Science method

Transfection - Science method

The introduction of DNA into a recipient eukaryote cell and its subsequent integration into the recipient cells chromosomal DNA.
Questions related to Transfection
  • asked a question related to Transfection
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 Transfection
Question
5 answers
Hi,
This is actually just a general question out of curiosity. I have tried transfection using PEI both in suspension and attached cells, with and without antibiotic before and I don't see any difference. I understand that in lipid-based transfection, antibiotic can hinder the complex formation of lipid and DNA, but because PEI works differently I don't see why it is still advisable to use antibiotic-free media?
In our lab but we even don't change media in culture vessel for lipofectamine transfection and it still work perfectly, as long as we perform the DNA-lipofectamine complex formation in OPTIMEM first. It is also easier because we don't need to wash or change media prior to transfection.
Any other opinion?
Relevant answer
Answer
Hi Lia,
I hope all is well. Since this is an old post, I am not sure if you still work on the Expi cells and PEI transfection. I am currently doing a large-scale Expi cell suspension culture and will use PEI MAX for the transfection. I have my seed culture growing with 1xPen-Strep (1x defined as 100 units/mL), and am considering diluting the culture into antibiotic-free medium for spliting purposes, which will reduce the Pen-Strep concentration in the culture. Do you think 0.02x Pen-strep concentration in the final culture will interfere with the transfection?
  • asked a question related to Transfection
Question
3 answers
I have transfected neurons using electroporation before playing plasmid with Cas9 which makes a single double stranded cut. But I am not able to see any results. Is there a way I can delete the whole gene using CRISPR in primary neuron culture?
Relevant answer
Answer
Thanks for the advice. The knock out mouse is not available but I will try the lentivirus .
Tripti
  • asked a question related to Transfection
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 Transfection
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 Transfection
Question
5 answers
HI
i transfected adenovector to hek293 cell line. i don't know Adenovirus CPE.
this picture is hek 293 cell line 8 days post transfection. do you observe any CPE?
Relevant answer
Answer
Certainly, I can guide you on how to evaluate and analyze the cytopathic effect (CPE) caused by adenoviruses in cell cultures, which is crucial for identifying virus infection and determining its intensity. Adenovirus CPE involves characteristic changes in host cells induced by viral invasion, which can be used effectively to assess the progress and severity of infection. Here’s a detailed approach to study adenovirus CPE:
  1. Cell Culture Preparation:Cell Line Selection: Choose a suitable cell line that is susceptible to adenovirus infection. Human cell lines like A549 (lung carcinoma) or HEK293 (human embryonic kidney cells) are commonly used for adenovirus studies due to their high susceptibility. Culture Conditions: Maintain cells in an appropriate growth medium at 37°C with 5% CO2, ensuring they reach 70-80% confluency by the time of infection, optimal for observing CPE.
  2. Virus Inoculation:Virus Preparation: Use a known titer of adenovirus to infect the cell cultures. The amount of virus used can vary depending on the desired multiplicity of infection (MOI). Infection Process: Remove the growth medium and add the virus in a small volume of serum-free medium to allow close contact with the cells. After allowing virus adsorption for 1-2 hours, add growth medium back to the cells.
  3. Monitoring CPE:Daily Observation: Examine the cells daily using a light microscope to observe the development of CPE. Adenovirus typically induces rounding, clumping, and detachment of cells. Keep detailed records of the progression and intensity of these effects. Photographic Documentation: Capture images of the CPE at various time points post-infection to document changes and compare with uninfected control cells.
  4. Quantitative Assessment:CPE Scoring: Develop a scoring system for CPE based on severity and extent (e.g., 0 = no CPE, 1 = mild CPE, 2 = moderate CPE, 3 = severe CPE). Viability Assays: Use assays such as MTT or trypan blue exclusion to quantify cell viability and correlate with visual CPE observations.
  5. Viral Titer Estimation:Plaque Assay or TCID50: Following the observation of CPE, perform a plaque assay or TCID50 to quantify the viral titer. This helps in correlating the extent of CPE with the viral load.
  6. Data Analysis:Data Compilation: Compile observational and quantitative data to analyze the relationship between the extent of CPE, cell viability, and viral titer. Statistical Analysis: Apply appropriate statistical methods to validate the observations and assess the reproducibility of the results.
  7. Reporting and Review:Documentation: Prepare a comprehensive report detailing the methodology, observations, data analysis, and conclusions. Include all photographic evidence. Peer Review: If applicable, have the study peer-reviewed to ensure accuracy and validity of the experimental approach and findings.
This structured approach allows for a thorough investigation of adenovirus cytopathic effects, providing valuable insights into the virus-cell interactions and the efficacy of potential antiviral treatments. Regular and meticulous monitoring coupled with detailed documentation are key for a successful analysis of adenovirus CPE.
Reviewing the protocols listed here may offer further guidance in addressing this issue
  • asked a question related to Transfection
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 Transfection
Question
1 answer
I am performing PCR as a QC test to look for a transcription gene that should be negative after a CAR T therapy process. As we are comparing against a CAR transduced patient's cell, we require a used transduced ATCC cells. However, the ATCC cells have a low transfection titer, which makes the PCR band faint and when kept for long, it becomes fainter and fainter.
I was thinking of using another different grade of cells such as transduced research grade cells as it was observed that the bands tend to be much brighter than the ATCC grade cells.
Is it possible to use transduced research grade cells instead of ATCC grade?
Relevant answer
Answer
Using a different grade of cells for your positive control could potentially introduce variability or inconsistency into your experiment. It's generally recommended to use the same grade or quality of cells for both experimental and control groups to ensure reliable and accurate results. However, if you have valid reasons for using a different grade of cells for your positive control, it's essential to thoroughly validate and justify this decision to ensure the integrity of your experimental findings.
  • asked a question related to Transfection
Question
2 answers
Hi, I have transfected my HEK293 cells with pcDNA3.1_plasmid of interest with EGFP by using lipotransfection method. Post transfection 48h the egfp expression looks good with more than 50% transfection rate. Started G418 selection (DMEM+10% FBS+ 2mg/mL G418). Post transfection 48h, the cells transfer from 24wp to 12wp to create space for G418 medium to get to non-transfected cells. Medium replacement done daily, and after a week, fresh selective medium is prepared. Massive cell death observed after 2 days of G418 selection.
Day 7, 95% of non transfected cell die. Colonies/ egfp rounded cell clump together can be observed and a lot of resistant cells survived but did not attached under the G418 selective stress.
Day 8, the selective medium reduce to 1mg/mL and Day 10 reduce to 500ug/mL and day 12 reduce to 250ug/mL hoping that the resistant cells will attach and starting to expand.
Unfortunately, the non transfected cells starting to grow rapidly and the cells with EGFP did not grow so much. Any comments and thoughts are welcome. The reason I use 2mg/mL from to start becuase I have done 1mg/mL to kill the HEK293 but it wasn’t effective. The active G418 percentage is 80.9% and I have reconstituted G418 with 100mM HEPES buffer And filtered.
I have read about for the survival cells to expand it might take a long time under the selective pressure. When is the right time to reduce the antibiotic?
the attached image is Day 10 under G418 selection. Should I be more patient and keep the G418 concentration high and wait for the colonies to attach and expand?
Relevant answer
Answer
Sabine Strehl thank you for answering! Under G418 selection pressure, the resistant cells did not die but not exert their typical morphology even after 10 days. The non resistant cells die 99% but not completely, as I reduce the antibiotic level the non resistant cells overgrowth than the resistant cells that express EGFP and slowly my resistant cells reduce and die off and non resistant cells overgrowth. I use medium with G418 for 7 days then prepare a fresh one, but on 7th day of the medium, the antibiotic seems wear off the potency after a few water bath thaw.
I will repeat the transfection and selection process. Thank you for your suggestion.
  • asked a question related to Transfection
Question
1 answer
I am working on the Primary neuron cell. I am trying to observe the stress granule in the cell. For that, I have done transfection with my gene of interest and treated the cell with Arsenite. I want to do Fluorescence recovery after photobleaching assay to know the mobility of the proteins. Can anyone tell you how you have performed the FRAP assay?
Relevant answer
Answer
Fluorescence Recovery After Photobleaching (FRAP) is a technique used in cell biology and microscopy to study the dynamics of molecules within living cells. Here's how it works:
  1. Principle: FRAP involves selectively bleaching a small region of fluorescently labeled molecules within a cell using a high-intensity laser. This bleaching process irreversibly destroys the fluorescence of the molecules in the targeted area.
  2. Observation: After bleaching, the recovery of fluorescence intensity in the bleached region is monitored over time. The recovery occurs as fluorescent molecules from the surrounding unbleached regions diffuse into the bleached area.
  3. Analysis: The rate and extent of fluorescence recovery provide information about the mobility, turnover, and interactions of the labeled molecules within the cell. Different models can be used to analyze FRAP data, such as diffusion, binding, or compartmentalization models, depending on the biological context.
  4. Applications: FRAP is used to study various cellular processes, including protein diffusion, membrane dynamics, cytoskeletal dynamics, and protein-protein interactions. It can also be used to assess the effects of drugs or genetic manipulations on molecular mobility within cells.
FRAP is a powerful tool for understanding the spatiotemporal dynamics of molecules in living cells, providing insights into their behavior and function in complex biological systems.
  • asked a question related to Transfection
Question
5 answers
Hello. I've been trying to transfect plasmid DNA to my breast cancer cell lines (BT-474 and SK-BR-3) with Thermo Fisher's Lipofectamine 3000 reagent. According to the thermo fisher, those cells have almost %50-70 efficiency but I only managed %10.
I use 25 µL optimem- 1 µL Lipofectamine and 25 µL optimem- 250 ng DNA- 0.5 µL P3000 for BT-474 and
25 µL optimem- 0.5 µL Lipofectamine in one tube and 25 µL optimem- 500ng DNA- 1 µL P3000 for SK-BR-3 as thermo's protocol says. I am using GFP expressing PX458 plasmid to transfect but I don't know why I'm facing this low efficiency problem. Has anyone face that before?
Relevant answer
Answer
honestly whatever works, works. Try out electroporation and see what happens.
  • asked a question related to Transfection
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 Transfection
Question
4 answers
We are using pMSCV for transit expression of a protein.:
1. The gene was cloned in (between XhoI/EcoRI).
2. Transfection with lipofectmin 3000 to 293t cell.
3. After 48 hrs, GFP can be observed. 4. But my target by WB.
Please suggest what could be the problem
Relevant answer
Answer
if "there's no promotor before my target gene"... then you have the answer
  • asked a question related to Transfection
Question
3 answers
Quite a naive question
I am looking for the most optimal way to transfect different cell lines with the same construct. Some of them are notoriously difficult to transfect, like RWPE1
I am satisfied with the quality of transient DNA transfection, which I did on simple lines like HEK293 or HeLa. But maybe it’s time for me to somehow optimize the process? Please advise, maybe it’s time for me to learn CRISPR? pLenti? Something else?
talk to me please
Relevant answer
Answer
It is difficult to have an single optimal way for every cell lines, each cells have their own strength driving multiple signaling pathway, upper limit of the plasmid copy received and interaction between GOI and its own proteins, hence affecting gene expression facility. We had tried at least four different transfection method with more than ten different reagents just to make a cell population to express 0.1% of GOI, while in 293T the expression was almost hitting the upper limit of detection in flow cytometry. Therefore, to generate an optimized protocol for each cell lines or primary cells are considered easier then to generate a single protocol for universal use. The cloning process or to achieve a single double KI/KO clone by CRISPR is very time consuming and troublesome, if you want to go through CRISPR, try look at the newest technique such as base/prime editing by Dr. David Liu. In the other hand, lentivirus is not, in our lab, lentivirus is an useful tool for large scale cell line production, a single production process may grant us enough amount of lentiviral particle for more then 100 times of usage. However, if a gene is already being difficult to express in such cell lines, I would rather look into an optimization of DNA construct, a small twist such as codon optimization or driving promoter sometimes grant you more then 50-fold of difference, that will be far practical then transfection process.
Best
  • asked a question related to Transfection
Question
5 answers
I have transfected cells with GFP, which emit in close range to FITC and used apoptosis kit that have annexin conjugated with FITC to measure apoptosis and PI to measure death. I am gating for GFP separately but feel it is probably false positive for apoptosis because the fluorescence spill over? Is this correct? need some thoughts? thank you
Relevant answer
Answer
Asma El-Howati Yes, that is true. But why bother with unmixing GFP from FITC, if you can simply run the assay using AnnV-APC for instance. The cleaner the setup the better :)
Cheers.
