific primers for detecting mutant expression. In addition, you can consider to perform RT-qPCR using two sets of primers: one targeting the wildtype sequence, and the other targeting the mutant sequence.
  • asked a question related to Plasmids
Question
5 answers
I am currently trying to generate expression constructs of truncated proteins for x-ray crystallography. It is my first time doing cloning. I have had success so far in PCR amplifying my inserts using primers with NdeI, Bam HI, XhoI, and EagI restristion enzyme sites. My amplified inserts are running at the correct molecular weight as confirmed by agarose gel. I always ethanol precipitate my PCR produce overnight and resuspend the DNA pellets in 10mM Tris pH 8.0 and then do a double digest of the insert using NEB's restriction enzyme cloning tool. I typically digest my inserts for ~4-6 hours and then ethanol precipitate the digestion reaction. At the same time I do a double digest my pet28 or pet 15 vector with the same restricion enzymes under similar conditions, I have checked that each enzyme has linearized my plasmid on a gel before I add the other enzyme and then combine the digestion reactions. The uncut plasmid migrates slower than the digested plasmid and the digested plasmid runs at its expected molecular weight. I also ethanol precipitate my vector after the digestion. Finally, I use T4 DNA ligase from NEB and perform the ligation reaction. I then transform 200uL of competent cells with 10uL of the ligation reaction usually. I have tested my comp cells and used a control plasmid and was able to get good transformation efficiency with only 1ng of DNA and 50uL of comp cells but since the ligation reactions tend to give little colonies I have scaled up the transformation. I mini-prep some colonies from the transformation and have done double digests for over 100 colonies now and I do not ever see a band corresponding to my insert being there. Lately I have been doing a single digest to see if I can see the plasmid molecular weight increase as a result of the insert being there, but I do not think I see an insert also. For my most recent ligation I did a 7:1 molar ration of insert to vector in a 20uL reaction with ~20ng of vector. I have also tried 50ng of vector and 20:1 10:1, 1.2:1 ratios and still i get no insert. My most recent transformation for example gave 3 colonies for my insert reaction, 0 colonies for my control of no insert +ligase, and 5 colonies for my control reaction with no insert or ligase. I haven't digested the mini-preps yet but I feel like I will not see an insert again. I would say these results have been typical for my ligations, sometimes I get ~7-27 colonies for my reaction with the insert and 0 colonies for both of my no insert controls. When I test the colonies though, I do not see an insert or shift in molecular weight indicating the insert is not in my vector. Does anyone have any suggestions how I can get a clone successfully ?
Relevant answer
Answer
Hi,
for PCR digestion I generally prefer to purify the PCR product on column directly (there is conditions where you can get rid of the primers and primer dimers) before digestion then repurify (on column if the PCR product is clean or on agarose gel if there is other unwanted bands (or improve your PCR)). For the vector you can do the same (to get rid of the multicloning site fragment after double digest. then when you clone with two different restriction sites you don't need to dephosphorylate (the vector will not recircularize if correctly digested) you don't need neither to have a vast excess of fragemnt/vector ratio 2/1 will be enough, then if there is a restriction site in the vector in between the two restriction sites you are using (and not present in your insert) you can also digest the ligation reaction (after inactivating the ligase 65°C 10min) with .5ul of this enzyme; linearized plasmid do not give transformant...
  • asked a question related to Plasmids
Question
4 answers
Greetings for all of scientist using this platform. I have a little problem. Recently I had done reconstruction of my plasmid (Named PDR111, length = 11,8 kb). Transformed culture i named it W1 Transformant. After the transformation being done, I isolated the plasmid with Geneaid Presto Mini Plasmid Kit and i had done electrophoresis after i got the isolated plasmid. The results will be displayed, PDR111 is my plasmid before reconstruction (circular) as a negative control. As you can see, the band from transformed product seems to be nicked or linear. Does its mean that my transformation success? Because my supervisor told me that isolated plasmid from Presto Kit usually circular. Is it possible that my transformation product be nicked/linear plasmid? Please answer me, thank you
Relevant answer
Answer
Jeremy Mullesa I think your plasmid is ok. PDR111 is overloaded, therefore, it appears to run faster and looks smaller/different from your clone. Load equal amounts. To ensure you have the correct fragment cloned, sequence your construct.
Kais Khudhair al Hadrawi you answer is off-topic and simply generated by ChatGPT. The RG community for sure knows how to use this tool.
  • asked a question related to Plasmids
Question
4 answers
I'm currently trying to capture a biosynthetic gene cluster using Transformation-Associated Recombination (TAR) in yeast. After identifying positive yeast clones, I extract, via an alkaline lysis method, the plasmid from yeast and electroporate it into E. coli DH10B.
However, I have not been able to find any positive hits upon cPCR on Eci clones despite testing about 100 colonies using the same diagnostic primers I used to identify the yeast positive clones. Then, on some of the E coli that I pick and miniprep, I consistently only see my original capture vector (with no gene cluster inserted).
The issue may lie in the yeast plasmid extraction, the transformation, or the plasmid isolation prep from E coli. I know that yeast plasmid extractions are hardly ever clean and tend to be "dirty," contaminated with yeast gDNA and other DNA it has inside due to the IPA precipitation required to perform the method. That tends to lead to poor transformation efficiency into E coli. But I feel like I'm stuck going in circles trying to bring the plasmid into E coli and isolating it. if anyone has ever worked with TAR and has experienced any troubleshooting at this stage of the process, I would appreciate insight. Or any advice on things I can try to better improve yeast plasmid extraction or transformation/isolation in E coli. Many thanks!
Relevant answer
Answer
Transforming and isolating a large plasmid in E. coli can sometimes be challenging, but there are several troubleshooting steps you can take to optimize your protocol:
  1. Quality of Plasmid DNA: Ensure that the plasmid DNA you are using for transformation is of high quality and free from contaminants. Purify the plasmid using a reliable method such as commercial kits or CsCl gradient centrifugation.
  2. Transformation Efficiency: Optimize the transformation efficiency by using competent E. coli cells that are prepared fresh and are highly competent. You can prepare your own competent cells or purchase commercially available ones.
  3. Transformation Protocol Optimization: Review your transformation protocol and ensure that it includes proper heat shock conditions, incubation times, and recovery steps. Optimizing these parameters can significantly improve transformation efficiency.
  4. Plasmid Copy Number: Large plasmids may have lower copy numbers compared to smaller ones, making them harder to isolate. Consider using bacterial strains that are optimized for maintaining large plasmids, such as certain E. coli strains like DH10B or DH5α.
  5. Selection Markers: Ensure that your plasmid contains appropriate antibiotic resistance genes for selection. Use antibiotics at the correct concentrations to select for transformed colonies while suppressing growth of non-transformed cells.
  6. Screening for Transformants: After transformation, streak the transformed cells on selective agar plates containing the appropriate antibiotic. Incubate the plates overnight at the optimal temperature for E. coli growth.
  7. Isolation of Large Plasmids: Large plasmids may require special isolation techniques to prevent DNA shearing during purification. Consider using methods such as alkaline lysis followed by cesium chloride (CsCl) gradient centrifugation or commercial kits designed for isolating large plasmids.
  8. Verification: Once you have isolated the plasmid DNA, verify its size and integrity by running it on an agarose gel or using other analytical techniques such as restriction digestion or sequencing.
By carefully optimizing each step of the transformation and isolation process and troubleshooting any issues that arise, you can increase your chances of successfully transforming and isolating large plasmids in E. coli.
  • asked a question related to Plasmids
Question
4 answers
Hi all,
I want to have 301 copies/ul for my downstream experiments. The sample I am using is a purified plasmid, of which I know the concentration (ng/ul), volume (ul), and molarity(g/mol). How can I get my desired DNA copies?
Relevant answer
Answer
Creating desired DNA copies typically involves a process called DNA amplification, commonly done through polymerase chain reaction (PCR) in molecular biology labs. Here's a simplified overview of how you might go about it:
  1. Design Primers: Primers are short DNA sequences that bind to the target DNA you want to copy. You need two primers, one for each end of the sequence you want to amplify. These primers are designed to match specific regions of the target DNA.
  2. PCR Setup: Prepare a PCR reaction mixture containing the DNA sample you want to amplify, primers, DNA polymerase enzyme, nucleotides (the building blocks of DNA), and buffer solution. This mixture is then placed in a thermal cycler, a machine that can precisely control temperature changes.
  3. PCR Cycling: The PCR machine heats and cools the reaction mixture in cycles. Each cycle typically consists of three steps:Denaturation: The reaction mixture is heated to around 95°C, causing the double-stranded DNA to separate into single strands. Annealing: The temperature is lowered to around 50-65°C, allowing the primers to bind (anneal) to their complementary sequences on the target DNA. Extension: The temperature is raised to around 72°C, and the DNA polymerase enzyme extends the primers by adding nucleotides, creating new DNA strands complementary to the target sequence.
  4. Multiple Cycles: The cycling process is repeated typically 20-40 times, resulting in exponential amplification of the target DNA sequence. After each cycle, the number of DNA copies doubles.
  5. Analysis: Once the PCR is complete, you can analyze the amplified DNA using various techniques such as gel electrophoresis or sequencing to verify the presence and quality of the desired DNA copies.
This process allows you to generate millions to billions of copies of a specific DNA sequence from a tiny amount of starting material, enabling various applications such as genetic testing, sequencing, cloning, and gene expression analysis.
  • asked a question related to Plasmids
Question
3 answers
I want to copy a target gene from cDNA into a plasmid. The primers were designed according to the CDS sequence from NCBI. But when I performed PCR reactions I could not get any target bands. So the CDS sequence was synthesized into my plasmid vector. When used the plasmid as template and the above primers to run PCR, target bands were quite clear which means the primers can work. I know the gene copy number in plasmid must be much higher than in cDNA. So I increased the amount of cDNA template and cycle numbers (from 35 to 45 cycles), no target bands showed. Could anyone tell me what the problem might be. Is that possible that the CDS sequence in my cells has changed? If yes, is there any other ways to get the CDS sequence except artificial synthesis.
Relevant answer
Answer
Robert Adolf Brinzer Thanks a lot. I will try again.
  • asked a question related to Plasmids
Question
5 answers
i need some help setting up my culture plate having negative controls,I am going to infect HEK293T cells in a 6-well plate... to one well I will add my lentivirus particles(packaged already), and to 2nd well, I will not anything and let only HEK293 T cells grow, and to the 3rd well can I add the empty lentivirus vector(having GFP and PUROMYCIN gene)? if yes,so will I add it without the packaging and envelop plasmids? and how this 3rd well can act as a control.
Relevant answer
Answer
If you really want to avoid gfp, and you don't have any other plasmid to substitute it, then I would recommend transfecting only the other 2 plasmids. At least this way the control cells will go through the transfection process.
  • asked a question related to Plasmids
Question
7 answers
I am currently expressing a recombinant protein through the transduction of my plasmid into HEK 293T cells. Historically, I've encountered minimal issues and achieved high yields of protein at a satisfactory concentration. However, I've recently faced challenges as the protein expression has significantly decreased. Alongside investigating the underlying cause, I am interested in concentrating the protein obtained from my recent experiments. I am considering utilizing the Amicon Ultra for this purpose. However, I require clarification on the protocol specifics. Which buffers are recommended for this procedure? Additionally, I seek guidance on the recovery process for the concentrated protein. Your insights would be greatly appreciated. Thank you!
Relevant answer
Answer
the use of amicon ultra is quite simple.
You can equilibrate the amicon ultra with the buffer in which your protein is stored and stable, add the protein, centrifuged and recover the concentrated protein from inside. For small volume amicon, eg 0.5ml, you can recover the protein by inverting upside down the membrane in an empty tube and centrifuging at low speed (e.g 1000g) while for higher volumes, eg amicon ultra-4 or amicon-ultra 15 you can directly recover the protein from inside using a gilson pipette or similar (200ul tips is suggested, since 1000ul tips is to big)
In the following link of my blog, you can find some tips about a possible mistake that you can do in ultrafiltration
best
Manuele
  • asked a question related to Plasmids
Question
4 answers
I tried several times using the Lipofectamine 2000 reagent, extracted the proteins but couldn't detect on Western blot. My plasmids were constructed with PcDNA 3.1+ Please does anyone have any suggestions? Thank you.
Relevant answer
Answer
Thank you Sir, Bruno Salomone Gonzalez de Castejon, we changed the method and got positive results. We still used the HEK 293 cells but with the PEI 40K reagent this time.
  • asked a question related to Plasmids
Question
2 answers
My experiment requires using blasticidin selection marker in lentivirus transduction and I did cut the sequence of interest with restriction enzyme and insert it into the backbone carrying BSR (blasticidin resistant gene). The backbone previously worked very well, and with a concentration of 10ug/mL of blasticidin, I got my stable cell line. However, this time, the transduction seemed not to work and all the cells in titration were killed by blasticidin. I first thought the concentration too high for selection. Though I switched concentration of blasticidin to 5ug/mL, no cell in transduction group could survive, which suggested something more crucial, like plasmid design, might go wrong. Here is a short sequence showing the cut sites of both plasmids for getting the insertion part and BSR backbone. Both plasmids worked very well previously.