  • asked a question related to Transfection
Question
7 answers
Hello ! I m a master just starting work in CRISPR genome editing ,
and trying to knock-in a reporter in my interesting gene
my method is transfect RNP and dsODN by lipo2000 to 293T cell line ,there is variety RNP and donor DNA concentration found in paper (few nM to 60nM RNP , about 500ng or even 18nM ODN ect)
In my experiment , I have a donor DNA about 4kb ,
so I perform a set of test using 60nM RNP (1:1 Cas9/gRNA) with 50 to 400 ng dsODN , but get no successful
at next condition test , I perform 0,15,30 nM of RNP and positive plasmid control to test transfection efficiency and get the result EGFP may get lower with higer RNP concentration.
whether the too giant size of molecular and negative charge give rice to barrier when liposome formation in my condition?
is my donor DNA too big or it must should be plasmid or ssODN?
or just this method working in even low efficiency?
please any good condition and advice !
sincere thanks.
and sorry for too much question and typo
Relevant answer
Answer
Dear [Recipient],
I hope this message finds you well. Your inquiry regarding the use of liposomes for the delivery of Cas9/gRNA ribonucleoprotein (RNP) complexes and double-stranded oligodeoxynucleotides (dsODNs) is both timely and relevant in the context of genome editing technologies. This delivery strategy is increasingly recognized for its potential to facilitate efficient, targeted genome editing by reducing off-target effects and enhancing delivery efficiency. Below, I provide detailed advice and considerations for employing liposomes in this cutting-edge application.
Understanding the Delivery System:
Liposomes are versatile, lipid-based vesicles capable of encapsulating nucleic acids and proteins, facilitating their delivery into cells. Their biocompatibility, low immunogenicity, and ability to merge with cell membranes make them an attractive delivery system for Cas9/gRNA RNP and dsODNs.
Preparing Cas9/gRNA RNP and dsODNs:
  1. Cas9/gRNA RNP Complex Formation:Prioritize the use of high-purity, recombinant Cas9 protein and synthetically derived gRNA. Mix them in a stoichiometric ratio to form the RNP complex. Incubation conditions for complex formation can vary, so optimization may be required.
  2. dsODN Preparation:Ensure that the dsODNs, used as repair templates for homology-directed repair (HDR), are synthesized with high fidelity and purified to remove any contaminants that might affect liposome encapsulation and cell uptake.
Liposome Preparation and Complex Loading:
  1. Liposome Selection:Choose liposomes with a composition and size suitable for RNP and dsODN delivery. Cationic liposomes are often preferred due to their ability to form complexes with the negatively charged nucleic acids and proteins.
  2. Encapsulation Efficiency:Optimizing the encapsulation efficiency is crucial. This may involve adjusting the ratio of liposomes to the RNP/dsODN complexes, the concentration of components, and the incubation conditions. Utilize assays to quantify encapsulation efficiency and adjust protocols accordingly.
  3. Protective Measures:Incorporate strategies to protect the integrity of the RNP and dsODN during the encapsulation process. This might include the use of buffers that stabilize the RNP complex and prevent dsODN degradation.
Cell Targeting and Uptake:
  1. Cell Type Considerations:The efficacy of liposome-mediated delivery can vary significantly between cell types. Characterize the uptake efficiency and cellular toxicity in your target cells, optimizing liposome size and surface charge as necessary.
  2. Enhancing Cellular Uptake:Explore strategies to enhance cellular uptake, such as incorporating targeting ligands into the liposome formulation, which can facilitate receptor-mediated endocytosis in specific cell types.
Monitoring Editing Efficiency and Off-target Effects:
  1. Assessment of Genome Editing:Employ sensitive and specific assays to evaluate the efficiency of genome editing and HDR-mediated insertions, such as T7 endonuclease I assay, PCR-based assays, or next-generation sequencing.
  2. Evaluation of Off-target Activity:Utilize computational tools and experimental assays to predict and assess off-target effects, ensuring that the delivery method does not exacerbate unwanted genomic modifications.
Conclusion:
Liposome-mediated delivery of Cas9/gRNA RNP and dsODNs represents a promising avenue for enhancing the specificity and efficiency of genome editing applications. Success in this approach requires careful consideration of liposome composition, optimization of encapsulation processes, and thorough evaluation of delivery outcomes in target cells. Continued innovation and optimization in this area are expected to further refine these techniques, broadening their applicability and effectiveness in both research and therapeutic contexts.
Should you require further assistance or wish to explore additional aspects of this delivery method, please do not hesitate to reach out. I am here to support your research endeavors.
Best regards,
Take a look at this protocol list; it could assist in understanding and solving the problem.
  • asked a question related to Transfection
Question
2 answers
kindly provide some insights
Relevant answer
Answer
Transfecting macrophages is especially difficult since macrophages are professional phagocytes that are very sensitive to foreign materials. Introduction of foreign DNA activates the Toll-like receptor 9 (TLR9) pathway leading to the production of cytokines and nitric oxide. These activated macrophages may then be less responsive to treatment that one may intend to examine.
You may need to optimize the protocol for various parameters such as timing of transfection, type of transfection reagents, amounts of transfection reagents and of plasmid DNA, as well as ratio of transfection reagent to plasmid DNA.
One critical step for RAW264.7 which you need to consider is the health of the cells. Overgrown cultures are not suitable for transfection as their physiology changes, and continuous culturing of RAW264.7 cells for a long period of time can also change the phenotype and function of the cells. Freshly thawed cells that have a low passage number are recommended for transfection.
Another important consideration is the choice of transfection reagents. Lipid-based transfection reagents are typically used in research due to its ease of use and commercial availability. The cells should be incubated with the transfection reagents for a few hours instead of overnight. Longer incubation time with the transfection reagent will increase transfection efficiency, but it can also be harmful to the cells either causing cell death or cell activation, both of which can interfere with the experimental design. Also, the time between transfection and experimental treatment (the rest time) is crucial.
You may want to refer to the paper attached below. It will be helpful!
Good Luck!
  • asked a question related to Transfection
Question
1 answer
Hi all,
My gene causes the cancer cells to grow very slowly when it is overexpressed. Is there a method to make the cells move faster?
Relevant answer
Answer
Your gene may be stressing your cells. This may be because the gene is so overly expressed it's consuming too much of the cells' resources, leading to stress and slow growth. The gene itself may be toxic or problematic in some way too. Not knowing the cell line or the gene, it's hard to say what may be happening.
  • asked a question related to Transfection
Question
10 answers
Hello Good people
When I stained my adherent non-transfected cells with Hoechst 33342 staining it showed blue fluorescence but dull staining happened with GFP transfected cell
I used 2ug per molar
30 min incubation at RT
300ul per well in 12 wells plate
So, what's your suggestion for better procedure to be able to see cell segmentation more clearly!
What's the benefits from PBS washing as recommended by some protocols at the beginning or the end!
Relevant answer
Answer
May consider using Permai fluorescence dye.
  • asked a question related to Transfection
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 Transfection
Question
5 answers
Dear All,
I am encountering challenges with AAV-mediated neuronal transduction, where high volumes lead to significant cell death, while low volumes yield inadequate signal levels.
Here's a brief overview of my AAV production process:
  1. I produce AAV in HEK cells using Fugene transfection.
  2. Following transfection, I lyse the cells through freeze-thaw cycles.
  3. I purify the lysate overnight using PEG8000 without chloroform purification.
  4. After purification, I pellet the virus and dilute it in Tris buffer.
For titration, I've tried volumes ranging from 10 microliters to 0.2 microliters. At 1 microliter, the signal is optimal, but it coincides with significant cell death. Volumes lower than this are not feasible for analysis due to inadequate signal.
I'm seeking insights into the possible reasons behind the observed cell death during transduction. Your input would be greatly appreciated.
Relevant answer
Answer
Hey,
I prepared the buffer.
When I transduce, my neurons are in neurobasal phenolred negative, I cannot check colour change.
But I tried only buffer titration from 1 microliter to 50 microliter. It made no cell death.
  • asked a question related to Transfection
Question
5 answers
Hello! I am growing transfected cells in 24 well plate. on the bottom of each well i have a small glass cover-slip. so the cells adhere to that cover slip. I am using this method because its very easy to transfer that glass to a slide and then analyse for fluorescence. the only problem is that DAPI staining efficiency is super low. I am simply covering the glass on which the cells are growing with DAPI for 5 minutes and then analyzing. Is there another protocol that I should use in this case?
Thank you!
Relevant answer
Answer
May consider using Permai fluorescence dye.
  • asked a question related to Transfection
Question
2 answers
Hello everyone,
I am trying to make a CRISPR-Cas9 knockout on lung cancer primary cells using the RNP complex. I am using the Neon Transfection System but so far it failed - all the cells that took the complex were dead. The settings I tried were: puls voltage 1200 v, width 30 ms, pulse number 2.
I am planning to use their optimization protocol on the 24-well plate but maybe any of you have already tried a similar edition on any type of primary cancer cells and can share the settings used?
Relevant answer
Answer
Sure. The settings working the best for me were 1400 voltage, 20 pulse width and pulse number 2. The other one that was working quite fine was 990/40/1 (better transfection but higher mortality) and 1200/40/1 (worse transfection rate but higher viability). Hope you find it helpful!
  • asked a question related to Transfection
Question
1 answer
Hi there, as asked in the title, because we already have these two transfection reagents in lab for Cas9/sgRNA complex transfection, I just want to know if I can use any of them instead of Lipofectamine RNAiMax when I need to transfect only the sgRNA into a cas9-expressing tumor cell line. If so, any experiences on how you used it and how did it work? Thanks!
Relevant answer
Answer
Never used these two before. But I used Lipofectamine 2000 to deliver the sgRNAs to the Cas9-expressing cells back in 2017, and it worked.
  • asked a question related to Transfection
Question
2 answers
Hello,
I am Mahmuda, now I am working with DG44 cell culture. So far, my cell culture viability improved to 90%. However, before transfection, my cell viability decreased to 70%. I am very disappointed with the results of this culture.
Is there any suggestion or input that can be given so that I can solve this problem?
Then, is there any particular trick to do for this DG44 culture?
Thank you for the help
Relevant answer
Answer
DG44 cells are a type of Chinese hamster ovary (CHO) cell line commonly used in biopharmaceutical research and production, particularly for the production of recombinant proteins and monoclonal antibodies. Several factors can affect the viability of DG44 cells, including:
  1. Culture Medium and Nutrients: The composition of the culture medium and the availability of essential nutrients are critical for cell viability. Ensure that the medium is properly formulated with appropriate concentrations of glucose, amino acids, vitamins, salts, and growth factors necessary for cell growth and metabolism.
  2. pH and Buffering Capacity: Maintaining the pH of the culture medium within the optimal range is crucial for cell viability. Fluctuations in pH can adversely affect cell growth and metabolism. Additionally, ensure that the culture medium has adequate buffering capacity to resist pH changes over time.
  3. Temperature: DG44 cells are typically cultured at 37°C in a CO2 incubator. Maintaining the proper temperature is essential for cell viability. Fluctuations in temperature can affect cellular processes and compromise cell health.
  4. Osmolality: The osmolality of the culture medium should be maintained within the physiological range to prevent osmotic stress on the cells. Hypo- or hyperosmotic conditions can lead to cell swelling or shrinkage, respectively, affecting cell viability.
  5. CO2 Levels: DG44 cells are typically cultured in a humidified atmosphere containing 5% CO2. CO2 is necessary for buffering the culture medium and maintaining the optimal pH for cell growth. Ensure that the CO2 levels are properly regulated to support cell viability.
  6. Cell Density and Confluence: Cell density and confluence in the culture vessel can affect cell viability. Overcrowding can lead to nutrient depletion, waste accumulation, and reduced viability. Conversely, low cell density may result in suboptimal growth and viability.
  7. Cell Passage and Subculture: Proper handling during cell passage and subculture is essential to maintain cell viability. Avoid over-trypsinization or excessive mechanical stress during cell dissociation, as it can damage the cells and decrease viability.
  8. Contamination: Contamination with bacteria, fungi, mycoplasma, or other microorganisms can compromise cell viability. Follow strict aseptic techniques and regularly monitor cultures for signs of contamination.
  9. Cell Line Health and Stability: The genetic stability and health of the DG44 cell line can impact cell viability over time. Monitor cell morphology, growth rate, and productivity regularly to ensure consistent performance.
By carefully optimizing these factors and maintaining appropriate culture conditions, you can enhance the viability and robustness of DG44 cell cultures for various applications in biopharmaceutical research and production.
  • asked a question related to Transfection
Question
2 answers
Hi everyone. I want to DNA isolate from transfected cell culture medium (transfected with lentivirus). I tried lots of kit but DNA cons and quality not very well and I didnt show at agarose gel.
Relevant answer
Answer
DNA extraction from cell culture medium can be challenging due to the presence of various contaminants such as proteins, salts, and other cellular debris. However, several methods can be used for DNA extraction from cell culture medium, with some modifications to traditional DNA extraction protocols. Here are two commonly used methods:
  1. Phenol-Chloroform Extraction:This method involves organic extraction of DNA using phenol-chloroform followed by ethanol precipitation. Collect the cell culture medium and centrifuge it to pellet any cells or debris present. Transfer the supernatant to a fresh tube and add an equal volume of phenol:chloroform:isoamyl alcohol (25:24:1). Mix the solution thoroughly and centrifuge it to separate the organic and aqueous phases. Transfer the aqueous phase containing DNA to a fresh tube and precipitate DNA by adding ethanol and salt (e.g., sodium acetate). Centrifuge to pellet the DNA, wash with ethanol, and resuspend the DNA in an appropriate buffer.
  2. Commercial DNA Extraction Kits:Many commercial kits are available specifically designed for DNA extraction from cell culture supernatants. These kits typically use silica-based columns or magnetic beads to capture DNA selectively, removing contaminants. The procedure generally involves binding DNA to the column or beads, washing away contaminants, and eluting pure DNA in a low-salt buffer.
Regardless of the method chosen, it's essential to consider the following tips for successful DNA extraction from cell culture medium:
  • Remove any cellular debris by centrifugation before starting the extraction process.
  • Use fresh reagents and ensure proper handling to minimize contamination.
  • Optimize the extraction protocol according to the specific characteristics of the cell culture medium and the intended downstream applications.
  • Evaluate the quality and quantity of extracted DNA using spectrophotometry or fluorometry.
  • Store extracted DNA appropriately at -20°C or -80°C for long-term stability.
The choice of method may depend on factors such as the available equipment, time constraints, and the downstream applications for the extracted DNA. Always follow the manufacturer's instructions or established protocols carefully to obtain reliable results.
  • asked a question related to Transfection
Question
2 answers
I want to use the TriFECTa RNAi kit for transfection, but I dont know how much RNA and control transfection should I use.
Relevant answer
Answer
assistant avatar
. مجموعة TriFECTa RNAi تستخدم لتقوم بصمم تحدبات RNA لتثبيط الجينات المحددة في الخلية. لاستخدام هذه المجموعة في الترانسفيكشن، يجب عليك أولاً تحضير ال
RNA
المستهدف باستخدامها.
لتحديد كمية وتركيز ال
RNA
المستهدف الذي يجب استخدامه، يعتمد ذلك على عوامل عدة مثل نوع الخلية المستهدفة والجين المراد تثبيطه وكفاءة عملية التحويل (transfection) الخاصة بك. عموماً، يمكن البدء بتجارب تجريبية باستخدام تراكيز مختلفة لـ RNA ومراقبة التأثير على تعبير الجين المستهدف.
بالإضافة إلى ذلك، من المهم أيضاً استخدام جهاز للتحكم في عملية الترانسفيكشن لضمان انتقال الRNA بشكل فعال إلى الخلية. يمكنك الاعتماد على العديد من جهوز الترانسفيكشن المتاحة مثل Lipofectamine أو Polyethylenimine.
  • asked a question related to Transfection
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 Transfection
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 Transfection
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 Transfection
Question
2 answers
For transfection we have used 500uL of transfection complex for 1 well of 6 well plate. so total media added to each well was 2mL. According to this 100nM of sirNA and 100ug/mL of nanoparticle was added in the transfection complex (500uL). What is the ratio of SiRNA to media. (we have a stock of 100micro molar of siRNA )
Also what if we lower down the volume of media to 50uL. Then how to maintain the ratio of siRNA to media?
Relevant answer
Answer
125ul optimem + 1,5ul 20microM RNAi
125ul optimem+2,5 ul RNAi max mix togheer and wait 20min RT
in the meantime replace the medium on your cells (i plate 2 10e5cells/2ml 6wells
with 1,25ml DMEM FBS witout ATbio
add the transfection medium overthe cells, mix..final volume 1,5ml analyse at 48 or 72Hours...work great (some people remove the medium after 2h and add complete media!)
work great see photo
  • asked a question related to Transfection
Question
5 answers
Hi everyone. Has anyone worked with Raji cell transfection? I recently used AMAXA technology to transfect Raji cells with a CRISPR construct that resulted in a very poor efficiency (~1%). When I tried the same construct in HEK293T cells with Xtreme gene9 I got more than 80% transfected cells plus very high levels of GFP expression that was part of the cassette. Any alternative suggestions for RAJI transfection? I know that Xtreme gene9 probably won't work well on them...
Relevant answer
Answer
Transfecting Raji cells efficiently can be achieved by optimizing several factors, including the choice of transfection reagent, cell density, and transfection conditions. Here's a general protocol along with some suggestions to enhance the efficiency of transfection in Raji cells:
Materials:
  1. Raji cells
  2. Plasmid DNA or siRNA/RNAi oligonucleotides
  3. Transfection reagent (e.g., Lipofectamine 3000, TransIT-X2, or other suitable reagents)
  4. Opti-MEM or RPMI medium
  5. Fetal bovine serum (FBS)
  6. Antibiotics (optional)
  7. Culture dishes or plates
  8. Pipettes and tips
Protocol:
  1. Cell Preparation:Culture Raji cells in RPMI medium supplemented with 10% FBS and antibiotics (if necessary) in a humidified incubator at 37°C with 5% CO2. Ensure that cells are in log-phase growth and have a high viability (>90%).
  2. Transfection Complex Preparation:Dilute the plasmid DNA or siRNA/RNAi oligonucleotides in Opti-MEM or RPMI medium without serum according to the manufacturer's instructions for the chosen transfection reagent. Prepare the transfection complexes by adding the diluted nucleic acids to the diluted transfection reagent. Incubate the mixture at room temperature for 15-30 minutes to allow complex formation.
  3. Cell Transfection:Seed Raji cells in culture plates or dishes at the desired density the day before transfection to ensure that they reach 60-80% confluency on the day of transfection. Replace the growth medium with Opti-MEM or RPMI medium without serum just before transfection. Add the transfection complexes dropwise to the cells and gently swirl the plate to ensure uniform distribution of the complexes. Incubate the cells with the transfection complexes at 37°C in a CO2 incubator for the recommended duration according to the transfection reagent's protocol.
  4. Post-Transfection:After the transfection period, replace the transfection medium with complete growth medium containing serum to support cell recovery and expression of the transfected genes. Incubate the cells for an additional 24-48 hours to allow for gene expression or knockdown to occur.
  5. Analysis of Transfection Efficiency:Analyze the efficiency of transfection by monitoring the expression of a fluorescent reporter gene if using a reporter plasmid or by assessing the knockdown/knockout efficiency of the target gene if using siRNA/RNAi. Perform downstream experiments or analyses to investigate the functional consequences of gene expression modulation.
Optimization Tips:
  • Test different transfection reagents and concentrations to find the most efficient one for Raji cells.
  • Optimize the ratio of DNA or RNA to transfection reagent for maximum efficiency and minimal cytotoxicity.
  • Consider using a selection marker (e.g., antibiotic resistance gene) in plasmid transfections to enrich for transfected cells and improve efficiency.
  • Try different cell densities and seeding conditions to find the optimal conditions for transfection efficiency.
By carefully optimizing these parameters and following the protocol, you can achieve efficient transfection of Raji cells for your experiments.
l With this protocol list, we might find more ways to solve this problem.
  • asked a question related to Transfection
Question
2 answers
what are the possible reason(s) that make AsPC-1, 293T and MCF-7 cell lines failed to be transfect/transduced?. Please help me with possible hints?
Relevant answer
Answer
You may consider the following reasons.
1. The choice of which cell type to use for transfection is a critical factor that is often overlooked. Since each cell type is likely to respond differently to a given transfection reagent, choosing an appropriate cell type is necessary to maximize results. For instance, some cell types like MCF7 or HepG2, prefer to grow in clumps or clusters which is not ideal for transfection because minimal membrane surface is exposed which compromises uptake. Blood or immune cells that lack the proper endocytic machinery can minimize uptake and there are the macrophages that have an evolved uptake mechanism, but quickly breakdown and destroy endosomal contents. So, select the right cell line for transfection.
2. Make sure that the cells are not 100% confluent (should be about 60-80% confluent) or in stationary phase at the time of transfection because actively dividing cells take up foreign nucleic acid better than quiescent cells.
3. Most cell types should be used between 4 and 25 passages for optimal transfection.
4. There could be a possibility that the DNA concentration may be too low, or DNA may be degraded. In such a case, you may increase the ratio of DNA :transfection reagent, and you may confirm DNA integrity by A260/A280 spectrophotometer reading (should be at least 1.7).
5. You may use serum-free media for DNA dilution because some serum proteins may interfere with complex formation. You may also increase complexing reaction incubation time.
6. Test the culture for contamination.
7. Do not use antibiotics at the time of transfection because cationic lipid reagents increase cell permeability. As a result they may also increase the amount of antibiotics delivered into the cells, causing cytotoxicity and low transfection efficiency.
8. Cells could suffer mechanical damage during the experimental steps. So, do not vortex or spin cells for extended period.
9. Some transfection reagents could be compromised due to improper storage.
Best.
  • asked a question related to Transfection
Question
1 answer
Background: I recently seeded HEK cells on a poly-L-lysine coated plate and used those for transfection. My vectors are backsplicing vectors with the ZKSCAN introns which generates circular RNAs so it takes a while for the GFP signal to be observable with a microscope, even for my most active IRES of interest (more active than EMCV and comparable to c-myc 5'UTR). Most papers, like this one:
grow cells for 4 to 5 days. However, I found that cells would become more confluent, acidify the media too fast and die. Then, I might lose the GFP-expressing cells. I tried changing media everyday when cells reach high confluency, but the media always turn very yellow the next day. If I seed fewer cells, then they may become too sensitive to the transfection, as I have noticed especially for the backsplicing vectors. Coating the plate with poly-L-lysine did help tremendously to prevent cell death after transfection, but after 2 days cells begin to die.
Question: So for experiments that require longer incubation/treatment periods, what do people do to maintain cell health at high/100% confluency?
Relevant answer
Answer
You can add less serum to the culture medium (e.g. 2%)
  • asked a question related to Transfection
Question
3 answers
Hello everyone,
I use several pcDNA3.1 expression vectors to transfect cells.
The vectors were prepared by midi-prep a year ago and diluted in TE buffer.
Now that I run new experiments, I decided to measure plasmid concentrations again, prior to transfection.
All their concentration have droped by 2 to 3-fold.
260/280 ratio are still good (over 1.8), but strangely 260/230 ratio have risen (from 2 to 2.3~2.5).
Given the good 260/280 ratio, the presence of EDTA in the buffer and the -20°C storage, I'm pretty sure it is not degradation.
It could be adsorption of DNA on eppendorf tube wall but given the 100~500ng/µL range of concentration, I don't think any tube surface could sequester this much vector quantity.
Anyway I heated my vector for 15min to 60°C and votexed it without increasing the measured concentration ?
The only thing I see would be freeze/thaw cycle maybe ? (I did 10 to 20 such cycles...)
Should I add glycerol to my TE so that freezing and ice crystals don't shear my vector ?
Or just aliquot my vector?
Where did my vectors go guys ???? ^^
Thanks for the help you can provide,
Philippe.
Relevant answer
Answer
But remember it might be the original measurement that was off. Lastly it might not matter, use the lower concentration as real. and It won’t hurt if there is a bit more than expected Unless you need to be very quantitative
  • asked a question related to Transfection
Question
1 answer
Hello! Can you give me some tips how to do CRISPR-Cas9 more effectively? Maybe you can give me some advice on this topic. This technology is new for me, I tried to conduct the experiments to knock-out genes, but they weren't successful. How do you detect knock-outs? What is the best combination of conditions to carry out CRISPR-Cas9?