The yellow marked region is same in both donor plasmid and BSR backbone. (including Hind III site)
The insertion part has a longer gap between its ORF and IRES. The shorter gap shows region of BSR-carrying backbone. (including Not I site)
I cut them by Hind III and Not I, and after midi prep I verified the BSR marker is inside the ligated product.
The packaging region for lentivirus is within the length limit.
Protocol for producing lentivirus was same with my previous experiment wherein same backbone was used.
Does the gap length between 1st ORF and IRES influence the expression of 2nd ORF? Though the donor plasmid can well express its selection marker?
Relevant answer
Answer
Michael Marino As for the vector size, the recombinated plasmid is same size with either donor plasmid or backbone plasmid, around 7kbp.
Thank you for your suggestion on RT-qPCR. I will check the virus product sufficiency.
  • asked a question related to Plasmids
Question
3 answers
When making a substitution to a plasmid using Q5 should I use methylated plasmid as my template? If I have a plasmid purchased from a vendor should I first transform it into E.coli?
Thank you for your suggestions. DL
Relevant answer
Answer
thank you. very helpful
  • asked a question related to Plasmids
Question
1 answer
Hello. I'm writing with courage to seek advice from professors and researchers. Currently, I am researching the impact of introducing a B plasmid into E. coli carrying an A plasmid, to study the effect of B genes on A genes.
After transforming the A plasmid (ampicillin resistance) via electroporation, I culture the cells to create electroporation competent cells, and then perform electroporation transformation with the B plasmid (gentamicin resistance).
While transforming A and B separately into DH10B Competent cells works well, colony formation does not occur when transforming B into A-harboring competent cells.
A plasmid: ColE1 origin, B plasmid: oriV, so there should be no incompatibility issues.
I wonder if adding ampicillin (1X, 100mg/ml) during the culturing process after transforming A could affect the cells. I tried dividing the cultures into small cultures, always adding 1x ampicillin, and when doing large cultures, I tried not adding antibiotics, or adding them at 0.2x concentration, but in all cases, transformation hardly occurs.
Should I consider anything else? Could co-transformation be the solution?
I would greatly appreciate your help. Please let me know if you need more information for your response.
Relevant answer
Answer
It is hard to know what the problem is, could the genes you have cloned on the plasmids somehow be incompatible?
But you could try to reverse the order (do plasmid B first).
Secondly co-transformation usually works fine, however what you are doing should also work. So if there is some compatibility problem then it would occur regardless.
As a control you could try to transform the plasmid B parent plasmid into your competent cells.
  • asked a question related to Plasmids
Question
2 answers
Lipofectamine is a commonly used transfection reagent known for its efficiency in delivering nucleic acids into mammalian cells. However, the optimal transfection conditions can vary depending on cell type, transfection reagent, and experimental setup. Therefore, I am seeking advice from the scientific community on the specific Lipofectamine protocol that would be most effective for transfecting plasmid DNA into PC3 Eb KO cells. Any insights, recommendations, or protocols shared will greatly contribute to the success of my research project."
Relevant answer
Answer
I wanted to extend my heartfelt thanks for taking the time to respond to my question. Your insights were incredibly valuable and have provided me with a deeper understanding of the topic. Your willingness to share your expertise is truly appreciated.
  • asked a question related to Plasmids
Question
3 answers
I am trying to clone a large (11kb) and toxic gene in an expression vector. Therefore, I am using the EPI400 CopyCutter strain that is maintaining the plasmid at a low copy number to allow bacteria growth. Attached is the protocol for this strain.
I have to grow the bacteria on plate at 28 C for 2 to 3 days to be able to get colonies.
I did a colony PCR and got 20 positive colonies.
When I put them in liquid culture (5mL to 10mL), some colonies never grown, even at 28 C. Some of them did grow (2 days incubation 28C, 250rpm in aeration culture tube, OD600 above 2.0), but then I only get a really low yield after induction of the high copy production in 100mL culture in flasks.
I tried this several times but still got a really low yield (10ng/uL, maximum 15).
I used this strain for other toxic genes and was able to get more than 40ng/uL (which is around what I need to be able to sequence my plasmid and verify the correct insertion of my gene).
I am using this kit for plasmid extraction Wizard® Plus SV Minipreps DNA Purification Systems
Any suggestions on how I could improve my plasmid yield?
Any suggestions to be able to grow the bacteria in liquid culture after getting them selected on the plate?
Thank you.
Relevant answer
Answer
To optimize plasmid DNA yield when working with toxic genes:
  1. Consider using a lower copy number plasmid backbone to reduce the burden on host cells.
  2. Adjust induction conditions to balance gene expression and cell growth.
  3. Replace the promoter with a weaker or less toxic one, if possible.
  4. Experiment with different growth temperatures to find the optimal balance.
  5. Utilize competent cells with enhanced tolerance to toxic proteins.
  6. Employ an iterative culturing approach to allow cells to adapt to toxicity over time.
  7. Monitor cell density and harvest cells at the optimal density for plasmid yield.
  8. Ensure efficient cell recovery during plasmid isolation to maximize yield while minimizing DNA damage.
By implementing these strategies, you can improve plasmid DNA yield while working with toxic genes, such as when using CopyCutter EPI400. Experimentation and optimization may be necessary to find the best approach for your specific gene and experimental setup.
  • asked a question related to Plasmids
Question
3 answers
Hi everyone,
I'm in the process of creating a zebrafish Knock-in line. In order to verifying that my integration has worked, I've created a positive control plasmid with the fragment that I would expect to have in my transgenic line.
Typically, using plasmids as a positive control for PCR reactions would yield single bands due to the purity of the plasmid. My concern is that, once I optimise my PCR using the plasmid, the PCR might not actually work when using extracted gDNA from zebrafish as the template. Hence, I was wondering if it is sensible to mix the plasmid with wild type gDNA to create an unpure template. I could then use it to optimise my PCR reaction. Does this sound feasible?
Thanks :)
Relevant answer
Answer
Hi Golsana,
Although it seems like a feasible approach one of the problem is that how would you control for the amount of the plasmid that you mix with gDNA to create the unpure template. You don't know how much PCR amplification will be achieved in your zebrafish knock-in line. Therefore, everything is relative in this scenario.
One way to address this is to create a PCR standard with serial dilution of your plasmid alone and plasmid+gDNA unpure template. Once you have your knock-in line ready, you can compare the knock-in line PCR profiles with any of your standards to see if it matches to any of your PCRs. You can always scale up or down the amount of knock-in gDNA depending on what you see.
If it's a targeted knock-in, the other way to test this is to design a oligo pair which runs from the knock-in region and extends into the flanking region in the gDNA. This will be a specific PCR which will only amplify if your knock-in has worked.
Hope this helps!
Best,
Amit
  • asked a question related to Plasmids
Question
4 answers
I am currently running a plasmid isolation. I am using a bacterial stab to grow the plasmids. We have used this stab a few times now. Unfortunately, we have not had the chance to make glycerol stocks of these plasmids. I streaked both an LB agar and LB agar with ampicillin plate. I left them in the incubator overnight at 37 degrees C. I do not see any growth or colonies. I will leave it overnight again in the incubator. I doubt that this will work because I have done this once before. If you have any suggestions please let me know. Thank you!!
Relevant answer
Answer
I agree with Sofiane Benyamina as this has happened very often in my lab. What is most likely is that the plasmid has not completely closed, as even plasmids closed on themselves still create colonies.
I advise you check the process of plasmid formation (like digestion and ligation) or the ratio of lenght between insert and open plasmid. Also the lenght of the insert and its sequence can lead to a faulty ligation, since the insert bends onto itself.
  • asked a question related to Plasmids
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 Plasmids
Question
3 answers
In 84232 add gene plasmid, I am not sure at which site should I clone guide RNA
Relevant answer
Answer
You can use SnapGene to download the map of the 84232 Addgene Plasmid. Open the DNA sequence file in the Snapgene software and look for gRNA scaffold. When you select typeIIS enzymes in the Enzyme tab you will see two sites for BplI enzyme upstream of the gRNA scaffold. When you design gRNA you should add the overhangs to your gRNA oligos. Anneal the oligos and clone into the vector.
Good luck.
  • asked a question related to Plasmids
Question
1 answer
Hello!
When constructing the plasmid, I used the same promoter. However, the direction of each expression cassette promoter in the obtained plasmid is the same, and the plasmid with the opposite direction of promoter cannot be obtained. In this case, I am worried that homologous recombination within the plasmid may occur after integration into yeast, resulting in the loss of some expression cassettes. How should I do toget the plasmid with the opposite promoter direction?
Thanks in advance!
Relevant answer
Answer
To reverse the direction of two identical promoters in the construction of a plasmid, you will need to manipulate the DNA sequence of the plasmid. Here's a general guide on how you might accomplish this:
1. Identify the promoter sequences.
2. Design PCR primers to amplify the region containing the promoters in the reverse orientation.
3. Perform PCR to amplify the desired region.
4. Purify the PCR product.
5. Clone the purified PCR product into a suitable vector.
6. Verify the orientation of the promoters in the newly constructed plasmid.
7. Transform the plasmid into a suitable host organism.
8. Conduct functional assays to confirm the activity of the reversed promoters.
These steps involve manipulating the DNA sequence through PCR, cloning, and transformation techniques to achieve the desired orientation of the promoters in the plasmid.
  • asked a question related to Plasmids
Question
10 answers
Hi molecular biologists, I'm wondering if any of you might be able to help me with a question I have.
I am attempting to insert the DNA sequence coding for a protein domain into a plasmid (the plasmid is popinF). The insert DNA (E. coli optimised) was synthesised by Thermo (and it has passed their QA/QC), and I've successfully inserted it into popinF and transformed E. coli stellar cells, before collecting 3 different colonies from a plate to perform minipreps and acquire the plasmid with inserts. The sequencing results came back for all of them, and confirmed that the full (and correct!) DNA sequence had been inserted into one of the 3 plasmids.
However, I found it very peculiar that one of my plasmids appeared to have my DNA insert, but in a degenerated form with regards to the sequence. In the alignment shown attached, I can clearly see that there is very very strong matching of the sequenced result to the DNA from ~230 base onwards, showing that the synthetic DNA has inserted. But the sequence prior to this region does not show a high correlation to my DNA insert, and I'm wondering how this could be, and what could have caused this? I know that the synthesised DNA must be correct because I've successfully put the full length sequence into another identical plasmid - could it be that this particular plasmid showing a degenerate sequence could have undergone mutations within the E. coli or have degenerated in other ways, and if so could anybody please expand on the mechanisms and nature of these mutations? If anybody has any insight into mutation events of DNA inserts in plasmids within bacteria or knows of any good literature that reviews it and how to avoid them during recombination/transformation, I would be very appreciative for the help!
Thanks very much all,
Rob
Relevant answer
Answer
Plasmid insert mutations can occur due to various reasons, including errors during DNA replication, exposure to mutagenic agents, or incorrect handling during molecular biology techniques. Here are some common causes of plasmid insert mutations and ways to avoid them:
  1. Replication errors: During DNA replication, polymerase enzymes may introduce errors leading to mutations. This can happen due to misincorporation of nucleotides or slippage during replication. To minimize replication errors, use high-fidelity polymerases for PCR amplification and ensure proper primer design to reduce the likelihood of misincorporation.
  2. Exposure to mutagenic agents: Plasmid DNA can be exposed to mutagenic agents such as UV radiation, certain chemicals, or reactive oxygen species. These agents can induce DNA damage and mutations. To avoid exposure to mutagenic agents, handle plasmid DNA with care, use protective measures such as UV shields, and store DNA samples properly to prevent degradation.
  3. Errors during cloning: Mistakes made during cloning procedures, such as incorrect primer design, improper ligation, or inefficient transformation, can lead to mutations in the plasmid insert. To avoid these errors, carefully design primers, optimize cloning conditions, and use appropriate positive and negative controls during cloning experiments.
  4. Insert instability: Some plasmid inserts may contain repetitive sequences or regions prone to instability, leading to mutations such as insertions, deletions, or rearrangements. To mitigate insert instability, sequence the insert region to identify any repetitive sequences or unstable regions and avoid using them if possible. Additionally, consider using alternative cloning methods or vectors that are more suitable for stable insert maintenance.
  5. Contamination: Contamination with nucleases or other enzymes can lead to degradation of the plasmid insert, resulting in mutations. To prevent contamination, maintain sterile conditions during molecular biology procedures, use certified DNAse-free reagents, and regularly check equipment for cleanliness.
  6. Storage conditions: Improper storage conditions, such as exposure to extreme temperatures or repeated freeze-thaw cycles, can damage plasmid DNA and introduce mutations. To ensure stability, store plasmid DNA at appropriate temperatures (-20°C or -80°C), avoid frequent freeze-thaw cycles, and aliquot DNA samples to minimize exposure to light and air.
  • asked a question related to Plasmids
Question
2 answers
In addgene 84323, plasmid they have empty gRNA casette. I am not sure where to clin guide RNA of interest.
1. Is it should be between U6 promoter and gRNA casette or
2. In gRNA casette itself?
Relevant answer
Answer
I think you might have given the wrong plasmid number.
But usually it's between the U6 promoter and the gRNA scaffold.