Relevant answer
Answer
You usually detect knock outs by knocking in a cassette with a selectable marker gene or cutting in two locations which causes a deletion large enough to be detected with PCR. Double check that your plasmids are using promoters appropriate for your organism. If you are simultaneously trying to knock in a marker then you can combine the system with single strand annealing proteins used for recombineering (lambda red for prokaryotes and ICP8 for eukaryotes).
  • asked a question related to Transfection
Question
2 answers
I am trying to overexpress a TurboID construct in mouse primary culture cells for validation experiments. Unfortunately the transfection of this construct appears to be killing my cells while other constructs (i.e. GFP) transfect just fine. My current hypothesis is that the turboID is depleting the cell's biotin supply since they are grown in a serum-free media and the only source of biotin is the B27 supplement. In order to alleviate this, I'd like to supplement their media with free biotin, but have little experience in the use of biotin as a cell culture supplement. I was wondering if there there specific kinds of biotin I should purchase for this purpose and if anyone has experience with what a safe starting dosage for primary culture cells may be.
Relevant answer
Answer
Hi Nick,
Supplementation of media with biotin for serum-free culture of mammalian cells is a little complex by the fact that biotin is susceptible to oxidative stress and inactivation by UV radiation. Oxidation products of biotin are biotin sulfoxide and biotin sulfone. For more information on micronutrients in cell culture, you may want to refer to the article attached below.
The safe starting dosage for primary culture cells may be optimized as in cell culture media, the concentration range for biotin varies from 0.01 to 0.82 (μmol/L).
You may purchase biotin powder for use in cell culture from the link provided below.
Best.
  • asked a question related to Transfection
Question
1 answer
I infected the 293T cell line with TTV. Before that, I transfected the Jurkat and Raji cell lines with TTV, but after 10 days of infection, the viral titer dropped.
Relevant answer
Answer
it could be that the 293T cells may have more robust antiviral responses compared to Jurkat and Raji cells, leading to the inhibition of TTV replication. This could include the activation of interferon response pathways or the expression of restriction factors that limit viral replication.
  • asked a question related to Transfection
Question
3 answers
Hello,
Context: I am trying to purify and quantify my AAV containing my plasmid of interest. I used HekAAV cells from Takara and transfected the cells with my plasmid of interest, pRepcap and pHelper. I extracted and purified my AAVs with the Takara midi kit. I ran an 8% SDS gel and was able to visualize VP3s and feint VP1 proteins.
Problem: In order to quantify the titer of the AAV, I treat my samples with DNase and proteinase K and run a qPCR with primers for the ITR of the AAV. My standard shows expected Cq increase with higher folds of dilution but for the same folds of dilution, my AAVs show the same Cq values. I repeated and observed similar Cq value with no change with change in dilutions.
Assumptions: The AAV are not well transfected and only the capsids are expressed. Both the plasmid of interest and pHelper were not transfected well. So, I only see VPs on SDS gel but no change in Cq values in PCR.
Looking for advice or suggestions as to what may have happened and how I can improve my titer quantification data.
Relevant answer
Answer
@Hieu Nguyen
My negative control (water + qPCR mix) was expected to have the higest Cq but it had Cq similar to my samples at its highest dilution. This makes me think my water or master mix was contaminated and my titer was very low so it amplified the contaminant i guess.
For primers, I have been using primers for the ITR regions. They have been yielding reliable results till now.
  • asked a question related to Transfection
Question
4 answers
I have utilized pET26b(+) NS2B-NS3 DENV2, and transfected it in E.coli. After transfection the selected cells were characterized based on the kanamycin resistance present in the vector. The selected cells were incubated with 1 millimolar of IPTG at 28 C for 4 hrs induction. While the results seem positive for the experiment, multiple bands occur underneath the protein of interest. the selection was done based on the histidine tag using penta-his antibody. Please help me with this concern. I am using TMB for the development of the blot.
Relevant answer
Answer
If you have not yet tried adding protease inhibitors when preparing the protein lysate then that would be the first thing I would try. Secondly you might reduce the time after induction or reduce the temperature a bit more.
Lastly the level of these products is fairly minor and you might just ignore them.
However I would note that you DO NOT know if your protein is pure or not. Since you are doing a western blot by definition you are only seeing the proteins that are detected. You might wish to run a protein gel and stain to get an idea of the actual purity, if this is something important to you. But truthfully for many applications you do not need pure protein.
  • asked a question related to Transfection
Question
3 answers
Hi everyone, I transfected HEK cells with a protein that is intracellularly expressed through flow. Then I isolated the protein using RIPA buffer protocol, however, I accident added IP buffer instead of RIPA and now I do the know if it’s worth going through the whole western blot process.
For more context, I only know the protein shows intracellular expression through flow but don’t know exactly where it’s expressed. I looked up and ThermoFisher said IP is less harsh than RIPA so I don’t know if it collects everything or not.
Any advice is appreciated!
Relevant answer
Answer
Can you please share the composition?
  • asked a question related to Transfection
Question
4 answers
I have been transfecting a dendritic cell line for about a year now with about a 15-30% transfection efficiency that I would like to increase to 40-50%. I have used Neon Transfection (electroporation) and chemical transfection (JetOptimus) and changed multiple parameters for the electroporation and the chemical transfections with no success. My largest plasmid is ~6.8 kb and is being co-transfected with GFP at ~4.5 kb.
Relevant answer
Answer
I see your plasmid size is high and that's why instead inserting GFP inside your plasmid (Not to exceed the sized more that can be linked with multiple issue even cell death) you are co-transfecting the cells with GFP plasmid.
Now, there is something missing in your question, how much of your plasmid you use along with how much of GFP concentration, here for sure you will use higher concentration of your blind plasmid and a little of GFP just to get a little signal of transfection. If this is the case, then 15 to 30% transfection is quite fine because your blind plasmid would be more in number as compare to the lower amount/concentration of GFP that is co-transfected with your plasmid, so ideally more of your plasmid would have been transfected as compare to lower amount of GFP plasmid.
If this is the case and let say, you use 1 ug of total both plasmid (100 ng GFP+900 ng Your plasmid) or (300 ng GFP + 700 ng Your Plasmid) in 24 well plate, just have a try to use the total amount of GFP plasmid alone to mimic your original condition, you will get good transfection because there will be only GFP (in total) that is responsible for the signal, and hence you can have a comparison justification for actual transfection (Your plasmid+GFP).
If you can let us know the amount of plasmid that you are using (Both your plasmid+GFP plasmid).
kind regards
  • asked a question related to Transfection
Question
6 answers
I recently tested PEI for production of GFP lentiviruses in HEK-293T cells. Important parameters in my protocol were as follows:
- PEI solution (1 ug/uL): branched PEI (sigma408727) was diluted 1:1000 in water -> pH was adjusted to 6 -> filter-sterilization
- Transfection: 9 uL packaging plasmids mix (invitrogen) + 3 ug pLenti6.3-GFP + 1mL serum free DMEM + 36 ul PEI solution -> vortex (5 times, 1s each) -> 15 min @ RT -> added to cells dropwise (~5*10^6 cell/10 cm plate)
I checked the packaging cells for GFP expression. The lipofectamine control (with manufacturer protocol) was fine but PEI didn't work at all. What may be the problem with my protocol?
Relevant answer
Answer
Dear Esteemed Colleague,
Thank you for your inquiry regarding the use of Polyethylenimine (PEI) for cell transfection and lentivirus production. PEI is a cationic polymer widely utilized in biotechnology and research for the delivery of DNA or RNA into cells due to its efficiency and cost-effectiveness. Here, I provide a comprehensive guide on employing PEI for both transfection and lentiviral vector production:
Overview of PEI Transfection
PEI transfects cells by condensing DNA/RNA into nanoparticles through electrostatic interactions, facilitating the uptake of these nucleic acids by cells. Its high cationic charge density allows for effective complexation with negatively charged nucleic acids, promoting endosomal escape and enhancing gene delivery.
Preparation of PEI Solution
  1. Dissolve PEI: Dissolve linear PEI (25 kDa) at a concentration of 1 mg/mL in water. Adjust the pH to 7.0 with HCl. This solution can be aliquoted and stored at -20°C for long-term use.
  2. Sterilization: Filter-sterilize the solution using a 0.22 µm filter to remove any particulate matter and ensure sterility.
PEI Transfection Protocol
  1. Plasmid Preparation: For cell transfection, prepare your DNA plasmid mix. For lentivirus production, this typically involves a transfer vector containing your gene of interest, a packaging plasmid, and an envelope plasmid.
  2. PEI-DNA Complex Formation: Dilute the required amount of plasmid DNA in an appropriate volume of serum-free medium (e.g., Opti-MEM). In a separate tube, dilute PEI in the same medium at a ratio of PEI to DNA of 3:1 (µg:µg). Combine the DNA and PEI solutions, mix gently, and incubate at room temperature for 15-20 minutes to allow complex formation.
  3. Cell Transfection: While the DNA-PEI complexes are forming, plate cells in advance so they are 70-80% confluent at the time of transfection. After the incubation period, add the DNA-PEI complexes dropwise to the cells. Gently swirl the plate to ensure even distribution.
  4. Incubation: Incubate the cells with the DNA-PEI complexes at 37°C in a CO2 incubator. After 4-6 hours, replace the medium with fresh complete growth medium to remove any toxicity associated with PEI.
  5. Expression Analysis: Analyze the transfection efficiency and gene expression 24-72 hours post-transfection, depending on your experimental setup.
Lentivirus Production
  1. Transfection for Lentivirus Production: Follow the PEI transfection protocol using HEK293T cells plated in a suitable dish/flask. The cells should be transfected with a lentiviral vector plasmid, along with packaging and envelope plasmids using PEI.
  2. Virus Harvesting: 48-72 hours post-transfection, collect the supernatant containing the lentivirus particles. This can be done by gently pipetting out the medium.
  3. Virus Concentration and Purification (Optional): Concentrate the lentivirus by ultracentrifugation or precipitation methods if a higher titer is required. Filter the supernatant through a 0.45 µm low protein binding filter to remove cell debris.
  4. Storage: Aliquot the virus into sterile tubes and store at -80°C to preserve viral activity.
Key Considerations
  • Optimization: The PEI:DNA ratio may require optimization for different cell lines or plasmid sizes. Start with a 3:1 ratio and adjust as necessary.
  • Toxicity: PEI can be toxic to cells at high concentrations or with prolonged exposure. It's crucial to optimize the amount of PEI and the duration of exposure.
  • Sterility: Maintain sterile conditions throughout the procedure to prevent contamination.
By following these guidelines, you can effectively employ PEI for both efficient cell transfection and lentivirus production. Should you have any further questions or require additional details, please feel free to reach out.
Best regards.
This list of protocols might help us better address the issue.
  • asked a question related to Transfection
Question
6 answers
I’m currently trying to transfect LAN5 cells (human neuroblastoma cell line) using the Lipofectamine 3000 transfection reagent, both with the vector pCMV6-AC-GFP expressing the human heat shock 60kDa protein (Origene RG224428 vector) and with a control plasmid (the same vector without the insert, Origene PS101000 vector).
Using a DNA : Lipo ratio of 1:3, I obtained a good (but not high) transfection efficiency with the control plasmid, but no transfection, or a very low transfection efficiency with the expression plasmid.
Could the plasmid size be responsible for this different transfection efficiency?
Does anyone have any advice to improve the transfection efficiency for both plasmids?
Thanks for your suggestions!!!
Relevant answer
Answer
Dear Esteemed Colleague,
Thank you for reaching out with your query regarding the influence of plasmid size on the efficiency of transfection using Lipofectamine 3000. The question you've posed is essential for optimizing transfection protocols and ensuring the successful delivery of genetic material into cells. Let us explore this matter in a structured and detailed manner.
Lipofectamine 3000 is a widely utilized reagent in molecular biology for the transfection of nucleic acids into various types of cells. It has been engineered to enhance the delivery of plasmids, siRNA, and mRNA into both eukaryotic and prokaryotic cells. The efficiency of this transfection reagent, like others, can indeed be influenced by several factors, including the size of the plasmid DNA being transfected.