  • asked a question related to Plasmids
Question
2 answers
I am trying to do a golden gate cloning but the ligation-digestion seems to be failing
Relevant answer
Answer
The product insert that comes with the enzyme will have a standard protocol for how much to use.
Do you mean you have exactly 96 nanogram of plasmid OR do you have a plasmid at the concentration of 96 nanograms per microliter?
  • asked a question related to Plasmids
Question
4 answers
I am cloning a PCR blunt product into pJET1.2 vector. The colony PCR and plasmid PCR have confirmed the presence of insert. The insert size is 1.17kb and the plasmid with insert looks 4kb on the gel with liberalized plasmid However, when I do digestion with HindIII and EcoRI the insert is not released. The individual digestion seems to linearize the plasmid but the double digestion does not release the insert (I tried both sequential and combined double digestion). I have not yet sequenced the plasmid.
Relevant answer
We have included EcoRI and HindIII in our primers so PCR product we ligated should carry these 2 sites Tom Masi
  • asked a question related to Plasmids
Question
3 answers
I am performing a bacterial transformation on Mycobacterium abscessus spp. abscessus using pMSP12::mCherry. I need to know if this plasmid is integrative or replicative
Relevant answer
Answer
a replicative plasmid is a plasmid which, once inside its bacterial host cell, will remain in its extra-chromosomal form; its replication is autonomous and does not depend on the replication of the chromosome.
On the other hand, an integrative plasmid is a plasmid which cannot be found in an extrachromosomal form, it must integrate the bacterial chromosome, it cannot replicate autonomously, its replication depends on the replication of the chromosome.
  • asked a question related to Plasmids
Question
1 answer
Hello, I have conducted a QuikChange site saturation mutagenesis with degenerate NNS primers to generate mutant libraries of certain residues of my enzyme. For all I have used annealing temperatures of Tm - 5. The reaction mixture is the PfuUltra II Hotstart PCR Master mix: https://www.chem-agilent.com/pdf/strata/600850.pdf
Only for half of my chosen residues could I observe plasmid DNA amplification.
Generally, even if DNA amplification was successful , after transforming competent E. coli cells with the respective plasmid, only very few colonies could be seen.
What are some problems, that can occur during the PCR cycles? I dont have any major hypotheses other that primer design might need to be reassessed.
Relevant answer
Answer
hello,@
I would first lower the annealing temperature (Tm-10°C), then if you have clones but no mutant then check your dpn1 enzyme,
if you do not have transformants then make more cycles (for site directed mutagenesis you generally don't see the PCR product (13-16 cycles)) ...
you may also think to use another PCR enzyme (we shifted succecfully for the Q5 polymerase...
  • asked a question related to Plasmids
Question
2 answers
I have been trying to make a stable cell line expressing my GOI using lentiviral transduction of HEK293T cells.
To do so I ordered a lentiviral transfer plasmid from addgene which had a gene in it already. The gene is followed by an P2A sequence and then the puromycin resistance gene.
I have used this plasmid to make a lentivirus and transduce cells. This was no problem, the cells survived puromycin and expressed the protein.
But now for my GOI, I removed the present gene from the plasmid using restricition enzymes and added my GOI. Then I packaged the lentiviruses and transduced my cells. After adding puromycin, the mock transduced cells died and the transduced once survived. But, immunofluorescence staining was negative for my protein.
What puzzles me is that the puromycin resistance gene has no promotor of its own and uses the promotor and startcodon of my GOI and is expressed, but my GOI is not translated.
I checked with PCR and the gene for my GOI is present in the cells. The sequenceshow no mutations and the gene, P2A and puromycine resistance gene are all in frame.
I also tried transfecting HEK293T cells with the exact same GOI and then staining is positive.
Does anyone have a possible explanation and solution for this problem?
Relevant answer
Answer
The 2A peptides leave a fairly significant C-terminal scar on the upstream coded protein. Many GOIs I have worked with have had compromised function because of this extra peptide tag, particularly if it is a membrane-embedded protein.
Perhaps consider switching the positions of the puromycin and the GOI (i.e. put the GOI downstream of the P2A)?
  • asked a question related to Plasmids
Question
4 answers
I am trying to select E.Coli colony which has inserted gene on pk18mob plasmid from E.Coli DH5 alpha transformation. After transformation, I did colony PCR reactions from some small colonies and it showed my inserted gene on gel electrophoresis image.
However when i cultured these colonies in LB liquid culture and extracted plasmid for restriction enzyme reaction, it did not show my inserted gene on gel electrophoresis image.
My RE protocol is 1ug of plasmid, 1ul BamHI, 1ul HindIII, 5ul buffer and water to 50ul at 37oC for 15 minutes. Then pipet 10ul to gel electrophoresis.
Relevant answer
Answer
were your primers both inside the insert sequence or one in the plasmid and one in the insert?
If both were insert primers then you may just have a positive pcr from wetting of the plate with ligation mix and insertionn may not have taken place at all
  • asked a question related to Plasmids
Question
4 answers
I obtained a plasmid from a lab down the hall. The lab has never used the plasmid before. They just happened to have it. It's labelled pMSCV-STAT6. I looked up similarly named plasmids on addgene and they're retroviral plasmids (for packaging retroviruses). However, what's confusing is the person who gave me the plasmid told me this is for packaging lentiviruses. My PI said I can't just tell a plasmid from its name. People just name plasmids randomly. So I'm really confused here. Is there a way to tell? Like if there's a particular sequence present in the plasmid that's unique to retroviral vectors or lentiviral vectors, and I can just sequence that region to determine if it's for packaging retroviruses or lentiviruses. Thanks. 
Relevant answer
Answer
Hi
Well done !!!
Thanks
  • asked a question related to Plasmids
Question
4 answers
Two plasmids of different sizes were constructed based on the pBeloBAC11 backbone. One is 18kd and the other is 40kd. Choose to use TOP10 competent cells and transform in CaCl2 solution. The 18KD can be transformed successfully, but the 40kd size does not get colonies. How to optimize My method to transform a 40kd plasmid? Does the method of power transfer help?
Relevant answer
Answer
Dear Esteemed Colleague,
Greetings. I hope this message finds you well and immersed in the intricacies of genetic engineering, particularly in the context of plasmid-based transformations, which are fundamental to a wide range of molecular biology applications. Your inquiry regarding the effect of plasmid size on transformation efficiency is both pertinent and insightful, reflecting a critical aspect of experimental design and optimization in genetic manipulation. Below, I delve into the relationship between plasmid size and transformation efficiency, grounded in current scientific understanding and empirical evidence.
Overview of Plasmid Size Impact on Transformation Efficiency
Principle: Transformation efficiency, defined as the number of successful transformants per unit of DNA used, is influenced by several factors, including the size of the plasmid being introduced into the host cell. Generally, an inverse relationship is observed between plasmid size and transformation efficiency.
Key Considerations
  1. Physical Constraints:Larger plasmids encounter more significant physical barriers during the process of entering a cell, due to their increased size and potentially more complex conformation. This can reduce the likelihood of successful uptake by the host cell, particularly in methods relying on passive diffusion or electroporation.
  2. Replication and Maintenance:The cellular machinery must replicate and maintain introduced plasmids. Larger plasmids may impose a higher metabolic burden on the host cell, potentially leading to lower stability and copy number, which can indirectly affect transformation efficiency by reducing the proportion of cells that retain and express the plasmid.
  3. Preparation and Purity:The preparation of larger plasmids can sometimes yield lower purity or concentration, which may directly impact the transformation efficiency. Ensuring high-quality, high-concentration plasmid preparations is crucial for optimizing outcomes.
Strategies to Mitigate the Impact of Plasmid Size
  1. Optimization of Transformation Conditions:Tailoring the transformation protocol to accommodate larger plasmids, such as adjusting the electric field strength in electroporation or optimizing chemical transformation conditions, can help improve efficiency.
  2. Use of High-Efficiency Host Strains:Certain bacterial strains are engineered to improve transformation efficiency, including those with enhanced uptake mechanisms or reduced nuclease activity, which can be particularly beneficial for larger plasmids.
  3. Minimization of Plasmid Size:Where possible, minimize the size of the plasmid without compromising the necessary elements for expression and selection. This may involve the removal of non-essential sequences or the use of smaller backbone vectors.
  4. Incremental Increase in Selective Pressure:Gradually increasing the selective pressure on transformants can help in maintaining larger plasmids within the host cell population, thereby potentially increasing the overall efficiency of transformation.
Conclusion
The size of a plasmid plays a significant role in determining the efficiency of transformation, with larger plasmids generally associated with reduced efficiency. By understanding and addressing the underlying mechanisms through which plasmid size impacts transformation, researchers can employ strategic approaches to optimize the transformation process for plasmids of varying sizes.
Should you require further guidance or wish to discuss additional strategies for enhancing plasmid transformation efficiency, please do not hesitate to reach out. I am here to support your research endeavors and contribute to the advancement of your projects.
Warm regards.
With this protocol list, we might find more ways to solve this problem.
  • asked a question related to Plasmids
Question
5 answers
Generally CHO (DHFR -ve) cells on transfection with plasmid bearing DHFR gene + Gene of Interest and upon addition of MTX only thoose cells which take up plasmid (containing DHFR + Gene of Interest ) will survive others will die.
My doubt 1 is : Generally DHFR is involved in De novo synthesis of Nucleotides, then how the nucleotides are synthesized in CHO (DHFR -ve ) cells?
My doubt 2 is : CHO (DHFR -ve ) cells lack DHFR so they couldn't use De novo pathway for nucleotide syntheis but they can use salvage pathway, then after transfection with Plasmid (containing DHFR + Gene of Interest) all the cells will survive due to operation of salvage pathway, now how to distinguish between the transfected cells vs Un transfected cells.
I'm confused with this DHFR-MTX selection system, could someone please help me to understand this concept, Also please share any referance material.
Relevant answer
Answer
Dear Esteemed Colleague,
Greetings. I hope this message finds you well and deeply engaged in your research endeavors, particularly those involving the use of Chinese Hamster Ovary (CHO) cells for recombinant protein production. Your inquiry regarding the application of dihydrofolate reductase (DHFR) - methotrexate (MTX) selection system in CHO cells lacking endogenous DHFR activity (DHFR-negative cells) is of significant interest for enhancing gene amplification and expression. Below, I provide a detailed overview of employing this system for the selection and amplification of transfected genes in DHFR-negative CHO cells.
Overview of DHFR-MTX Selection in DHFR-Negative CHO Cells
The DHFR-MTX selection system is a powerful tool for the selection and subsequent amplification of transfected genes in mammalian cell lines, including DHFR-negative CHO cells. This system exploits the enzyme dihydrofolate reductase (DHFR), which is crucial for the synthesis of thymidine and purine nucleotides, thereby enabling DNA synthesis and cell replication.
Key Steps and Considerations
  1. Transfection:Transfect DHFR-negative CHO cells with a plasmid containing both the gene of interest and a functional DHFR gene. This allows for the restoration of the DHFR pathway in transfected cells, enabling them to survive and proliferate in a medium lacking nucleosides.
  2. Selection with Methotrexate (MTX):After an initial selection in nucleoside-free medium, apply methotrexate (MTX), a competitive inhibitor of DHFR, to the culture medium. MTX concentrations can be gradually increased to select for cells with amplified copies of the DHFR gene. As the DHFR gene is co-amplified with the gene of interest, this process also selects for cells with higher expression levels of the target protein.
  3. Optimization of MTX Concentration:Start with a low concentration of MTX and incrementally increase it to ensure the selection of clones with high levels of DHFR expression and, consequently, high expression of the gene of interest. The optimal MTX concentration must be empirically determined and may vary depending on the cell line and the construct used.
  4. Clone Isolation and Expansion:Isolate individual clones under selective pressure and screen for those exhibiting the highest expression of the gene of interest. Expand successful clones for further characterization and production.
  5. Screening and Analysis:Perform rigorous screening of isolated clones for desired traits, such as stability of expression, growth characteristics, and productivity. Analytical methods may include Western blotting, ELISA, or activity assays specific to the protein of interest.
Additional Tips
  • Plasmid Design: Ensure the plasmid backbone contains elements that facilitate high expression in mammalian cells, such as strong promoters and enhancers.
  • Cell Culture Conditions: Maintain optimal cell culture conditions to support the growth and selection of transfected cells, paying careful attention to media formulation and culture environment.
  • Record-Keeping: Meticulously document all experimental conditions, observations, and results throughout the selection and amplification process to facilitate reproducibility and downstream analysis.
Conclusion
The DHFR-MTX selection system offers a robust strategy for the selection and gene amplification of transfected DHFR-negative CHO cells, facilitating high-level expression of recombinant proteins. By carefully designing the selection strategy and rigorously screening for high-producing clones, this system can significantly enhance the yield and stability of protein production in CHO cells.
Should you require further assistance or wish to discuss additional strategies for optimizing recombinant protein expression in mammalian cells, please do not hesitate to reach out. I am here to support your scientific journey and contribute to the success of your research projects.
Warm regards.
This protocol list might provide further insights to address this issue.