  1. Impact of Plasmid Size on Transfection Efficiency: It is generally observed that the size of the plasmid can affect the efficiency of transfection. Smaller plasmids tend to be transfected more efficiently than larger plasmids. This phenomenon can be attributed to the physical and biological barriers larger plasmids encounter during the transfection process. Larger DNA molecules may have a more challenging time entering the cell due to their size and conformation, which can hinder their passage through the cell membrane, even when using highly efficient transfection reagents like Lipofectamine 3000.
  2. Mechanistic Considerations: The process of transfection involves complex interactions between the transfection reagent, the nucleic acid, and the cell membrane. Lipofectamine 3000 forms complexes with plasmid DNA, facilitating its entry into cells. However, as the plasmid size increases, the formation of these complexes might be less efficient, leading to reduced transfection efficiency. Additionally, larger plasmids may be more susceptible to shear forces during the preparation and handling processes, potentially compromising their integrity and further affecting transfection outcomes.
  3. Optimization for Larger Plasmids: When working with larger plasmids, it may be necessary to optimize transfection conditions. This optimization can include adjusting the ratio of Lipofectamine 3000 to DNA, increasing the amount of DNA or reagent, or altering the incubation times. Empirical testing is often required to determine the optimal conditions for each specific plasmid and cell type combination.
  4. Considerations for Experimental Design: For researchers, acknowledging the impact of plasmid size on transfection efficiency is crucial for experimental design. It is advisable to consider the use of smaller plasmids when feasible, or to ensure that transfection conditions are optimized for larger plasmids to achieve desired levels of gene expression.
In summary, the size of the plasmid is an important factor that can influence the efficiency of Lipofectamine 3000-based transfection. Smaller plasmids generally exhibit higher transfection efficiencies, but with careful optimization, successful transfection of larger plasmids can also be achieved. It underscores the importance of considering plasmid size in the planning and execution of transfection experiments to ensure the reliability and reproducibility of the results.
I trust this detailed explanation sheds light on the complexities of plasmid size in relation to Lipofectamine 3000 transfection efficiency and assists in guiding your future experimental approaches.
Best regards.
This protocol list might provide further insights to address this issue.
  • asked a question related to Transfection
Question
4 answers
My IPSC clones (not estbished lines) are fargile that when i transfect them with Cas9 palsmids - the cells die. I have used lipofectamine, and electroporation method for transfection. But cells dont survive.
what should i do different?
Relevant answer
Answer
You can transfect iPSCs with RNPs using a gentle lipofectamine transfection reagent. By this, the transfection efficiency will increase up to >97%. The next step is to single-cell sort the CRISPRed iPSCs on a 96-well plate in an enriched environment using Stemflex medium, cloneR supplement, and laminin-521. Using this combination you can achieve up to 70% clonal survival rate while ensuring they grow as homogenous clones. I discussed this in detail in a methodology paper here:
Contact me if you have further questions about the method.
  • asked a question related to Transfection
Question
3 answers
I‘ve transfected K562 cells by electroporation on Biorad Gene Pulser Xcell, the conditions was: 155V, 1000μF, 56.8ms, 0.2 cm Cuvette Gap, 106 cells in 100μL Entranster-E, 2μg plasmid
The transfection rate was around 30%, how can I improve it?
Relevant answer
Answer
To improve the transfection efficiency of K562 cells via electroporation, you can consider the following strategies:
  1. Optimization of electroporation parameters:Vary the voltage, capacitance, and pulse length: Try different combinations of voltage, capacitance, and pulse length to find the optimal conditions for your specific cell type and plasmid. You can perform a parameter optimization experiment by testing a range of values for each parameter. Adjust the cuvette gap: Altering the cuvette gap can influence the electric field strength experienced by the cells during electroporation. Experiment with different cuvette gap sizes to determine the most effective configuration for your cells.
  2. Optimization of cell and DNA concentration:Titrate cell number: Test different cell densities to identify the optimal number of cells for transfection. Too many cells can lead to increased cell-cell impedance and reduced transfection efficiency, while too few cells may result in lower overall transfection rates. Titrate DNA concentration: Determine the minimum amount of plasmid DNA required for efficient transfection. Lowering the DNA concentration can help reduce cellular stress and improve transfection efficiency.
  3. Optimization of transfection reagent:Explore alternative transfection reagents: Consider testing different transfection reagents optimized for electroporation in suspension cells like K562. Some reagents may exhibit better compatibility and efficiency with specific cell types. Evaluate different incubation conditions: Experiment with varying the duration and temperature of incubation after electroporation to optimize the interaction between the transfection reagent, cells, and DNA.
  4. Cell preparation and handling:Ensure cells are in optimal condition: Maintain cells in a healthy and actively proliferating state prior to electroporation. Check for cell viability, passage number, and culture conditions to minimize stress and improve transfection efficiency. Handle cells gently: Avoid excessive centrifugation, pipetting, and agitation during cell preparation and handling, as these can affect cell viability and transfection efficiency.
  5. Consider additional techniques:Explore alternative transfection methods: In addition to electroporation, consider other transfection techniques such as nucleofection or viral transduction, which may offer higher efficiency for certain cell types. Use reporter assays: Incorporate reporter genes (e.g., GFP) into your plasmid construct to easily visualize and quantify transfection efficiency by flow cytometry or fluorescence microscopy.
By systematically optimizing these parameters and experimental conditions, you can enhance the transfection efficiency of K562 cells via electroporation. It may require iterative testing and adjustments to achieve optimal results.
  • asked a question related to Transfection
Question
3 answers
We have been trying to produce lentivirus with the Tet inducible system for a while, but we are getting no signal in the infected cells after infection. However, after transfection, there is a higher signal in the cells. Has anyone had this problem, or has any explanation for this situation?
Relevant answer
Answer
Thanks for the information.
A few possibilities on why you are not seeing the signal after transduction:
1. The titer of your lenti is very low therefore very few/if any cells are being infected. Have you titered the lenti?
2. You are not using a high enough MOI. Typically transducing human cells requires an MOI of 5 to achieve >95% transduction efficiency.
3. You are attempting to induce expression with tet too soon after transduction. It takes a few days for the lenti to integrate into the genome. For instance, if you had mCherry under a CMV promotor, you would not see expression until about 4 days post transduction.
Regarding the transfection, since you are transfecting a plasmid you would see the signal much sooner since there is no integration into the genome.
  • asked a question related to Transfection
Question
6 answers
Does anyone work with expression in S2 cells? How efficient is the transfection using Lipofectamine 2000? Everytime I try to transfect using CaCl2 I get a cloudy media, and it seems that the culture is infected with a small bacteria, even filtrating all reagents with a 0.1 filter.
Relevant answer
Answer
The S2 kit comes with reagent for CaCl2 transfection. However, I used Cellfectin2 to transfect S2 cells and the efficiency is very high.
  • asked a question related to Transfection
Question
2 answers
I am currently doing a final year project on electroplating RNP complexes into Kasumi 1 cells - I have researched all the voltages published in papers and they range from 7500 kV to 90V - I am unsure as I have a 0.1 cm electroporation width gap and a BTX Harvard ECM electroporation and most papers have used the Neon Transfection System.
Relevant answer
Answer
Narges Zargar-Balajam Thank you so much for your recommendation!
  • asked a question related to Transfection
Question
1 answer
I transfected pcDNA and Eprotein in 293T cells, after 48 hours the supernatant was collected and exosomes were isolated, after lysing the exosomes
, an IP was performed on the CD1d, and the final membrane that was revealed was like this picture I put, it was very unclear and uneven, can anyone tell me why this is?
I've repeated this several times and the bands are this uneven, but the variety in the whole cell is very uniform and nice looking. I'm so confused.
Relevant answer
Answer
It is kind of tricky to troubleshoot this issue based on the limited information provided. You may want to label the lanes, such as "input", "IP", "IgG control", "exosome marker", and "Positive control", so people can understand the readout better.
Not sure if you use the homemade gels or purchased ones, but the rule of thumb is the pH of the gel may dramatically affect the protein electrophoresis. In addition, you may want to make sure the buffer is not too hot during the electrophoresis and transfer.
Good luck.
  • asked a question related to Transfection
Question
3 answers
We generated two Crispr KO cell line by PlentiCRISPR Puro and transfection of pSpcas9-GFP plasmid. After s.c. injection of cells on BALB/c immunecompetent mice, I successfully generated the formative tumor as expected at day 7. However, to my surprise, the tumor seems to disappear between days 10-14, no matter MOCK or KO cell line.
Does anybody have any suggestion or explanation?
Relevant answer
Answer
I would need to know what genes you had knocked out to make the cell line tumorigenic. It might be possible your cell line hit the Hayflick limit.
  • asked a question related to Transfection
Question
1 answer
I'm wondering if anyone has had similar experiences and/or solutions for the problems I've encountered when trying to generate stable Hepa1-6 reporter cells. I have GFP- and RFP-tagged plasmids in pCDNA6 myc/his B backbone which I'm planning to stably express into Hepa1-6 cells. The transfection efficiency and fluorescence intensity were great. I reseeded the cells 24h post-transfection and started blasticidin (10ug/ml) selection 48h post-transfection. Around Day 4-5 after blasticidin treatment most of the non-transfected Hepa1-6 cells died. The transfected cells grew well however none of the cells were fluorescent. The fluorescence intensities were already lower at Day 2-3 of blasticidin treatment. Seems the cells I have now are still blasticidin resistant but all my GOIs are lost. I've made stable lines in other cells using pCDNA plasmids. The genome integration efficiencies may vary but I was always able to get stable expression. This is the first time I use Hepa1-6 cells though. Would putting my GOI and the antibiotics under the same promoter using P2A or IRES help? Or Hepa1-6 is just not suitable for stable line generation?
Thank you very much,
Grace
Relevant answer
Answer
I would recommend using lentivirus for this. Its pretty easy to get custom lentivirus with your GOI/insert and GFP/RFP reporters and a drug selection marker from LipExoGen. Not very expensive either. We just email them flr this kind of thing and usually they will get back to us in the same day.
  • asked a question related to Transfection
Question
6 answers
I am currently working with HT-22 cells by transfecting them with bacterial plasmids to perform IHC. However, we have not had much success with reaching a 50% transfection rate. I usually do about a 40 hour incubation post transfection. I can not allow the cells to incubate any further because they will die due to the lipofectamine. Last week we tried a 60 hour incubation post transfection to have a better transfection rate. Unfortunately, we did have more transfected cells but many dead cells as well which defeats the purpose of our transfection. The one thing that did help us get a higher transfection rate was adding more plasmid, lipofectamine and P3000 to our cells. Does anyone have suggestions on how to improve the transfection rate without exceeding the 40 hour incubation?
P.S. I have attached files related to the products I use for my transfection and the protocol.
Relevant answer
Answer
I would try other transfection vectors (TransIT, jetPei, ....) there is a lot of them, change the DNA/lipofectant ratio... but there is cells that never transfect well.... tried transduction with lentivirus ?....
  • asked a question related to Transfection
Question
2 answers
Dear all, I recently had trouble analyzing my data by Western blot. I'm using the cell line model and transfection performance to analyze the importance of my target protein to cellular signaling. When I transfected and visualized the protein location or cell vibration, migration, or proliferation, the results turned out as expectation. However, when I tried to extract the protein for western blot and RNA for q-PCR analysis, the data became inconsistent and the phosphor form was the same between all conditions. I had changed my sample buffer, lysis buffer also other buffers to make SDS-PAGE but the results were the same.
Could you please give me suggestions to solve the trouble?
Thank you so much and best regards.
Relevant answer
Answer
Maybe you are sampling in a late time point, and the phosporilation occurs at early stage, try to do a time lapse experiment, as early as posible 0.5-2 h
  • asked a question related to Transfection
Question
2 answers
I want to transfect neuronal cells with my gene of interest. The lab used AAV vectors containing hrGFP. Hence want to know how it is better than the other variants.
Relevant answer
Answer
hrGFP fluoresces brighter in mammalian cells and is less toxic than GFP/eGFP, making it easier to use for mammalian expression experiments. It has a higher extinction coefficient and is more resistant to pH induced conformational changes as well as to solvents, detergents, and proteases.
However, in the paper attached below, use of hrGFP instead of GFP did not result in a larger proportion of transfected cells expressing fluorescence for extended period of time after transfection. Therefore, hrGFP does not appear to offer a significant advantage over GFP in transfection experiments.
Best.
  • asked a question related to Transfection
Question
1 answer
If cells were transfected with a plasmid containing a gene of interesed (the resulting protein is intracellular) fused to GFP, would the GFP signal be detected by flow cytometry? Or can we only detect those proteins on the cell surface?
Relevant answer
Answer
Yes, the GFP signal can be detected by flow cytometry no matter if intracellular or on the surface.
  • asked a question related to Transfection
Question
1 answer
I know several others have struggled with this but maybe someone has some current insight? I am trying to transfect NIH3T3 ms fibroblasts. I have tried Lipofectamine 3000 and JetPrime and JetOptimus by Polyplus and achieved very low transfection efficiencies (at times ~1%). I have played around with cell denisty, serum starvation, DNA: reagent ratio etc. No real difference. What else could I try?
Has anyone tried using PEIs (Polyethylenimin) for fibroblast transfection? If yes, any specific derivative? Maybe you even have a protocol to share?
I could use all your help! Thanks in advance.
Relevant answer
Answer
Dear Stefanie
First, examine the efficacy of your DNA construct and transfection reagents in HEK cells. Let us know about the results.
  • asked a question related to Transfection
Question
4 answers
I run qPCR to titer the AAV ( which i got by transfection in 100mm dish). I make four dilution of my sample ( 12 well and triplicate). I take GFP plasmid as a standard and were diluted to 1ng/ul, accordingly, 8 dilution to 0.05ng/ul (24 well and triplicate). Now i got the Ct, Ct Mean, Ct SD, and quantity. I need to calculate the the quantity in picogram/well, picogram/ml, genome copy and genome copy per ml. kindly pls suggest me any way, how to calculate it. Thanks.
Relevant answer
Answer
Calculating the genome copy number of Adeno-Associated Virus (AAV) titer by quantitative PCR (qPCR) involves several key steps. This method is widely used due to its sensitivity and accuracy. Here is a detailed guide:
  1. Preparation of Standards: Begin by preparing a standard curve using a known quantity of the AAV vector. This standard is typically a plasmid containing the same sequence as the AAV genome that will be amplified in the qPCR assay.
  2. Serial Dilution of Standards: Perform serial dilutions of the standard to generate a range of concentrations. These dilutions will be used to create a standard curve that correlates the cycle threshold (Ct) values from the qPCR with the known quantities of the template.
  3. DNA Extraction from AAV Samples: Extract the AAV DNA from your samples. This step is crucial for removing inhibitors and ensuring the efficiency of the qPCR reaction.
  4. qPCR Setup: Set up the qPCR reactions for both the standard dilutions and your AAV samples. Use primers specific to a sequence within the AAV genome. Ensure all reactions are performed in replicates to increase the reliability of the results.
  5. Running the qPCR: Perform the qPCR assay following the standard protocols of the qPCR machine. Carefully monitor the amplification and melting curves to ensure the specificity of the reaction.
  6. Analyzing the Standard Curve: After the qPCR run, analyze the standard curve. Plot the Ct values against the log of the known quantities of the standard. The resulting curve should be linear, and you can use it to calculate the efficiency of the PCR reaction.
  7. Calculating the AAV Genome Copies: Use the standard curve to determine the genome copy number in the AAV samples. For each sample, find the corresponding quantity of AAV genomes from its Ct value using the standard curve equation.
  8. Normalization: Normalize the calculated genome copy numbers to the volume of the AAV sample used in the DNA extraction to get the genome copies per milliliter.
  9. Quality Control: Ensure quality control by including no-template controls (NTCs) and positive controls in your qPCR run. NTCs should show no amplification, while the positive controls should align with expected Ct values.
  10. Data Interpretation: Analyze the data in the context of your experimental design. Take into consideration any potential factors that might affect the accuracy, such as PCR inhibitors or variations in DNA extraction efficiency.
  11. Replication for Accuracy: Repeat the assay for each sample multiple times to ensure accuracy and reliability of the results.
By carefully following these steps, you can accurately determine the genome copy number of AAV in your samples using qPCR. This method is highly sensitive and allows for precise quantification of the viral titer, which is crucial for many applications in gene therapy and virology research.
This protocol list might provide further insights to address this issue.
  • asked a question related to Transfection
Question
4 answers
Here is my problem: The day before transfection, I seed the HEK293FT cells into 100mm dish(cell density 4*10^6 cells), the next day before I preform the transfection, cell confluency like 90%, and I change the medium (warm the medium before). When I did that, the cells did not detach immdiately, but after I move the dish into incubator, and wait for the plasmid package, the cells become detach from the edge and almost the whole cell layer become detach. My concern is can I re-seed these cells by pipette? Can these work? Will it effect the transfection efficiency or the lentivirus production?
Relevant answer
Answer
probably they will re adhere if you put them in a new plate at a lower density (and after separating the agregates by flushing up and down) but I am not sure that the transfected cells will reatach... to much work for an unsure result... HEK-FT are suppose to grow in suspension (F) and they adhere really poorly (you can improve adhesion by coating your plates with poly L lysine (or buy coated plate) but I would rather shift to HEK-T cells; they adhere much better on plastic (and even better on coated plates)
  • asked a question related to Transfection
Question
2 answers
I've been having issues with viral production for the last couple of months. I was able to successfully make lentiviruses using a 2nd generation system 3 months prior and have a few aliquots frozen, but have unable to make more viruses. We recently tested the frozen nonconentrated virus by transducing a leukemia cell line and our transduction efficiency is about 80-90%.
My current protocol is:
Day -1) Seeding 293T on a 10 cm2 plate.
Day 0) Check to see if 293T confluence is 70-90%.
Prepare transfection complex:
Dilute packaging (psPax2), envelope (pRD114a) and transfer plasmid at 1:1:1 molar ratio in optimem.
Dilute PEI at 3X DNA MW in optimem.
Incubate for 10 mins.
Aliquot the PEI into the diluted DNA mixture. Incubate for 20 mins.
Add PEI:DNA mixture to 293T.
Day 1) Change media with fresh DMEM + FBS.
Day 2+3) Harvest supernatent. My transfection efficiency is about~80% evident by my GFP/mcherry reporter gene.
When I attempted to transduce the same leukemic cell lines, I was unable to detect my fluorescent reporter. It seems like even though my transfection was successful, my 293T are just not packaging the virus. Its not our transduction method because we are able to transduce with our old virus stock just fine. I tried different envelopes, different transfer plasmids, different aliquots of the packaging plasmids, freshly thawed 293T, different incubators, and different FBS manufacturers (both HI and non-HI). I could try different base medias incase the DMEM lot is bad, but i'm not sure if thats the case. I don't think it's out PEI because our transfection has been working well.
Relevant answer
Answer
You could also try reaching iut to a company that specialize in lentivirus production. That could save a lot of time. We always use LipExoGen for any kind of lentivirus we need. They have the most variety and also work realky great :)
  • asked a question related to Transfection
Question
2 answers
Hello dear all, at the moment I have a plasmid exhibiting 2 luciferase signals to monitor 2 cell signaling pathways.
Since this Luciferase reporter was originally designed to create a stable cell line, the transfection efficiency can't be assessed yet and it is a subject of concern to me, do you know how I can attach to the current reporter plasmid a fluorescence signal that will detect which cells had the insertion of the plasmid in a fluorescence-based approach and select them more specifically post-transfection by flow cytometry?
Thanks in advance.
  • asked a question related to Transfection
Question
3 answers
Hi all,
I did a mock transfection using pGL3 vectors with different promoter inserts (plasmids range from 10 to 14kb). However, I have gotten unexpectedly low luminescence ratios (firefly luc / renilla luc) and I was wondering if it would be logically sound to transfect mols of DNA instead of a certain weight for all of them (eg. 200ng per well). Thoughts?
Thanks!
Relevant answer
Answer
No, I haven't. But it is logically sound.
I would just use ~40% more µg of the 14kb plasmid, compared to the 10kb version....
  • asked a question related to Transfection
Question
10 answers
I have cloned my genes in a CMV vector and transfected them in HEK293 cells. After harvesting the cells, the expression level at the transcriptional level is increased compared to the control well. However, the peptide sequence is not seen to be expressed as analyzed by mass spectrometry.
Can you please give an insight into the possible reason for the same?
Thank you.
Relevant answer
Answer
Hi,
some expression vectors also carry sequences for in vitro cell-free translations. If your vector does not have it, try different one with these additional pieces. It will allow you to have your peptide or, at least, will show that problem with expression (on any level) in original setting. Good luck.
  • asked a question related to Transfection
Question
1 answer
I am trying transfection of SiRNA of COPB2 to pancreatic cell lines in 96 well.
Bxpc-3 is difficult to transfect SiRNA following the manufacture's protocol (lipofectamine 3000). So, I have devised some protocol (extend the transfection duration 4hr→15hr, 3000 reagent ammount 0.15μl→0.3μl/ well, DNA amount 0.1ng→0.15ng /well). But after transfection, almost all cells are died.
Please advise me the technical tips of transfection.
Relevant answer
Answer
Hello,
An important and simple thing to try is to skip antibiotic when transfecting and/or the serum. In my cells, antibiotic-free condition made the biggest difference in survival but other colleagues have reported the serum or both.
If you have tested this in parallel with the other variables (time, lipofectamine reagent amount and siRNA amount), perhaps is best to change to a different transfection reagent, fugene is one of the best, but I usually use it for bigger DNA transfections, such as plasmids. For siRNAs I've used SAINT_siRNA transfection reagent, with 12h-24hour exposure and very little loss of viability and for me it worked better than lipofectamine 3000.
Ideally, at 40-50% confluency, if it's too high or too low it also interferes with viability and transfection efficiency. In my experience, optimizing using a 96 well plate is a bit tricky, and a larger format might help.
I would also recommend using a positive (like GAPDH) and negative/scramble control, to make sure the transfection efficiency is okay, control for off-target effects and to assess if perhaps the loss of viability is part of the knockdown of your target protein. In the last case, it's also important to then test with a different sequence of siRNA, to better control for off-target effects of targeted sirna.
Hope it helps ;)
  • asked a question related to Transfection
Question
2 answers
I am referring here predominantly to therapeutic CHO cell lines, although this trend does seem to be widespread. In the literature, often in the same study, electroporation is used to generate stable cell lines, whereas a chemical method such as PEI-mediated transfection is utilised for transient gene expression. This is true for industry and academia as far as i can tell. Reviews on mammalian transfection methodologies tend to argue that chemical methods are by far the most common, for a list of reasons that make it more advantageous. Does anyone know of any reason why people continue with electroporation for stable work? If I was to guess I would say that it is more efficient at DNA delivery and that the hit taken in cell viability is not so important, because stable cell line generation allows plenty of time for recovery and perhaps also because regulatory bodies might not be comfortable with potential lingering chemicals in formulated products. However, I cannot find any literature to support this. Any help would be greatly appreciated. 
Thanks in advance,
Joe
Relevant answer
Answer
Hope someone can answer this question. From a few pieces of literature I read, electroporation could transfect more linearized plasmid into cells in a short time, which indirectly increasing the DNA concentration in nuclear. Compared to this, chemical transfection needs 4-6 hours to intake those DNA. and the transfect efficiency of linearized DNA in chemical transfection is low. Also, you cannot transfect too much DNA by using chemical transfection, which may cause some immune resistance and then decrease the transfection efficiency. For transite transfection I don't know, both electroporation and chemical transfection could be used. I guess chemical transfectin kit is much more easier to approach, and the procedure is more easier to follow.
  • asked a question related to Transfection
Question
2 answers
I would like to ask a question about constructing stable cell lines.
If someone has the whole genome sequencing results of their overexpression stable cell line that would be really helpful. That would give us a clear solid example of what going on during the fregment integration steps.
  1. I would like to ask, when using vector transfection to construct stable cell lines, is gene recombination inclined to randomly insert the entire part of the transfected vector into one random position (I mean a whole block integrated into the genome, the target gene and the resistance gene will be integrated near by as they were in plasmid). Or is it inclined to random integration, that is, the target fragment and the resistance gene are integrated in different spots? In addition, in the final cell line obtained, how many copies of the fragment are integrated into the genome? (Because the results we often get like, final seed 30 clones that can survive under puromycin, but only 3-5 contain the target fragment, which seems to answer this question, that is, random fragments of random integration are high probability events in this case.)