  • asked a question related to Plasmids
Question
8 answers
Hi all. I´m using calcium-phosphate transfection for 293T. I had never transfected cells with any method. I used two DNA concentrations, 5ug and 10ug per well (6-well plate). I expected lower than optimal results as it was my first time trying the procedure. I obtained about 40% efficiency with 5ug and about 10% with 10ug. My professor says that is inconsistent. I thought perhaps the DNA concentration was too high, as I seem to gather most people use 2.5ug or less, but as an undergrad I don't really know. The plasmid is TurboGFP (SHC003) and had a desirable ratio; and diluted to 5ug/ul as it was highly concentrated. 2xHBS was prepared by myself at 7.11 pH as my professor indicated; after filtering I aliquoted and froze it inmediatelly. I also prepared CaCl2 2.5M; which I also filtered and aliquoted, then kept it at 4°C.
And so I prepared:
5ug GFP: 1 ul GFP (5ug/ul), 8 ul CaCl2, 91 ul molecular grade water.
10ug GFP: 2 ul GFP (5ug/ul), 8 ul CaCl2, 90 ul molecular grade water.
I added each solution dropwise to an equal volume of 2xHBS; this was done under constant vortexing. The solution initially had a yellow-ish colour that went away quickly, and then after a couple minutes it looked white-ish.
I replaced the medium, which I had changed and hour before transfection, with 1 ml DMEM 10%SFB, 1% PenStrep.
I incubated the mixtures 10 minutes and then added it to the cells drowise and trying not to go over the same spot twice. After each drop I shook the plate gently, so the DNA could cover as much of the plate as possible. The medium turned slightly orange. I left this medium for 4 hours.
After the 4 hours I washed the cells with PBS 7 times; as I had seen very big calcium precipitates the only time I had tried it before. Then I replaced with DMEM full. I noticed while washing that some cells were lifting; even though I always take precautions not to treat them roughly.
After 24hrs I took the cells to the cytometer and got the results I already mentioned. I may have done something wrong. My professor expected at least 70% efficiency. I also added 7AAD to check for cell death/damage but I got 0%.
Any tips? Tricks? Thanks to all in advance.
Relevant answer
Answer
Dear Esteemed Colleague,
Greetings. I trust this message finds you well and deeply engaged in your critical research endeavors, particularly those involving the transfection of HEK293T cells. Achieving high transfection efficiency in HEK293T cells is pivotal for a broad range of molecular biology applications, including but not limited to, gene expression studies, viral vector production, and protein expression. Below, I provide a structured approach to optimizing transfection efficiency in HEK293T cells, incorporating best practices and strategic considerations.
Strategies for Enhancing HEK293T Transfection Efficiency
  1. Cell Health and Culture Conditions:Ensure cells are healthy, actively proliferating, and maintained under optimal culture conditions. Transfect cells at 70-80% confluency to balance accessibility to the transfection reagent-DNA complex and adequate space for growth post-transfection.
  2. Quality of DNA:Use high-quality, endotoxin-free plasmid DNA. The purity of the DNA (A260/A280 ratio of ~1.8) significantly influences transfection outcomes. Prepare DNA using reliable purification kits or methods to achieve optimal purity.
  3. Transfection Reagent Selection:Select an appropriate transfection reagent or method tailored to HEK293T cells. Lipid-based reagents (e.g., Lipofectamine 2000, Lipofectamine 3000, or PEI) are commonly used due to their efficiency in these cells. Evaluate different reagents to determine the most effective one for your specific application.
  4. Optimization of DNA and Reagent Ratio:Titrate the amount of DNA and transfection reagent to identify the optimal ratio that maximizes efficiency while minimizing toxicity. A starting point could be following manufacturer recommendations, then adjusting based on empirical results.
  5. Transfection Protocol Refinement:Follow the manufacturer’s instructions for the chosen transfection reagent closely, then tweak parameters such as incubation time with the DNA-reagent complex and the volume of the culture medium.
  6. Serum and Antibiotics:Perform transfections in serum-containing medium but without antibiotics, as antibiotics can increase cell sensitivity to transfection-induced stress.
  7. Post-Transfection Care:Change to fresh culture medium 4-6 hours after transfection to remove any residual transfection reagent and mitigate cytotoxicity, unless otherwise optimized for your specific protocol.
  8. Use of Enhancers:Consider using transfection enhancers or boosters that some reagents offer, which can further improve efficiency by facilitating the entry of the DNA-reagent complex into cells.
Additional Considerations
  • Experimental Replicates and Controls: Include appropriate controls and perform replicates to ensure reproducibility and to discern the true effects of the transgene from transfection variability.
  • Monitoring and Analysis: Utilize quantitative and qualitative methods to assess transfection efficiency, such as flow cytometry for GFP or other fluorescent markers, and qPCR or Western blot for gene of interest expression.
  • Record-Keeping: Maintain detailed records of all transfection conditions and outcomes to identify the most effective protocols and to facilitate troubleshooting in future experiments.
Conclusion
Improving transfection efficiency in HEK293T cells involves a combination of optimizing cell culture conditions, carefully selecting and using transfection reagents, and fine-tuning the transfection protocol. By systematically addressing these areas and incorporating empirical optimization, you can significantly enhance the success of your transfection experiments.
Should you require further assistance or wish to explore additional aspects of transfection methodologies, please do not hesitate to reach out. I am here to support your research efforts and contribute to the advancement of your scientific work.
Warm regards.
Take a look at this protocol list; it This list of protocols might help us better address the issue.
  • asked a question related to Plasmids
Question
4 answers
I have a plasmid with kanamycine antibiotic resistant gene and Bar as a marker gene. I need to transfer this plasmid into AGL-1 strain of agrobacterium tumefaciens. I faced a problem when I follow the protocol steps of transformation. I did not get bacterial colony even after 2-days of a LB media plate having Kan 50ug/ml.
Protocol steps
1- Take competent cells from -80 C.
2- Add 5ul of plasmid having conc. 50ng/ul in 100 ul of AGL-1 BACTERIA.
3- Keep on ice for 30 min.
4- put in liquid nitrogen for 5 min
5- keep on heat bath for 5 min.
6- keep on ice for 5-min again.
7- add 800ul of LB without antibiotic (Kan)
8- Shake for 2-hrs at 28 C.
9- spread on LB media plate with kan 50ug/ml. Keep these plates on 28 C for 2-days.
But did not get the bacterial colony.
These are the protocol steps which I followed. Anyone can guide me where I am doing mistake?
Relevant answer
Answer
Hello, remove kanamycin from nutrient media and see do you have a grow of culture or not, if you see grow it mens kanamycin resistance gene transfection or expression problem.
  • asked a question related to Plasmids
Question
3 answers
In some gene editing studies using mice, the delivered plasmid carrying the editing tool (e.g. TALEN) also carries both an HA-tag and GFP. The GFP is separated from the editing tool with a T2A sequence. Why are HA and GFP both needed?
Relevant answer
Answer
if the constuct is HAtag/Talen/T2A/GFP : the GFP is used to look in vivo at the transfected cells (%, localisation...), the HA tag is used to detect the editing tool by indirect immunofluorecsnce (IF) or western blot (the level of GFP can be different from the level of expression of the Talen du to difference in protein stability for example)
if the constuct is /Talen/T2A/GFP/HAtag , then the HA tag is not very usefull; GFP can be seen in transfected cells and antibodies against GFP can be used to see the expression but this will give you the level of expression of the GFP not of the talen protein.
  • asked a question related to Plasmids
Question
5 answers
Previously I have cloned estA gene in pET28b with restriction sites EcoRI and BamHI and stored it in -80˚C. I also proceeded with site directed mutagenesis (SDM) after confirmation of the cloning via PCR gene amplification. However, I didn't get positive result post SDM. Now, as I'm trying to repeat the experiment, I'm unable to exact the plasmid from the previously cloned bacteria and stored as glycerol stock. The bacteria shows antibiotic resistance but when I try to extract plasmid and confirm through gel electrophoresis, I'm not getting any band. Please can anyone guide me and help me troubleshoot the problem.
Relevant answer
Answer
Robert Adolf Brinzer I used Promega's PureYield Plasmid miniprep system kit for isolation of plasmid DNA. other samples are working fine only this particular cloned gene is showing problem
  • asked a question related to Plasmids
Question
2 answers
I have plants (L. japonicus) that have been transformed with an overexpression plasmid. How can I know that these plants are homozygous for the insertion?
Relevant answer
Answer
One solution is to do a restriction digest of the genomic DNA, dilute the concentration and ligate to form circularized pieces of DNA. You can then use PCR to amplify outwards from the ends of your insert and sequence the product to identify where the insertion is in the genome. You can then design primers to amplify across the insertion locus to allow for genotyping.
Alternatively make several separate lines and sequence a large number of progeny from self fertilization and only keep lines that do not produce any offspring lacking the insert. After two generations you can be confident that it is not heterozygous.
  • asked a question related to Plasmids
Question
2 answers
I noticed that a lot of plasmid annotations tend to find ColE1 origins, but I can never usually find the b. subtilis origin of replications from annotations.
Relevant answer
Answer
Else use a B. subtilis strain that expresses a filamentous phage GPII (replicase) as many plasmids also have a f1 or m13 ori.
  • asked a question related to Plasmids
Question
7 answers
I am doing plasmid isolation by Omega Plasmid DNA Minikit I #D6942-02 and Thermocycler GeneJET Plasmid Miniprep Kit #K0502. Tried both separately and no results.
I am following the instructions and using freshly prepared primary culture. All incubation periods and mixing either in the resuspension or in the lysis steps are followed carefully and I observe the viscosity of the solution after the lysis step. The white ppt. also observed after the neutralization solution. I do not know which step I might be doing wrong although my plasmid is around 6Kbp, so it does not require warming up my elution buffer. I am using sterilized distilled water as my elution. Cross checked the pipetting, chemicals, and the primary culture is really cloudy meaning that it is a rich bacterial culture. Any help?
Relevant answer
Answer
Amy Klocko Hey I have just seen this. Thank you so much!
Unfortunately, I have lost the plasmid, so I did use another bacterial strain Top10 as it is more suitable for cloning than the BL21. I have also used another plasmid (pACYC-Duet1) and transformed it into my Top10, and colonies were observed then I have made a primary culture from this freshly prepared Petri dish. Then I have tried to isolate the plasmid again from Top10 bacteria, but it did not give me any results. What do you think might be the reason?
  • asked a question related to Plasmids
Question
5 answers
I would to delete up to 33 bp from a plasmid to generate deletion mutation. Has anyone tried using Agilent Quikchange lightning site direct mutagenesis to delete such a long region? How feasible is this?
I think NEB Q5 site directed mutagenesis should work but this take me 2 weeks for delivery which I cannot wait for such a long time.
Any other methods will be appreciated.
Thank you.
Relevant answer
Answer
The Method I like is inverse PCR with 5'- phosphorylated forward and 5'-phosphorylated reverse primers facing away from each other and blunt end ligation with T4 DNA ligase and ATP and magnesium.
I the linked article, the method is Mutagenesis protocol B with deletion:
Note: the primers must be 5'-phosphorylated. Primers are not 5'-phosphorylated unless you order them from the manufacturer as phosphorylated. Otherwise, you can phosphorylated primers with T4 DNA ligase and ATP and magnesium. Then ligate the phosphorylated PCR product with T4 DNA ligase and ATP and magnesium.
  • asked a question related to Plasmids
Question
6 answers
Hi all,
I am doing a transient transfection to my cells with GFP-tagged plasmid and sending them to be FACS sorted and I need to replate them once I get them back. This is my first time ever doing this kind of experiments. would you please kindly help on how I could replate them? is it casual cell culturing? I heard that you send your plate and get a tube back?! would you be so kind to elaborate? Thank you
Relevant answer
Answer
Dear Esteemed Colleague,
Greetings. I trust this message finds you well and progressing in your endeavors within the field of cellular biology. Your inquiry about the proper methodology for replating cells after Fluorescence-Activated Cell Sorting (FACS) is both crucial and timely for ensuring the viability and proliferation of sorted cells. Below, I outline a comprehensive and structured protocol to efficiently replate cells post-FACS sorting, ensuring optimal recovery and growth.
Preparation Before Sorting
  1. Culture Conditions: Prior to sorting, ensure that your cells are in optimal condition, ideally in the exponential growth phase. This enhances their resilience and recovery post-sorting.
  2. Media Preparation: Prepare fresh culture media appropriate for your cell type. Consider supplementing the media with antibiotics and additional serum to support cell recovery if compatible with your experimental design.
Sorting Considerations
  1. Sorting Parameters: Choose gentle sorting parameters to minimize cell stress and damage. This includes using a larger nozzle size and lower pressure, if possible.
  2. Collection Tube Preparation: Pre-fill collection tubes with culture media containing serum to provide immediate nutrients and reduce shock upon collection. Keep the tubes cold to preserve cell viability.
Replating Protocol
  1. Immediate Handling: Process sorted cells as soon as possible after sorting to minimize stress and cell death. Keep cells on ice if immediate processing is not feasible.
  2. Centrifugation: Gently centrifuge the sorted cells at a low speed (approximately 200-300 x g for 5-10 minutes) to pellet the cells while minimizing stress.
  3. Resuspension: Carefully aspirate the supernatant without disturbing the cell pellet. Resuspend the cells in fresh, pre-warmed culture media tailored to your cell type's needs, gently pipetting to ensure a homogeneous cell suspension.
  4. Plating Density: Determine the appropriate plating density for your cell type and experimental requirements. Plating at a higher density can promote cell recovery and growth by facilitating cell-to-cell interactions.
  5. Culture Conditions: Transfer the cell suspension to culture vessels pre-coated if necessary with extracellular matrix components to enhance cell attachment and growth. Place the vessels in a humidified incubator set to the appropriate temperature and CO2 levels.
  6. Monitoring and Care: Monitor cell attachment and recovery closely over the next several hours to days. Change the media the following day to remove any dead cells and debris, and then regularly according to your standard culture practices.
Post-Sorting Analysis
  • Viability Assessment: Perform a viability assay (e.g., trypan blue exclusion or ATP-based luminescence assays) within 24-48 hours post-sorting to evaluate the success of the replating process and adjust culture conditions if necessary.
  • Expansion and Analysis: Once the cells have sufficiently recovered and proliferated, proceed with your experimental analyses or further expand the culture as required.
Conclusion
Following this protocol for replating cells after FACS sorting can significantly enhance cell viability and proliferation. The gentle handling of cells, coupled with optimized culture conditions, lays a solid foundation for the successful continuation of your cellular studies post-sorting.
Should you require further assistance or wish to discuss specific aspects of your cell sorting and replating procedures in greater detail, 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 Plasmids
Question
3 answers
Hi, I ran the digested plasmid on gel and purified it separately with NEB and QIAgen gel extraction kits, but the results came like this, how should I interpret these results? Where am I going wrong or are these results normal for digested plasmid?
Relevant answer
Answer
No problem! A piece of advice I learned a long time ago is your eyes do not lie to you, a spectrophotometer will.
Good Luck!
  • asked a question related to Plasmids
Question
6 answers
I have designed a genetic fusion of two separate constructs. One of them was in pET-21a and the other one was in pT7/7. Both of them expressed significantly in their respective plasmids but when I fused both of them such that one in pET-21a was tethered at 3'end followed by the other one i.e. pT7/7 and tried to express them in pET-21a, no expression was found and when I transferred my gene fusion to pET-28a, very high expression levels were monitored. I am unable to justify the reason that why is it happening as both pET-21a and pET-28a have similar sequences except the presence of his tag which only assists in purification.
Relevant answer
Answer
Hi,
pet28a comes with a ribosome binding site, while you need to add ribosome binding site yourself. May be that is the reason.
  • asked a question related to Plasmids
Question
7 answers
After transformation in DH5 alpha, I got positive colonies. I have confirmed it by PCR (using an isolated plasmid of positive colonies as a template to run PCR by Takara Taq) and restriction digestion. In PCR, I got exactly the same size of band as my desired interest in the gene but did not get fall out of my gene in restriction digestion.
I have attached a gel pic of PCR and restriction digested . 20 ul of restriction digestion was put at different amount of plasmid ( 5 ul and 8 ul of 140 (C1 )and 305 ng/ul (C2 )
Relevant answer
Answer
Thank u Ananthi Rajendran i will definitely try this.
  • asked a question related to Plasmids
Question
4 answers
Hi. For gene deletion, I need huge quantities of highly concentrated linearized plasmid for electroporation, but, after restriction digest, I have hard time to recover satisfying quantities by ethanol or isopropanol precipitation (plasmid starting material used in restriction digest as well as linearized DNA recovered have been dosed using Qubit). Does anybody have some suggestions ?
Relevant answer
Answer
add salt (NaCl or ammonium acetate (1/10volume of a 5M solution ph5) before adding 2 volumes of cold ethanol
  • asked a question related to Plasmids
Question
2 answers
I cloned 3HR and 5HR of p.falciparum on a plasmid and the next step is gRNA cloning. I did colony PCR for verification it is perfect but the thing is after taking the toothpeak of the colony in the petri dish should I also stream the leftover bacteria on the tip to a new plate of LB or not? is it necessary? cause the main colony already streamed once and for the second colony PCR do I need to streak it again? I will be thankful if you kindly share your experience.
Relevant answer
Answer
Robert Adolf Brinzer thank you so much
  • asked a question related to Plasmids
Question
10 answers
We have recently aquired plasmids with the following configuration:
EF1A>{gene of interest}:P2A:Bsd
When used for transfection (293FT cells and SH-SY5Y cells), these cells expressed the desired protein after 48 hrs (verified several times by western blot and viewing of GFP which is encoded in some of our plasmids). When the selection antibiotic is added, most cells survive, which is expected. However, after a few days, the cells no longer produce the desired protein (verified many times by western blot and viewing GFP).
To be sure, we always use a negative control for the antibiotics, cells which were not transfected, and they all died quickly (36 hrs at most).
Oddly enough, when used for lentiviral infection, there is no issue, and the cells continue expressing the protein even after a few weeks of antibiotic selection.
We have not run into this problem with other vectors acquired from other sources.
Thanks in advance
Relevant answer
Answer
That's true. We've been pondering it, it's good to hear someone from the outside recommending it.
Thanks
  • asked a question related to Plasmids
Question
1 answer
Hello. I am currently attempting to select single U 87 MG cells with red fluorescence by cell sorting after transfection with Lipofectamine 3000 of a plasmid containing mCherry. The problem is that the cells do not survive after the hole process or there are few cells left that die after a few days. Does anyone have an optimized protocol for transfection and selection of U 87MG cells by cell sorting? I would appreciate.
Relevant answer
Answer
The survival rate of cells after sorting depends mostly on three factors, the shear force created by the flow rate, the duration of the whole sorting process and the collection medium after cells been "shocked" with the given electric charge. In my years of sorting practice, I would suggest a 4 degree Celcius sorting for no more than an hour per sample, else increase the serum amount from 20-50%, just for cell recovery, increase also cell concentration to obtain target number faster, but do not exceed 1e7/mL.
Best.
  • asked a question related to Plasmids
Question
4 answers
Can I use 0.8% Agarose gel for 13.5 kb plasmid DNA?
Relevant answer
Answer
Just to be clear the issue is not the size of your plasmid but the size of the fragments you are going to look at. If you just wish to look at uncut or linearized plasmid then 0.5%-0.8% is fine. But if you are digesting with restriction enzymes then it depends on the size range of the fragments you intend to look for. 0.8% is ok down to 500bp fragments but for smaller you probably need to go to higher percents.
  • asked a question related to Plasmids
Question
8 answers
I am co-transfecting NIH3T3 cells with two plasmids (rasV12 mutant + gene of interest) for a transformation assay. My question is: how much reagent (FuGENE HD) should I use? I typically use a 3:1 reagent:DNA ratio for single transfections. But as I am adding twice the amount of DNA in total, should I use a 3:1 ratio for only one plasmid or both plasmids? Out of situations A and B below, which would you recommend? 
Situation A: rasV12 (1 ug) + Gene X (1 ug) = 3 ul FuGENE HD
Situation B: rasV12 (1 ug) + Gene X (1 ug) = 6 ul FuGENE HD
Relevant answer
Answer
When co-transfecting plasmids, the amount of transfection reagent to use can vary depending on several factors, including the transfection reagent used, the cell type, the size and concentration of plasmids, and the desired transfection efficiency. However, a common approach is to use a total amount of transfection reagent that is sufficient to effectively deliver the combined amount of DNA while minimizing cytotoxicity.
Here are some general guidelines for determining the amount of transfection reagent to use when co-transfecting plasmids:
  1. Ratio of DNA to Transfection Reagent: Follow the recommended DNA to transfection reagent ratio provided by the manufacturer of the transfection reagent. This ratio is typically optimized for maximal transfection efficiency and minimal cytotoxicity for a given cell type and transfection reagent.
  2. Total Amount of DNA: Calculate the total amount of DNA to be transfected by summing the amounts of each plasmid in the co-transfection mix. Typically, each plasmid is used at a concentration ranging from 0.1 to 1.0 µg per well of a 24-well plate, depending on the specific experimental requirements.
  3. Optimization: Perform pilot experiments to optimize the amount of transfection reagent and the ratio of plasmids to achieve the desired transfection efficiency while minimizing cytotoxic effects. Titrate the amount of transfection reagent and/or the total amount of DNA to identify the optimal conditions for co-transfection in your specific experimental system.
  4. Considerations for Individual Plasmids: Some plasmids may have different transfection efficiencies or cytotoxic effects compared to others. Adjust the amount of each plasmid in the co-transfection mix accordingly to achieve balanced expression levels if necessary.
  5. Cell Type and Culture Conditions: Different cell types may require different amounts of transfection reagent and DNA for efficient transfection. Consider the specific characteristics of your cell type, such as its sensitivity to transfection reagents and the culture conditions, when determining the optimal transfection conditions.
  6. Quality Control: Include appropriate controls, such as mock transfections or cells transfected with individual plasmids, to assess transfection efficiency, cytotoxicity, and specificity of the co-transfection.
  7. Scale-Up Considerations: If scaling up the transfection volume or using larger culture vessels, ensure that the total amount of transfection reagent and DNA is adjusted accordingly to maintain optimal transfection conditions.
By carefully titrating the amount of transfection reagent and optimizing the transfection conditions, you can achieve efficient co-transfection of plasmids while minimizing cytotoxicity and obtaining reliable experimental results.
l Check out this protocol list; it might provide additional insights for resolving the issue.
  • asked a question related to Plasmids
Question
3 answers
I have gel images of the plasmid that has been digested with various restriction enzymes and an image of the transcription of digested plasmid and a plasmid map with the cut sites located. I understand that the smallest transcription product on the gel is closest to the promoter. I get a band of  transcribed RNA approx 100bp with EcoR1 so I know that on the plasmid map the promoter is either 100bp upstream or downstream of the EcoR1 cut site but how do I then know the direction of transcription. 
Relevant answer
Answer
dentifying the position of a promoter on a plasmid and determining the direction of transcription involves several steps, including sequence analysis and understanding of promoter elements. Here's how you can approach it:
  1. Sequence Analysis:Obtain the DNA sequence of the plasmid of interest. This can be done using various methods, such as sequencing the plasmid directly or retrieving the sequence from a database or literature source. Analyze the DNA sequence using bioinformatics tools or software packages that allow you to visualize and annotate features such as promoters, coding sequences, and regulatory elements.
  2. Promoter Prediction:Look for sequences on the plasmid that resemble known promoter motifs or elements. Promoters typically contain consensus sequences recognized by RNA polymerase and transcription factors, such as the TATA box, CAAT box, and GC-rich regions. Use promoter prediction algorithms or databases to identify potential promoter regions based on sequence features and known promoter motifs.
  3. Orientation of Genetic Elements:Determine the orientation of genetic elements, such as coding sequences or selectable markers, relative to the identified promoter region. The directionality of these elements can provide clues about the direction of transcription from the promoter. In most cases, the promoter will be located upstream of the coding sequence it regulates. Therefore, the direction of transcription will typically be from the promoter towards the downstream genetic elements.
  4. Experimental Validation (optional):Conduct experimental assays to confirm the predicted promoter activity and direction of transcription. This may involve promoter reporter assays, such as measuring the expression of a reporter gene (e.g., GFP) driven by the putative promoter sequence. Use techniques such as RT-PCR or Northern blotting to analyze the direction of transcription and detect transcripts originating from the identified promoter region.
  5. Annotation and Documentation:Annotate the plasmid map or sequence file to indicate the position and direction of the identified promoter. This information will be useful for future reference and experimental design. Document your findings in a clear and organized manner, including the sequence coordinates of the promoter region, the predicted direction of transcription, and any experimental evidence supporting your conclusions.
By combining sequence analysis, promoter prediction, and experimental validation, you can identify the position of the promoter on a plasmid and determine the direction of transcription, providing valuable insights into the regulation of gene expression in the plasmid construct.
l This list of protocols might help us better address the issue.
  • asked a question related to Plasmids
Question
5 answers
Hi,
I used to use lipofectamine 3000 and it worked very well. But recently my same transfections are not working (No DNA editing, while before, the same transfection was giving me 25% editing). I don't know what is the cause. The FACS analysis seems to show expression of the GFP containing plasmids in 20 to 70% of cells.
I recently noticed that my lipofectamine 3000 reagents are expired. I used one expired since 2020 and one since April 2021. But none worked.
I also noticed my optimem is slightly expired since maybe beginning 2021.
Do you know if the lipofectamine 3000 or Optimem are reagents that cannot be used after expiring date (they are both stored in the fridge at +4)
Do you have any other idea what can be the problem? I ordered new reagents anyway, so I can compare the transfections once I receive them. But I would like some opinions if people have different ideas.
Relevant answer
Answer
Lipofectamine 3000, like many other transfection reagents, can be sensitive to its expiration date. The expiration date is determined based on the stability of the components in the reagent, and using the reagent past this date can affect its performance and efficiency in transfection experiments.
Here are some reasons why Lipofectamine 3000 might be sensitive to its expiration date:
  1. Decreased Efficiency: Over time, the reagents in Lipofectamine 3000 may degrade, leading to decreased transfection efficiency. This can result in lower levels of gene expression or knockdown in transfected cells.