  2. Online information has reported that, due to the LTR sequences on the lentiviral vector, during gene recombination, the whole sequence between LTR can be integrated into a specific sequence position of the genome, could someone help me to double confirm this information?
  3. About the role of resistance genes, could we understand it in this way? 1). In the overexpression period (2-7 days after transfection), screen out the clones that have not been successfully transfected; 2). In the integrated period (2-14 days after transfection), screen out the clones that have not successfully integrated the resistance gene. 3). After the stable cell line is constructed, maintain the purity of the single-cell clone. Am I right? I suppose that if random fragments of random integration theory is right. Then I would not expect that all the puromycin-resistant cells all have my target gene overexpressed.
  4. I saw a product sells on the takara website which are linearized resistance markers used for co-transfection with other overexpression vectors. I thought that linear resistance markers would increase gene integration efficiency, then it may indirectly increase the probability of simultaneously integrating the interested fragment and the resistance gene in the same cell(may in different spots of genomic). Compared to those methods that transfect vectors contains resistance gene, above linearized resistance markers co-transfection methods would have more 'positive' cell clones to survive(have both interested gene and resistant marker) , then increasing the possibility of getting those clones.
  5. In those easy transfect cells, such as HEK293 or CHO-K1, the transfection efficiency can easily reach more than 95%, so can we understand it in this way? Compared to co-transfecting overexpression plasmid along with a linearized resistance marker, to a vector containing both resistance marker and target fragments, for a single cell, the possibility to get both interested gene and resistant marker is almost the same, right? 95%*95%=90.25%
Thank you for your reading, and please let me know if I did not make my idea clear. It would be really helpful if you answered my question.
Best,
Le
Relevant answer
Answer
1. Depends on what is in the plasmid you are using. If your plasmid is using some transposons then you can get almost random integration. The copy number will depend on if it is a jumping or replicating transposon. Viral vector derived plasmids tend to integrate into specific regions (eg. Lentivirus derived ones) but can be multiple copy. If integrated using recombinases (like the CRE/Lox system) then insertion will be site specific and usually single copy depending on the number of landing sites and if the genome is diploid. Similarly plasmids that integrate by homologous recombination will have 1-2 copies. The part of the plasmid integrated is usually a specific cassette flanked by regions used for integration (eg. homology arms, recombination sites or ITRs). Most plasmids try to avoid backbone integration into the host genome as that is prone to gene silencing by methylation.
2. You are correct everything between the LTRs is integrated. You will find most plasmids of that variety have a cassette with your GOI and the selection marker that is flanked by the LTRs with the replication machinery on a separate helper plasmid.
3. Your GOI and selection marker should be integrated together. After transfection treat each clone as a separate cell line and maintain selection for a least a month. It is good to periodically screen for the marker as that will eliminate any clones that have had the integrated cassette silenced.
4. I would have to look the maps of those specific vectors.
5. Yes the probabilities are combinatorial to an extent though with enough different construct being co-transfected there would eventually be signs of toxicity.
  • asked a question related to Transfection
Question
2 answers
Hello. I plan to do a simple immunofluorescence procedure with HT-22 cells. I plan to transfect the cells and fix them. I do not believe I will have time to come in on the weekend. Is it possible to store my cells after fixation? If so, should I dehydrate them using ethanol? Are there any other recommendations? After fixing with PFA the next step is to wash and incubate with Triton 100x. In addition, will this disrupt my signal or my cells?? Thank you
Relevant answer
Answer
Hi,
You can fix your cells with PFA (room temperature) or ice-cold methanol (4 C) for 15 minutes, then rinse with PBS 2-3 times. Cells can be left in PBS in the fridge and this won't affect fluorescence. When you are ready you can resume the experiment starting with the permeabilization/blocking step (with triton and BSA).
Worked all the time for me
Hope it helps
  • asked a question related to Transfection
Question
2 answers
The expiration date of my PEI transfection reagent is out and I do not know how to check it. I only use siRNA in my work and I can't figure out what I'm doing wrong or what's wrong with PEi. Can I check the PEI somehow, for example, using DLS or chromatography or spectrophotometry?
Relevant answer
Answer
Choose a plasmid with fluorescence and test it on 293 cells. You can see if it is inactivated.
  • asked a question related to Transfection
Question
1 answer
I am supposed to do luciferase assay for HBV enhancers EnhI and EnhII. I am quite new to luciferase assay and never done on hands before. So, I have to test certain protein effect on HBV enhancers through Luciferase activity. If there is anyone familiar with that protocol. I am looking protocol or cell scheme with details on culture amount of cells (HepG2 in 12 well plate), Transfection details and other necessary details. Thank you.
  • asked a question related to Transfection
Question
1 answer
Hi everyone,
I need to transfect 5'ppp-dsRNA (invivoGen) to stimulate RIG-I pathway using Lipofectamine 2000 for Bone marrow-derived macrophage.
If you have experience with this, please share your protocol.
The company manual only mentions about DNA transfection or siRNA transfection, however, I need specific protocol for 5'ppp-dsRNA.
Thank you so much.
Relevant answer
Answer
You can use the same ratio as recommended by the manufacturer for siRNA. At the end of the day it is a matter of phosphodiester charged versus cationic lipids...so you should be OK. Best
  • asked a question related to Transfection
Question
6 answers
Hi all,
We have a lentiviral vector with a gene of interest that we would like to express. There is no requirement for stable cell line construction, so we will only be transfecting transiently. However, our most proliferative cell line is not HEK, and therefore I'm curious for transient purposes, if the lentivector could be expressed in other cell lines, e.g. CHO?
I see some other posts on RG, e.g. https://www.researchgate.net/post/Why_are_293_HEK_cell_used_as_viral_vector_production_cell_over_other_cell_lines_What_advantage_does_it_have. But again to our interest, we're not producing the viral particles.
Our viral vector consists of an LTR followed by a CMV promoter before our gene of interest. Would this suffice for transient transfection? Or would binding to LTR limit CMV binding in other cell lines? Thanks for any advice.
Relevant answer
Answer
Thanks so much Kajs. It's really an open-ended question and your insights are very valuable :)
  • asked a question related to Transfection
Question
3 answers
I'm doing T7E1 experiment to check CRISPR-Cas9 transfection efficiency. Although i see my PCR products in the true site on the agarose gel, i do not see any band when i do T7E1. It is as if no DNA was loaded on the gel (My T7E1 condition is for 1 hour at 37 C degree). What could be the reason for this?
Thank you very much for answers!
Relevant answer
Answer
Thank all of you very much for answers. I tried to optimize (amount of DNA, T7E1 concentration, incubation time) but i observed overdigestion. Our final decision is to send sanger sequencing.
  • asked a question related to Transfection
Question
2 answers
Hello! I have a question regarding transfection stability in MCF7 and IMR32. For my experiment I need to transfect them with a plasmid with my gene of interest and Kan/Neo-gene which confers resistance against G418, as a selection tool. And after 5-6 passages in G418-containing medium I need to co-cultivate them with some other intact cell lines (i.e. further usage of G418 on this stage is impossible).
So, my question is - how long cells can retain a plasmid after removal of selecting agent?
Relevant answer
Answer
I think it depends on more than one factor, including the characteristics of your cell line, your plasmid, the concentration of G418 that your cells are resistant to ...
Maybe if you can, try to cultivate the transfected cells with and without G418 (in parallel) for ~5 passages : you might see a difference if the cells lose the plasmid, if you can test for the presence/absence of your gene of interest after this culture.
If the cells don't lose the plasmid after 5 passages, maybe you can go up to 10 passages or more. It depends on how long you will need to co-cultivate them with the non-resistant cell lines.
I hope I could help a bit ! Good luck with your experiments.
  • asked a question related to Transfection
Question
4 answers
I still don't figure it out yet how exactly a virus particle is formed by adding three plasmids (transfer plasmid with transgene, packaging plasmid and envelope plasmid) in a cell by transfection?
How the plasmids connect to each other in the host cell so that the transfected cell produces virus particles?
In addition to that, how does this method improves the security? Why the virus is not able to reproduce again after the infection of the host cell? I thought it is necessary, so that more cells can be infected with the viral genome to integrate the transfer gene/ gene of interest (so that the cell is able to express it).
And is it even right that I achieve my gene of interest/ the expression of the transgen through lentiviruses? Through the stable integration in the host cell genome?
I would be very glad, if someone could describe to me the process with lentiviruses in a detailed but easy way. I would like to be able to fully understand the method.
Relevant answer
Answer
The plasmids do not "connect" to each other in the host cell, they're just co-expressed in the cell which means all the different components needed to make the lentiviral particle are expressed in the cell.
This improves safety because the genome of the lentiviral particle that is produced in this way will only contain the transgene from your transfer plasmid*, since that's the only plasmid containing LTRs which are required for the RNA genome to get packaged into the lentiviral particle. The lentiviral particle won't contain the envelope genes or packaging genes (which are on the other plasmids), and those genes are necessary for making the lentiviral particle. So the only time the lentiviral particle can be made is when you co-transfect all three of those plasmids. Once you isolate the lentivirus and then use it to infect other cells, it won't be able to replicate because it doesn't express the necessary genes.
And for your last question, it is completely normal to test overexpression of a gene by integrating copies into the genome with lentivirus. This is very common. But it's important to keep in mind that your gene of interest may behave differently because it will be expressed at different levels than endogenously.
*not completely true but I'm simplifying things.
  • asked a question related to Transfection
Question
1 answer
I'm going to transiently transfect primary pre-adipocytes with His-tagged plasmid DNA [pcDNA3.1(+)] containing ADIPOQ gene with a SNP to study the gene and protein expression by using FuGENE 4K transfection reagent.
I would like to ask what is the most appropriate protocol to use in order to determine the successful rate of the mentioned transfection?
I was planning to use Pro-DetectTM Rapid His Competitive Assay Kit from ThermoFisher (A38507) for transfection confirmation. However, I am wondering how do I select the best transfection dilution that give the best transfection rate using the competitive assay kit.
Is it possible for me to solely rely on the number of lines appearing on the strip?
(As the concentration increases, the number of test lines will decrease until all test lines disappear. The concentration of the His-tagged proteins is inversely related to the number of test lines appearing on the strip).
Detail of the competitive assay kit is provided in the attachment, and this is the product link:
https://www.thermofisher.com/order/catalog/product/A38508?ef_id=CjwKCAiAmZGrBhAnEiwAo9qHia_rdYYp1DiRAC8VMtmH3mj0MsVJCVwy2qfr7Q5teJ67iIw-otp2SRoCLfgQAvD_BwE:G:s&s_kwcid=AL!3652!3!384464758933!!!g!!!6538554939!82560538550&cid=bid_pca_wwr_r01_co_cp1359_pjt0000_bid00000_0se_gaw_dy_pur_con&gad_source=1&gclid=CjwKCAiAmZGrBhAnEiwAo9qHia_rdYYp1DiRAC8VMtmH3mj0MsVJCVwy2qfr7Q5teJ67iIw-otp2SRoCLfgQAvD_BwE.
Million thanks in advance for suggestions and generous support.
Relevant answer
Answer
I would recommend a Western Blot analysis first in order to determine the concentration of the His-tagged protein in your lysate. Use a His-tagged protein with known concentration (if availble) to determine (roughly) your expression level.
These lateral flow tests are less sensitive than Western Blots and if you are using a cell line that is not normally used for protein production, the yields might be too low to get a good signal. Especially, if you are trying to optimize transfection conditions where differences of e.g. 20-50% more in one condition could hardly be detected.
I have produced and used similar His-tag lateral flow tests in the past and compared them to the ones from Thermo Scientific. Small differences are not easy to detect using this principle plus it is protein dependent (sometimes the tag is not well accessible and the result might be misleading). Western Blot is not exactly quantitative either but definitely more sensitive and not so much dependent on the structure of your protein.
  • asked a question related to Transfection
Question
3 answers
Dear Sir,
I'm trying to generate lentivirus with Tet-pLKO-puro construct with several packaging systems, but neither of them worked at all.
1) pLP1 + pLP2 + and pLP/VSVG (Invitrogen) 2) psPAX2 + pMD2.G 3) pMDLg/pRRE + pRSV/REV + pMD2.G
The cell line I used: 293FT cells (Invitrogen)
Transfection reagent: PEI-max
When generating lentivirus with pLKO-puro or pLenti-puro construct, the lentivirus particle with high titer is obtained.