  2. Increased Toxicity: Expired reagents may exhibit increased cytotoxicity, potentially causing cell death or damage to the transfected cells. This can compromise the viability of cells and affect the overall success of transfection experiments.
  3. Instability of Components: The individual components of Lipofectamine 3000, such as lipids and transfection enhancers, may degrade or become less stable over time, affecting the overall performance of the reagent.
To ensure optimal transfection efficiency and reproducibility, it's important to use Lipofectamine 3000 within its specified shelf life and expiration date. Using expired reagents can lead to unreliable results and may necessitate repeating experiments, wasting time and resources.
Always check the expiration date of Lipofectamine 3000 before use and store the reagent according to the manufacturer's instructions to maximize its stability and performance. If the reagent has expired, it's best to obtain a fresh aliquot for your transfection experiments.
l Reviewing the protocols listed here may offer further guidance in addressing this issue
  • asked a question related to Plasmids
Question
7 answers
I am looking to estimate the diameter (nm) of a variety of double stranded plasmids (pUC19, pMAL pIII, pKLAC2, etc.) when they are natively supercoiled and when they are relaxed.
If someone could point me towards a formula it would be much appreciated! Thanks. 
Relevant answer
Answer
Calculating the diameter of plasmids typically involves determining the length of the plasmid DNA molecule. Plasmids are circular, double-stranded DNA molecules, and their size is commonly expressed in terms of base pairs (bp). Each base pair corresponds to approximately 0.34 nanometers (nm) of linear distance along the DNA molecule's length.
Here's how you can calculate the diameter of a plasmid:
  1. Determine the size of the plasmid: The size of the plasmid is usually provided in terms of base pairs (bp). For example, if a plasmid is 5,000 base pairs long, its length would be 5,000 bp.
  2. Convert base pairs to linear length: Multiply the number of base pairs by the length of each base pair, which is approximately 0.34 nm. This gives you the linear length of the plasmid DNA in nanometers.Linear length (nm) = Number of base pairs × 0.34 nm/bp
  3. Calculate the diameter: Since the plasmid is circular, its diameter can be calculated using the formula for the circumference of a circle.Diameter (nm) = Linear length (nm) / π
Here's a step-by-step example: Let's say you have a plasmid with 3,000 base pairs.
  1. Determine the linear length: Linear length = 3,000 bp × 0.34 nm/bp = 1,020 nm
  2. Calculate the diameter: Diameter = 1,020 nm / π ≈ 325.05 nm
So, the estimated diameter of the plasmid is approximately 325.05 nanometers.
It's important to note that this calculation provides an approximation of the plasmid's diameter based on its linear length. In reality, the plasmid molecule is not a perfect circle, and its shape and size can be influenced by factors such as supercoiling and protein binding. Additionally, experimental techniques like electron microscopy can provide more accurate measurements of plasmid size and shape.
l Take a look at this protocol list; it could assist in understanding and solving the problem.
  • asked a question related to Plasmids
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 Plasmids
Question
7 answers
So let say I have circular plasmid named "PDR111". I did PCR with KOD ONE MIX KIT and the product is linear DNA. Then, I did self circularization of linear DNA with T4 DNA Ligase protocol (Thermo Scientific #EL0014). I did everything right as the protocol said, But my transformation keep fail. Here's how my electrophoresis looks. My Plasmid size is 11871 kb. Pls guide me
Relevant answer
Answer
Most people don't bother with checking for ligation & just proceed to transformation. Include a full set of controls, including the pUC18 plasmid that comes with most kits. That can help you determine if the issue is with the transformation protocol, the competent cells, or specific to your plasmid.
Good luck!
  • asked a question related to Plasmids
Question
1 answer
Most of the GFP-ATG8 studies use yeast-derived plasmids rather than integrating into the genome. For the ones that integrate GFP-ATG8 into the genome, they are using a commercially available URA3 marker for the URA3 locus, which is not available in BY4741. Is there any other way than making a new plasmid? Thank you!
Relevant answer
Answer
Hello Songlin Wu
GFP tagged Atg8 is stable when it is tagged at the N-terminal. You can prepare a PCR cassette with Atg8 N- and C-terminal homology following GFP sequence in frame. For the selection of the right transformation, you need a selection marker which can be either an auxotrophic marker (for yeast strain) or antibiotic marker (Kanamycin/Nourseothricin). BY4741 strain has uracil auxotrophy (ura3Δ 0). This URA3 gene will be integrated first on the 5'-UTR of the gene, but then you have to eliminate it using Cre-recombinase.
Best wishes.
  • asked a question related to Plasmids
Question
4 answers
I am doing plasmid isolation by Omega Plasmid DNA Minikit I #D6942-02 and Thermocycler GeneJET Plasmid Miniprep Kit #K0502. Tried both separately and no results.
I am following the instructions and using freshly prepared primary culture. All incubation periods and mixing either in the resuspension or in the lysis steps are followed carefully and I observe the viscosity of the solution after the lysis step. The white ppt. also observed after the neutralization solution. I do not know which step I might be doing wrong although my plasmid is around 6Kbp, so it does not require warming up my elution buffer. I am using sterilized distilled water as my elution. Cross checked the pipetting, chemicals, and the primary culture is really cloudy meaning that it is a rich bacterial culture. Any help?
Relevant answer
Answer
Ensure you use the selective antibiotic on your plasmid-containing bacterial culture. Use freshly prepared antibiotic solutions and to confirm if your cells contain plasmid plate these on antibiotic plates, and select antibiotic resistant cells and process them for plasmid isolation.
You may also try eluting it in TE buffer, but it might not really affect that much, NFW works equally good, unless you plan to store the plasmid long term at -80*C, where TE buffer might be better.
  • asked a question related to Plasmids
Question
2 answers
Hi, I' am trying to make stable cell line using plasmid transfection.
the plasmid are available for neomycin selection.
I want to know the appropriate G418 concentration and time forselection about Mlg cell line. Mlg cell line is mouse lung fibroblast cell line.
and also I wonder the concentration and time of selection about mouse primary lung fibroblast.
Thank you
Relevant answer
Answer
okay. thank you very much.
following your recommendation, I am going to try make a kill curve.
thank you.
  • asked a question related to Plasmids
Question
2 answers
Hi, I am trying to synthesise a custom DNA construct.
My end of DNA construct has a 6x His-tag attached for purification later on. However, I am not sure if I should include the stop codon before or after the 6x His-tag.
Also, I will be cloning the sythesised construct into a different plasmid and I will need to add restriction sites. Do I put the restriction sites after the stop codon or before?
Thank you in advanced.
Relevant answer
Answer
If you want the C-terminal His-tag to be part of the expressed protein, the stop codon should come at the 3' end of the gene, i.e., after the His tag.
The restriction sites should be outside the open reading frame, which includes the stop codon.
  • asked a question related to Plasmids
Question
3 answers
I have cloned one of my mutant gene constructs into pet28a and we confirmed the presence of insert on the vector backbone with colony pcr and plasmid pcr multiple times even expressed the protein but unable to get the insert released on plasmid digestion of the same. Can anyone suggest something for this?
Relevant answer
Answer
It may be that one site was destroyed in the clone you picked. Every once in a while you just get a bad clone.
  • asked a question related to Plasmids
Question
2 answers
I want to construct a plasmid (for Drosophila cell system) containing a intron in order to study splicing process. In detail, I would like to insert wether a weak or a strong splice donor site (followed by small intron+ splice acceptor) in the firefly luciferase to be able to easy monitor the splicing activity by standard luciferase assay. However, if I can find easily the consensus site for 5'SS and 3'SS, I am struggling to find the appropriate full DNA sequences that I would like to practically insert in my plasmid. Is there someone that can help me finding this sequence? (addgene reference? detailled publication with the DNAsequence fully available ?)
Thanks in advance for your help,
J
Relevant answer
Answer
It's a very good question. I'm now focusing on intron splicing. my previous data suggests that G|gt-intron-ag|N structure is adviced. However, this rule was not enough. I found despite the artificial intron locating behind the G, it has only 1/3 chance to be correctly spliced in expressing vectors. If you'd like to insert an intron into an ORF, you'd better try more positions. the splicing rules are too faraway from it could be easily used. Wish you good Luck.
  • asked a question related to Plasmids
Question
9 answers
Is there any reason for i failed getting any bands on electrophoresis after i did plasmid isolation from E. coli DH5a? My plasmid is ~17 kb in size and i already checked the transformant with colony PCR but strangely i did not get any plasmid band after i isolated them using alkali lysis method even i have confirmed the pellet presence.
I already include RNase A in my isolation protocol so i am quite sure that the pellet was not from RNA. I also have tried to upscale the culture volume to 50 ml and did the midiprep version of alkali lysis. I already reduced the elution volume to 30 ul. I even tried to use Presto Mini Plasmid Kit from Geneaid but i got no result. This is the first time i encounter such problem. If anyone could give me some suggestions i will be very glad.
p.s. currently i don't have access to nanodrop because the facility that has it currently on lockdown
Relevant answer
Answer
Bruno Ricardo Villanueva Orozco Back then we asked our neighboring lab for a new batch of competent cells as a last attempt (they're on a different building). I don't quite remember but i suppose it was caused by blackouts resulting in a mess of our original stock of competent cells.
  • asked a question related to Plasmids
Question
3 answers
Dear researchers,
Lately in our lab (Unimore, Italy) we wanted to tag a Pseudomonas strain (Biocontrol) with gfp or mCherry in order to use it for localization assay, please could you help us, from where can we get the plasmid of gfp or mCherry with the cheapest way, for free if possible :)
Best regards
Fares
Relevant answer
Answer
Omar Hammam Salloum and Tom Masi Thank you so much for your time and for the very helpful advises !
  • asked a question related to Plasmids
Question
7 answers
While I was amplifying a certain gene from chromosomal DNA for my Gibson Cloning, I noticed after sequencing that there is one extra A on the middle of my gene of interest sequence. In the original sequence there are six A (AAAAAA) but when I cloned on the plasmid, the whole plasmid sequencing showed seven A (AAAAAAA) making a frame shift mutation. I used Hight Fidelity Phusion DNA polymerase for PCR amplification. Do you recommend any other polymerase?
Thank you so much for your suggestion.
Relevant answer
Answer
High Fidelity Phusion is very high fidelity, I don't think changing polymerases will really change things. I would agree with Sofiane Benyamina that you should just isolate a few independent clones and sequence them to find one that is correct before trying to change things.
  • asked a question related to Plasmids
Question
3 answers
Hi everybody,
I have two plasmids:
- One expressing the CRY2-CIB1 optogenetic system fused to the TetA transcription factor (TetA is reconstituted upon stimulation with blue light). This one has a constitutive promoter.
- The other one expresses a reporter fluorophore under the Tet promoter.
Does someone have any advice on how to join both in a single plasmid? Is it possible to have a plasmid with two different promoters?
I am trying to avoid co-transfection of the two, because the efficiency is obviously lower than by having a single construct.
Thanks to anybody that can help!
Relevant answer
Answer
There does not need to be any order since each promoter generates its own transcript and are therefore transcriptionally independent. They don't even need to be oriented the same way.
  • asked a question related to Plasmids
Question
3 answers
Thanks
Relevant answer
Answer
If anyone can guide me on how to design the primer and antibiotic gene cassette would be of great help because Im so new to this work.
  • asked a question related to Plasmids
Question
2 answers
1. My strain doesn't have a recA knock out.
2. I use a 2 plasmid system for my work. One replicates through rolling circle and the other through theta replication.
2. I have toehold switch-trigger pair on the plasmids which make strong secondary structures and are probably prone to recombination.
3. I get negligible titres for my product of interest.
Is it possible to prevent this problem through simple bioprocess methods (without cloning anything extra)?
Relevant answer
Answer
Before you worry too much about recombination, you should investigate why you are not getting product. If the plasmids are recombining in a way to prevent production of your product then you can sequence them again and see how they are altered. Or it could be that the problem is something else entirely and recombination is not actually a concern for you.
Lastly, can you not just switch strains?
  • asked a question related to Plasmids
Question
1 answer
Electrophysiology of 293 cells transfected with GABA plasmid could not detect current
Relevant answer
Answer
I'm not sure what the question here is. How did you attempt to detect current?
  • asked a question related to Plasmids
Question
1 answer
I am looking for exact copy number of pET28a plasmid. A citation would be great. Literature search only shows that it is low copy vector, but I haven't found any papers that mention the exact copy number or even an estimate.
Relevant answer
Answer
I believe that pET28 is based on the pBR322 backbone and that plasmid has a copy number of around 20 per cell. The precise number will vary by strain and growth conditions though.
  • asked a question related to Plasmids
Question
3 answers
Hello!
Recently, I ordered an FOLH1 ORF, which came inserted into the pUC-57 mini vector. Additionally, I requested BamHI and XbaI restriction sites to be included at each end of the ORF. When I performed the digestion of the plasmid, I observed that the single digestions with BamHI and XbaI linearized the plasmid as expected. However, when I performed the double digestion, I noticed two bands corresponding to the pUC vector and the FOLH1 ORF, as anticipated. Surprisingly, there was an additional band below them that I did not expect. What does this additional band signify? Could it be contamination, or is there a problem with the digestion process?
Relevant answer
Answer
some enzyme can have "star" activity where they cut to other stites than the regular one.
It can happen in specific conditions (for example to much glycerol in the reaction mix) or if the digesion is too long.
What king of enzyme are you using ? High fidelity from NEB ? and the buffer ?
  • asked a question related to Plasmids
Question
2 answers
I recently encountered an intriguing situation while examining a plasmid constructed by someone else for a eukaryotic expression system. This plasmid contains a unique arrangement of open reading frames (ORFs) that has sparked several questions regarding the potential outcomes of their translation.
In this plasmid, there is an ORF near the 5' end, where the translation initiation site is quickly followed by a stop codon, potentially resulting in a very short peptide. More interestingly, nested within this first ORF is a second ORF that begins inside the first ORF and could potentially translate into a much longer protein, consisting of 500 amino acids.
Given the common understanding that eukaryotic transcripts typically feature a single ORF, the discovery of this arrangement has led me to ponder the following questions about the translational dynamics in this specific scenario:
  1. In the context of this plasmid, will the translation machinery be capable of bypassing the short ORF to translate the longer protein, or will it prioritize the translation of the short peptide due to its proximity to the 5' end?
  2. If both peptides are indeed translated, what might be the expected ratio between the production of the long and short peptides?
  3. Is there a possibility that only the short peptide will be translated, effectively ignoring the translation potential of the longer, nested ORF?
Furthermore, I'm curious about how this scenario might differ if the plasmid were used in a prokaryotic system, which is known for its ability to translate multiple ORFs within a single transcript.
I'm seeking insights, experiences, or any relevant literature that could help shed light on the translational strategies employed by cells when faced with plasmids containing nested ORFs, especially in the context of eukaryotic expression systems.
Thank you in advance for sharing your knowledge and experiences.
Relevant answer
Answer
There are a myriad examples of this sort of ORF structure in eukaryotic viral systems (particularly RNA ones), where it has evolved to diversify/expand coding capacity and/or regulate viral gene expression. At first pass you can predict relative expression levels by examining the Kozak consensus around each AUG codon. We usually consider the -3 and +4 positions to categorise them as strong, middling and weak. If the first AUG is strong, you typically won't get much of the second product. If the first AUG is medium or weak and the second is strong, you'll get lots of the second product. Confounders to look out for; 5'-UTR length and protein stability. AUGs very close to the 5'-cap are often not translated well. Shorter proteins are harder to detect and if they're too short (or the wrong sequence) to fold up into a coherent structure, they'll be turned over rapidly and can be hard to detect unless (or even if) the proteasome is turned off.
After that, it can get properly complicated with all sorts of ribosomal gymnastics depending on the exact sequence of the transcript.
Bugs - need to consider Shine-Dalgarno (sp?) sequences or lack of, I think. But my undergrad lectures on this were many years ago :)
Cheers
Paul
  • asked a question related to Plasmids
Question
1 answer
Our lab is trying to use in vitro transcription to create mRNA of our inserted on a pcDNA 3.4 TOPO plasmid. I noticed it does not have a T7 promotor sequence. Are there other available promotors on the market we could use for in vitro transcription?
Relevant answer
Answer
The other IVT polymerase commonly used for molecular biology applications is SP6. I had a look at your plasmid backbone though, and I'm pretty sure it won't have the SP6 promoter either. I think you will need to clone your cDNA into an IVT vector for this experiment to work.
  • asked a question related to Plasmids
Question
5 answers
I am a graduate student doing sub cloning of two similar genes into pBC SK(+); one of the gene was inserted but few colonies appeared on the agar after transformation, the other is not inserting into the plasmid no matter how I elevated the ration of insert to plasmid.
What is the explanation?
Relevant answer
Answer
Hello, ethanol can have an effect, as well as other elements that have already been mentioned above. If you are using some carriers in your fusion such as glycogen it can also interfere with the binding.
If you have already tried everything else (as you told us) perhaps you can try cloning directly into plasmids prepared for direct cloning such as Topo.
  • asked a question related to Plasmids
Question
9 answers
I am planning to work with Addgene's pLKO-tet-on plasmid. I checked the protocol on the website and comments left here so far. I am a bit confused. Should I design the oligos exactly in the protocol (5'-CCGG for AgeI and 5'-AATT for EcoRI)? Because it is one bp missing in both RE sites and it results in oligos which have mutant RE sites. If they are mutant, how will the ligation work? I would be really happy if someone could explain me. One more thing, I came across so many people who had troubles of getting positive clones after ligation. I am wondering what is the latest situation. Is there any tricks that I should know? Thank you very much for your answers in advance.
Relevant answer
Answer
Designing shRNA (short hairpin RNA) oligonucleotides for the pLKO-tet-on plasmid involves several steps to ensure specificity, efficiency, and compatibility with the vector system. The pLKO-tet-on system allows for doxycycline-inducible expression of shRNA, offering controlled gene silencing. Here's a general guide on how to design shRNA oligos for this system:
1. Target Gene Selection
  • Choose the gene you wish to silence. It's crucial to select a target sequence that is unique to your gene of interest to avoid off-target effects.
2. shRNA Sequence Design
  • Target Sequence Length: Typically, 19-21 nucleotides long sequences within the target mRNA are chosen for shRNA design.
  • GC Content: Aim for a GC content of 40-60% to ensure stable shRNA formation and efficient target binding.
  • Blast Search: Use the NCBI BLAST tool to check the specificity of your chosen sequence against the genome of your organism to avoid off-target silencing.
  • Avoid Polypurine Stretches: Sequences with long stretches of Gs or Cs can form strong secondary structures or promote Pol III termination, reducing shRNA efficiency.
3. Loop Sequence
  • A loop sequence is required to connect the sense and antisense strands of the shRNA. A commonly used loop sequence is TTCAAGAGA, but others can also be effective.
4. Termination Signal
  • A string of 5-6 thymidines (T's) acts as a Pol III termination signal and should be added at the end of the antisense strand.
5. Incorporating Overhangs for Cloning
  • The pLKO-tet-on vector uses specific restriction sites for cloning (commonly AgeI and EcoRI). You'll need to add 5' overhangs compatible with these sites to your oligos. For example:Forward oligo: 5'-CCGG(N19 sense sequence)TTCAAGAGA(N19 antisense sequence)TTTTTG-3' Reverse oligo: AATTCAAAAA(N19 reverse complement of antisense sequence)TCTCTTGAA(N19 reverse complement of sense sequence)-3'
  • The CCGG and AATT sequences at the ends of the oligos correspond to the overhangs created by AgeI and EcoRI digestion, respectively.
6. Oligo Synthesis and Cloning
  • Have your designed oligos synthesized by a reputable company. Upon receiving, anneal the oligos to form double-stranded DNA and ligate into the digested pLKO-tet-on vector.
7. Validation
  • After cloning, sequence verify the insert to ensure correct shRNA sequence integration. It's also important to test the efficiency of gene knockdown by your shRNA in a pilot experiment before proceeding with more extensive studies.
8. Inducibility Test
  • Verify the inducibility of your shRNA expression by treating transfected cells with and without doxycycline and measuring the target gene's mRNA and protein levels.
Designing effective shRNA oligos requires careful consideration of the target sequence, shRNA structure, and cloning strategy. The pLKO-tet-on system's inducible nature adds an extra layer of control to your gene silencing experiments, making it a powerful tool for studying gene function.
l Perhaps this protocol list can give us more information to help solve the problem.
  • asked a question related to Plasmids
Question
4 answers
I am working with plasmids containing aion channel, with the goal of eventually using them for transfection in Hek cells.  The problem I am having is preparing these plasmids at the bacterial transformation stage.  I have 2 different channels (both from he HCN family), on of them (HCN2) grew perfectly on the first try in XL1 Blue cells.  However I am now doing point mutations on the channel (using a Quikchange kit) and I cannot get a colony that has my intact channel.  Additionally I am trying to use HCN1, another member of the same family, and it is giving me similar problems to the mutation reaction.  Here is what I have tried so far:
1.  I am using internal channel specific primers to screen picked colonies for the presence of my plasmid.  PCR of the unmated HCN2 plasmid produce a clean band of the appropriate size.  PCR of the mutation reaction prior to transformation produces a single band of the right size. but PCR's of the picked colonies for the mutants do not, they show multiple bands.
2. Used Stbl2 competent cell to hopefully prevent recombination of the plasmid,but the pct's looked the same as the XL1-Blue.  
3. Tried incubation at 37 and 30 degrees, and decreasing the antibiotic concentration, but still the same problem
I have tried these things with both the HCN2 mutation reaction and the wild type HCN1 plasmid and have had no luck.
Any advice would be much appreciated!  Also, if there are any extra details that would help please let me know 
Thanks! 
Anna
Relevant answer
Answer
Plasmid recombination following transformation can be a significant issue, especially when working with constructs that have repetitive sequences, large plasmids, or multiple plasmids being transformed into the same cell. Several factors can contribute to plasmid recombination:
1. Presence of Repetitive Sequences
  • Plasmids containing repetitive sequences are prone to recombination. During bacterial replication or repair processes, these sequences can misalign, leading to recombination events.
2. Plasmid Size and Complexity
  • Large plasmids or those with complex arrangements of inserts can be more susceptible to recombination. The physical stress during the transformation and replication process can lead to breakage and erroneous repair, facilitating recombination.
3. Host Strain Recombination Activity
  • The choice of bacterial strain for transformation can significantly affect recombination rates. Some strains have higher recombination activities due to their innate DNA repair and recombination mechanisms. Using recombination-deficient strains (e.g., recA mutants) can reduce this issue.
4. Transformation Method
  • Certain transformation methods may inadvertently promote recombination. Electroporation, for example, can create transient breaks in DNA, which under certain conditions might lead to increased recombination.
5. Multiple Plasmids in One Cell
  • Transforming multiple plasmids into the same cell increases the likelihood of recombination between them, especially if there are homologous sequences present.
Strategies to Minimize Recombination:
  • Use recombination-deficient strains: Strains like DH5α, STBL3, or those specifically engineered to reduce recombination (e.g., recA mutants) can help.
  • Minimize repetitive sequences: When designing plasmids, avoid or minimize the inclusion of repetitive sequences that can promote recombination.
  • Select appropriate cloning sites: Use unique restriction sites and cloning strategies to minimize recombination hotspots.
  • Optimize transformation conditions: Gentle handling and optimization of the transformation process can reduce stress-induced recombination.
  • Single-plasmid transformations: If possible, avoid co-transforming multiple plasmids into the same host to reduce recombination events between them.
  • Screen for recombination: After transformation, screen colonies carefully using PCR, restriction digestion analysis, or sequencing to identify and exclude recombinant plasmids.
Addressing these factors and implementing strategies to minimize their impact can significantly reduce the occurrence of unwanted recombination events during plasmid transformation.
l Take a look at this protocol list; it could assist in understanding and solving the problem.
  • asked a question related to Plasmids
Question
5 answers
I need bacterial DNA without endotoxin for cell stimulation. All the kits Ive found are for Plasmid DNA. Im dealing with Genomic DNA. I extract DNA from cultivated bacteria conventionally (phenol/chloroform/ethanol methods) and own a large amount of DNA (500-1000 ug/ml in 3-5 TE buffer approx) but they still contain endotoxin. I'm thinking about conventional method for endotoxin removal due to a very large DNA amount. Anyone can suggest me the conventional protocol or kits to remove LPS from Genomic DNA. pls help. Thanks in adance.
Relevant answer
Answer
Removing lipopolysaccharides (LPS), also known as endotoxins, from genomic DNA is crucial for experiments that require pure DNA, such as those involving sensitive downstream applications like transfection into endotoxin-sensitive cell lines. Here are some methods and tips for effectively removing LPS from genomic DNA preparations:
1. Endotoxin Removal Kits:
  • Commercial Kits: Numerous commercial kits are specifically designed for endotoxin removal from DNA, proteins, and other biological samples. These kits usually employ affinity chromatography or adsorption techniques to selectively bind and remove endotoxins while allowing the DNA to pass through.
  • Benefits: These kits are optimized for high efficiency, ease of use, and can often achieve endotoxin levels low enough for even the most sensitive applications.
2. Phenol-Chloroform Extraction:
  • Procedure: Phenol-chloroform extraction can help reduce endotoxin levels in DNA samples. After the aqueous phase is separated, the DNA in the aqueous layer can be further purified to remove residual endotoxins.
  • Consideration: This method may not remove all endotoxins and is less specific than kits designed for endotoxin removal.
3. Anion-Exchange Chromatography:
  • Mechanism: Anion-exchange chromatography is effective in separating endotoxins from DNA because of the significant difference in their charges. DNA binds to the column under certain conditions, while endotoxins do not.
  • Efficiency: This method can be highly efficient but requires optimization based on the DNA sample and the specific anion-exchange resin used.
4. Ultrafiltration:
  • Approach: Using ultrafiltration membranes with specific molecular weight cutoffs can help in reducing endotoxin levels. This method relies on the size exclusion principle.
  • Application: Suitable for larger volumes and can be effective if the DNA size significantly differs from that of LPS aggregates.
5. Polymyxin B Columns:
  • Principle: Polymyxin B binds to LPS due to its affinity for lipid A, a component of LPS. Columns containing immobilized polymyxin B can thus selectively remove endotoxins from DNA samples.
  • Usage: These are available as spin columns or beads and can be very effective, though they may have a binding capacity limit.
6. Dialysis:
  • Method: Dialysis against a suitable buffer can sometimes reduce endotoxin levels, especially if combined with other purification steps. However, this method alone is generally not sufficient for complete removal.
  • Application: Best used as an additional purification step rather than a standalone solution.
Tips for Effective Endotoxin Removal:
  • Assess Your Needs: Determine the sensitivity of your downstream applications to endotoxins to choose the most appropriate method.
  • Validate Removal: Use a Limulus Amebocyte Lysate (LAL) assay or similar tests to quantify endotoxin levels post-purification to ensure they are below your required threshold.
  • Optimize Protocol: Depending on the source and concentration of your DNA, some optimization may be required to achieve the best balance between endotoxin removal and DNA yield/purity.
Choosing the right method depends on your specific requirements, including the sensitivity of your downstream applications, the volume of DNA sample, and the acceptable level of endotoxin contamination. It's often beneficial to start with a method known for high efficiency and specificity, such as a dedicated endotoxin removal kit, especially for critical applications.
l Take a look at this protocol list; it could assist in understanding and solving the problem.
  • asked a question related to Plasmids
Question
1 answer
I would like to know what is the function of ara gene mutation in DH5 alpha E. coli.
The mutation chart says only this mutation will block arabinose metabolism.
But what will happen with the transformed plasmids if arabinose metabolism is blocked?
Relevant answer
Answer
Nothing will happen and everything will be fine.
The arabinose operon carries genes to permit growth on the sugar arabinose, E. coli carries many catabolic operons to permit growth on various sugars. So unless you intend to grow your strain on minimal media with arabinose as the sole carbon source (and I doubt you have any reason to do so), then you can ignore the ara mutation. It was just in the background of the strain but is not present for any particularly useful reason.
  • asked a question related to Plasmids
Question
2 answers
When we transfer plasmid in fungi for developing a transgenic fungi then how we can know that the fungi which grow on the media plates are transgenic or not?
Can we screen out fungi by adding antibiotics like Ampicine or kanamycine which is present in the plasmid?
Relevant answer
Answer
I agree with Amy Klocko that it will depend whether there is an appropriate promoter that will be expressed in fungi to express a resistance gene. Ampicillin is not going to work against fungi, however if expressed the kanamycin resistance gene will confer resistance to G418 which is a kanamycin analogue that works against eukaryotic cells.
However the best verification is to to PCR to verify that the gene of interest on your plasmid is actually present in your fungal strain after transformation.
  • asked a question related to Plasmids
Question
2 answers
I have tried to read about this but I don't find definitive answers.
I am trying to lentivirally transduce a mouse cell line with a few different plasmids (each encoding a different antibiotic resistance gene, namely puromycin, hygromycin and G418). After successfully transducing the cells a first time (with the puromycin resistance-containing plasmid), it now looks like when I transduce the cells with the hygromycin resistance-containing plasmid they don't die at the hygromycin concentration established during the antibiotic titration.
Has anyone experienced this?
I found this paper, but it doesn't seem to answer my question fully: https://www.sciencedirect.com/science/article/abs/pii/S0003269709008434
Relevant answer
Answer
Hi Carles Puig,
I haven't faced a problem like that. However I can share you some considerations:
1) Have you considered non-simultaneos transduction of your plasmids? For instance, first you transduce a plasmid, then you perform a selection of 2-3 days, then another plasmid with its subsequent selection and so on...
2) In our lab we had a couple of problems with G418, it was because of the analogy with neomycine and hygromicine. Maybe you can check thoroughly if the gene resistance can show cross-reaction, and if so, change one of the antibiotics
What type of cells are you transducing/transfecting?
I hope it helps
Best regards,
Francesc
  • asked a question related to Plasmids
Question
3 answers
hi, we are working on the puc57 plasmid that our construct designed with multiple RE sites and synthesized and cloned in it. we use fast digest re enzymes (thermofisher) to separate the parts of our construct for sub-cloning.
we have some issues with these enzymes that didn't work well on the electrophoresis with TAE buffer and the expected parts of the gene after restriction, are invisible, we did simple digest and double digest with one and two enzymes. what do you think is the problem?
thank you
Relevant answer
Answer
Performing an overnight digest may also help.
  • asked a question related to Plasmids
Question
3 answers
Hi. I am tring to express recombinant protein. I obtained the nucleotide sequence of the protein I want to express through cDNA cloning and obtained the ORF sequence of the protein inserted into the plasmid. I designed primers to introduce restriction enzyme sites and confirmed the desired sequence (primer sequence with restriction enzyme sites and the protein's ORF) through PCR and sequencing. The restriction enzymes used are Nde1 and BamH1.
I treated the PCR product and pET28a (a vector for recombinant protein expression) with restriction enzymes. The reaction conditions, including buffer and temperature, were determined according to the manufacturer's protocol, with a reaction time of two hours (manufacturer's recomandation is 1 hour). BamH1 was processed first, followed by PCR purification of the vector and insert. Similarly, Nde1 was processed, followed by agarose gel purification. The purified DNAs were ligated using Takara Mighty Mix and transformed into E.coli BL21 strain. The TF strain was spread on Kanamycin LB plates. The Kanamycin concetration is 50ug/ml. Although there were not many colonies, I obtained a few colonies after about two days. Colony PCR was performed on the obtained colonies, and bands of the desired size (the same size as the PCR for introducing restriction enzyme sites) were confirmed.
Therefore, we attempted to recover the plasmid from BL21 and hoped to confirm it again through sequencing before expressing the protein. However, there is a problem. Surprisingly, plasmid extraction from BL21 does not succeed. Typically, when we extract cloning plasmids in our laboratory, we obtain around 200-600 ng/ml. However, in this case, it is below 50 ng/ml. Despite ignoring the recommended concentration of 100 ng/ml by the sequencing company, we proceeded with sequencing, but no results were obtained. We have tried the described process several times, but we consistently encounter the same issue. Plasmid extraction seems impossible. By the way, the Nde1 site of pET28a exists in the T7 tag region. I am aware that this is necessary for purification. However, since I am going to use a 6xHis tag, I intended to remove it. I am suffering greatly due to these results. Thank you very much for taking the time to read through the lengthy text. I truly appreciate it. Is there something I have overlooked in this process? I seek your professional advice and will strive to follow it as much as possible.
Relevant answer
Answer
Because BL21 expresses the EndA nuclease, sometimes it can be difficult to get good plasmid DNA unless you take special precautions, it depends upon your procedure. However you might just take some of the DNA and retransform it into a normal strain (DH5alpha or similar) and prepare plasmid from there to sequence and confirm.
  • asked a question related to Plasmids
Question
11 answers
Hello, 
I am using a c-myc-tagged plasmid for coIP experiments, and i always get two specific bands on my western blot when I immunoblot with the c-Myc antibody (ab32). I dont understand what can be the second band i see on my western blot. Can anyone Help plz 
Thanks 
Pam
Relevant answer
Answer
When you observe two specific bands in a Western blot using an antibody against c-Myc, it could indicate several biological or technical factors. Understanding the context of your experiment, including the cell type, the nature of the c-Myc construct (if any), and experimental conditions, can help interpret these results. Here are some common reasons for observing two bands:
1. Post-Translational Modifications (PTMs)
c-Myc is known to undergo various post-translational modifications, including phosphorylation, acetylation, and ubiquitination. These modifications can alter the migration pattern of proteins on SDS-PAGE, potentially resulting in multiple bands. Different forms of modified c-Myc might have slightly different molecular weights, leading to the appearance of two distinct bands.
2. Alternative Splicing or Isoforms
The c-Myc gene might produce different isoforms through alternative splicing, leading to proteins of slightly different sizes. If your c-Myc antibody is capable of detecting more than one isoform, this could explain the presence of two bands.
3. Cleavage Products
Proteolytic cleavage of proteins can result in fragments of various sizes. If c-Myc is being cleaved in your cells, the antibody might detect both the full-length protein and a stable cleavage product.
4. Expression of Multiple Myc Family Proteins
If the antibody has cross-reactivity with other members of the Myc family, such as N-Myc or L-Myc, and these proteins are expressed in your cells, you might observe multiple bands. Check the antibody specificity to see if cross-reactivity is a known issue.
5. Technical Considerations
  • Loading Control: Ensure that a loading control is used to rule out unequal loading or transfer as a cause of varying band intensities.
  • Antibody Specificity: Verify the specificity of the antibody through controls, such as using cells with known c-Myc levels or overexpression/knockdown models.
  • Experimental Conditions: Changes in cell conditions, treatments, or stress could influence c-Myc expression or modification levels, potentially leading to variations in detected bands.
Troubleshooting Steps
  • Control Experiments: Use cells with known c-Myc status (overexpression, knockout, etc.) as controls.
  • Antibody Validation: Check if the antibody has been validated for Western blot and for detecting endogenous levels of c-Myc. Look for validation data or publications that used the same antibody.
  • Optimize Western Blot Conditions: Optimizing gel concentration, transfer conditions, and antibody dilutions can help clarify the nature of the observed bands.
  • Mass Spectrometry: For a definitive identification of the bands, consider cutting them out from a gel and analyzing them by mass spectrometry. This can confirm the identity of the proteins and any post-translational modifications.
In summary, multiple bands detected with a c-Myc antibody in Western blot could have various biological or technical explanations. Careful experiment design and additional controls can help determine the reason behind the observed bands.
l With this protocol list, we might find more ways to solve this problem.
  • asked a question related to Plasmids
Question
2 answers
Hi everyone,
I was looking for some tools (preferably web-based tools) to predict genes organized in operons in various bacterial sequences I currently work on (plasmids, chromosoms, etc.).
For example, I want to upload a sequence (gb, fasta, etc.) with either a single gene of interest, or a full genome sequence (annotated or not) and as an outcome I would like to receive information regarding operons.
Despite many examples of such tools in literature , it seems they are not operational.
Can you recommend a working web-based application for operon prediction in uploaded genome sequences?
Thanks in advance.
Relevant answer
Answer
I think the Operon-mapper will do what you are asking about. The link to the web portal is https://biocomputo.ibt.unam.mx/operon_mapper/out/out_3283038.html
  • asked a question related to Plasmids
Question
3 answers
I am currently doing my PhD project which consists of a lot of cloning of new plasmids I am assembling. Our laboratory generally maintains the collection on JM109 strain. But since I am doing a lot of Gibson Assemblies, I have been using electrocompetent DH10B cells for higher efficiency. My question is, can I use standard protocol of preparation of electrocompetent E. coli on JM109 instead of DH10B?
Relevant answer
Answer
Yes, you can adapt the protocol for preparing electrocompetent E. coli cells from DH10B to JM109. However, it's important to note that different strains of E. coli may have slightly different requirements for optimal transformation efficiency, so you may need to optimize the protocol for JM109 cells.
Here's a general outline of how you can adapt the protocol for preparing electrocompetent JM109 cells:
  1. Start with a fresh overnight culture of JM109 cells grown in LB medium at 37°C with shaking.
  2. Inoculate 50-100 mL of LB medium with the overnight culture and grow at 37°C with shaking until the culture reaches an OD600 of around 0.4-0.6. This typically takes 2-3 hours.
  3. Chill the culture on ice for 15-30 minutes to stop growth.
  4. Pellet the cells by centrifugation at 4°C for 10 minutes at 4000 rpm.
  5. Remove the supernatant carefully and resuspend the cell pellet gently in an ice-cold solution of 10% glycerol using a small volume (typically 10% of the original culture volume) to concentrate the cells.
  6. Centrifuge the resuspended cells again at 4°C for 10 minutes at 4000 rpm.
  7. Repeat the wash step with ice-cold 10% glycerol one or two more times to ensure the removal of any remaining LB medium.
  8. After the final wash, resuspend the cells in a small volume of ice-cold 10% glycerol to achieve a concentrated cell suspension.
  9. Aliquot the electrocompetent cells into small volumes suitable for single-use transformations (typically 50-100 µl).
  10. Flash freeze the aliquots in liquid nitrogen and store them at -80°C for long-term use.
  11. To use the electrocompetent JM109 cells, thaw an aliquot on ice, add your DNA (e.g., plasmid DNA for transformation) to the cells, perform the electroporation, and recover the transformed cells in SOC medium before plating onto selective agar plates.
By following this adapted protocol, you should be able to prepare electrocompetent JM109 cells for your Gibson Assembly experiments. It's always a good idea to perform optimization experiments to determine the optimal conditions for your specific application.
  • asked a question related to Plasmids
Question
4 answers
Hello everyone,
My question is regarding the behaviour of a (same) plasmid in two different forms: circular versus linear.
If two identical plasmid, whereas one is in circular form (undigested) and the other in linear form (once digested), is being run in a gel, which one is being expected to migrate faster (travel a longer distance) through the gel?
Thank you in advance.
Relevant answer
Answer
Michael J. Benedik Thank you very much for the clearance.