Does anyone know troubleshooting regarding this?
In previous post, I found several similar questions but precise answer has not posted yet.
Help me, please.
Relevant answer
Answer
Satoru Sasagawa Which reagent worked for you, please?
  • asked a question related to Transfection
Question
3 answers
Hello,
I am transfecting linear DNA along with an Adenovirus transduction to HEK cells and need to isolate the DNA from both the linearised plasmid and viral genome at different passages for restriction digest analysis. I am not interested in the nuclear DNA
Total DNA extraction will include all genomic DNA which I fear will interfere with the restriction digest and produce a highly visible smear on the agarose gel. Ideally, I would want to just isolate cytoplasmic DNA.
Will it be okay to use the total DNA? Could I isolate extrachromosomal DNA using a standard miniprep kit, although they are meant for bacteria? Or would it be better to perform cytosolic isolation followed by DNA analysis?
Relevant answer
Answer
Given the low quantities of linearized DNA that will be present you definitely need some form of enrichment protocol. A miniprep kit might work.
  • asked a question related to Transfection
Question
4 answers
transfection, lipofectamine 2000
Relevant answer
Answer
Hello Zohreh Haghri,
I would recommend the use of optiMEM which is a media with reduced serum to dilute lipofectamine and DNA before complex formation.
OptiMEM allows for reduction in serum by 50% with no change in growth rate and or morphology. So, OptiMEM would be an appropriate medium when using lipofectamine during transfection.
However, you cannot keep your cells in OptiMEM for longer duration. Preferably 5-6 hours in OptiMEM would be appropriate.
Best.
  • asked a question related to Transfection
Question
3 answers
For a project we needed to use a specific device (Bio-Rad Gene pulser) to electroporate mammalian cells. We set up the instrument with the following settings:
Voltage = 1.9kV (we had to use this voltage for a particular reason)
Capacitance = 25 uF
Resistance = 200 ohm
And proceeded with electroporating cells (COS7 and CHO) in 0.4 cm chambers (Bio-rad) in PBS (800 uL). Interestingly, with different cell lines and cell densities, we had identical time constants (0.8 ms). We were shooting for a relatively low time constant as I read in another RG post (<1 ms). But I would have thought this should vary to the specific experimental condition (volume, cell type and density, etc.). And so I am wondering if the experiment should proceed as such, or something went wrong and our cells were not electroporated?
I also tested just discharging the voltage in air (no cuvette) and the time constant was a bit higher (~1.8 ms, but also a constant if repeated).
We're just beginning with this instrument so hope to get some useful advice. I will know whether the cells are successfully transfected after a few days when I purify the protein, but any suggestions would be helpful. Thank you.
Relevant answer
Answer
Thank you for sharing your insights!
  • asked a question related to Transfection
Question
4 answers
is this contamination post transfection using DreamFect Gold ?
the transfection method is lipid based method
picture is attached i am concern about the one in the yellow box
Relevant answer
Answer
Mohamed Khashan بتتكلم عربي؟
كيف اعرف ان الخلايا healthy?
لانه في شخص قلي ان الخلايا غير سعيده بس شاف الصوره
  • asked a question related to Transfection
Question
3 answers
I want to stably transfect Min6 cell lines with ciruclar plasmid . Kindly suggest which method is the most efficient.
Relevant answer
Answer
Hello, I also want to transfect the Min6 cell. Could you please tell me the transfection efficiency of Min6 cell?
  • asked a question related to Transfection
Question
1 answer
I am trying to use MC-easy kit to generate a GFP expressing minicircle. The gel analysis suggests that the quality of my minicircle should be good. However, when I try to transfect cells with the minicircle, my transfection efficiency is pretty low (compare to the full-length control). Am I missing something from the gel analysis? Does anyone have suggestions for directions for troubleshooting?
More information about the experiment:
  • Transfection method: electroporation and lipofectamine 2000 give similar results
  • Cell line tested: HCT116 and HEK293
  • 1ug of full-length plasmid and 1:1 molar ratio of minicircle (~272ng) has been used for transfection.
  • DNase treatment is for minicircle purification which is to remove the parental plasmid and the plasmid backbone generated from minicircle production.
Relevant answer
Answer
Have you uncovered the reason for lower transfection rate? We are currently observing similar results for our minicircles generated with the SBI protocol.
  • asked a question related to Transfection
Question
5 answers
is this contamination post transfection using DreamFect Gold ?
the transfection method is lipid based method
picture is attached i am concern about the one in the yellow box
Relevant answer
Answer
Massar Alsamraae ممكن اتواصل معاك
انا جديده بالشغل بالخلايا كيف اعرف ان الخلايا كويسه وسعيده؟
  • asked a question related to Transfection
Question
2 answers
I have a question about negative control of a transfection experiment using siRNA in cells. the expression of the target gene for knockdown is decreased in the negative control (Thermo). What is the cause? The positive control is knocking down without any problem.
Relevant answer
Answer
Hi Simon. Thank you for your answer.
Confirmed. Target gene expression is down regulated in cells transfected with negative control compared to non-transfected native cells. GAPDH expression is down regulated in positive control, while no down-regulation of GAPDH is observed in native cells. Considering the possibility of off-targeting of negative control, I used two types of negatvie control, but both show decreased expression of target genes. What does this mean?
  • asked a question related to Transfection
Question
4 answers
I am performing siRNA transfection to inhibit a certain matrix RNA. Does it make sense in this case to determine the reduction of this RNA by reverse transcription on PCR?
Relevant answer
Answer
I haven't used a scramble siRNA but i used 2 housekeeping genes at the same time (GAPDH & ActinB)
  • asked a question related to Transfection
Question
4 answers
please refer to me the protocol of insertion of PXR gene in existing DNA which has to be transfected in HEK293 WT cell line
Relevant answer
Answer
Priyansha Singh, inserting a specific gene, such as the PXR gene, into existing DNA involves recombinant DNA technology. The specific method used can vary depending on the application, the host organism, and the research goals.
  1. Isolate the PXR Gene: Obtain the PXR gene by isolating it from an existing organism through polymerase chain reaction (PCR).
  2. Prepare the Vector: need a DNA vector, such as a plasmid, to carry the PXR gene. You may need to cut the plasmid with a restriction enzyme to create a "cut" site for the PXR gene.
  3. Ligate the Gene and Vector: The PXR gene and the vector are joined together using DNA ligase, creating a recombinant plasmid. This plasmid now contains the PXR gene.
  4. Transformation: The recombinant plasmid is introduced into the host organism through transformation. For bacteria, involves heat shocking the cells to make them more permeable to DNA. For other organisms, different methods like electroporation or viral vectors may be used.
  5. Selection and Screening: Not all host cells will take up the recombinant plasmid hence a selection marker (like an antibiotic resistance gene) often includes the plasmid. Only cells that successfully incorporate the plasmid will survive when exposed to the selection agent. This allows you to screen for and identify the host cells that now contain the PXR gene.
  6. Culturing and Expression: The selected host cells are cultured to allow the PXR gene to be expressed. This means the host organism will now produce the protein encoded by the PXR gene.
  7. Verification: To confirm that the PXR gene has been successfully integrated into the host DNA, apply PCR, DNA sequencing.
  • asked a question related to Transfection
Question
4 answers
Hi. I'm trying to transfect CAL27 cell line (squamous cells of the tongue) with an overexpressing plasmid (size 7.7 Kbp) in different concentrations (from 50 to 1000 ng) using different volumes of TransIT Reagent by Mirus for different times, with no results (while I had no problems with siRNAs), besides partial death (same death with the empty plasmid).
Do you have any experiences with cells hard to transfect with plasmids? Do you have any suggestions for me?
(Plasmid works very good in HEK293T)
Thank you in advance for your attention and answer.
Relevant answer
Answer
you should try a different transfection reagent. I do not see the point why a 7.7 kbp Plasmid is hard to transfect. I assume that you have used simple CaPO4 transfection for the Hek cells? Try to get Lipofectin 2000 or 3000. You might ask a colleague from a different department or your sales representative?
According to my quick literature search you might be even able to transfect them with the CaPO4 method. You should use an empty GFP coding plasmid a positive control.
Best wishes
Soenke
  • asked a question related to Transfection
Question
6 answers
I have worked with HEK293 FT cells for many years. Recently, they have mysteriously started to look abnormal (see attached). After being passaged and grown in complete DMEM media (10% FBS, Pen/Strep, L-Glut, Geneticin) for ~3 days, instead of being fully confluent, they become rather elongated, skinny and sparse with the media colour remained mostly red. When they were left to grow for another day without changing media, they started to die off. However, if I passaged them, they would grow normally for 2 days. Despite this issue, when cells were seeded and transfected, they expressed okay. Myoplasm was tested negative. I have tried cleaning the incubator, made up fresh media and recovered a new vial of cells but the issue remains unresolved. Could unstable temperature or low CO2 percentage in the incubator cause this kind of cell growth?
Relevant answer
Answer
Update: tested in 3 different incubators but the problem remained. However, I suspect the cells might have been adversely affected and thus not recoverable/reversible. New cells have been brought up.
  • asked a question related to Transfection
Question
1 answer
I'm encountering challenges with the health of my MOLT-4 cell line, even after culturing them at 2x10^5 cells/ml in RPMI 1640 (containing 25 mM HEPES and L-glutamine) supplemented with 10% FBS. Are there any additional supplements I should consider to enhance their growth?
Furthermore, has anyone employed the MOLT-4 cell line for lentivirus transfection and can offer insights or recommendations?
Thanks
Samah
Relevant answer
Answer
Recommended culture conditions:
RPMI 1640 + 2mM Glutamine + 10% Foetal Bovine Serum (FBS). To help establish a culture the concentration of FBS can be increased to 20%. Once a growing culture is established the FBS concentration can be reduced to 10%.
  • asked a question related to Transfection
Question
2 answers
I am having trouble overexpressing using a pCDH lentivector with our current plasmids vsv-g and psPAX2. Transfection into HEKs seems to be working fine as I'm getting RFP expression, but I'm not getting transduction into my target cells (also transduction into HEKs isn't working). Should I be using different packaging and envelope vectors? The protocol from the supplier suggests a mix of pPACKH1-gag, pPACKH1-rev and vsv-g, but they only supply as a ready mix of these, so I'd like to know if these are really necessary.
Relevant answer
Answer
Hi Ross,
I have a similar problem. Did you solve the problem finally?
  • asked a question related to Transfection
Question
4 answers
Dear All,
Good day
I trypsinized HEK 293 FT today and split them into two flasks but after loading the needed amount we noticed that cells are not separated well and some are in clumps, would that affect their viability and health in the molecular stage?
NOTE,
I don't need them for immediate experiments, just to maintain them until I decide on the transfection day!!
image attached
Thanks
Relevant answer
Answer
Dear Khawla,
Usually Cell clumps after trypsinization is due to poor disassembly while pipeting them after you resuspend in the new culture media. After centrifugation and resuspension of the pellet, you can always pipet up and down, from 10 to 15 times for example, to guarantee that you will get nice single cells/lower amount of clumps.
Regarding viability, it really depends on cell nature, but if they have space to grow, it should not be a problem. Just avoid confluencies higher than 80%, and next time you tripsynize, do it more carefully.
Best regards,
  • asked a question related to Transfection
Question
1 answer
Hi everyone. Does anyone try to transfect human astrocytes in culture (no primary) to introduce siRNA in order to silence gene expression? Which one would be the best option do do it effectively? Thank yoy
Relevant answer
Answer
If you already have the siRNAs and what you are looking for is only the method to transfect the cells I would suggest using Lipofectamine RNAiMAX. It has always worked perfectly for me with different cell types and I managed to achieve really good silencing of genes.
If you also need the siRNAs I would suggest to go with siPOOLs https://www.sitoolsbiotech.com/sipools.php. I have also had a really good experience with them.
  • asked a question related to Transfection
Question
1 answer
I tried transfecting HAP1 cells with electroporation programme by Lonza (X005,Y007,X007 and X001). Our plasmid size is 8.5 kb. Positive control plasmid eGFP is around 3.5 kb and that shows good transfection efficiency around 80 %. However, incase of our plasmid the transfection efficiency is very low just three to four cells. I tried chemical transfection using Lipofectamine 2000, XtremeGene9, Genjet Still the transfection efficiency is very low Just three to four cells. has anyone transfected HAP1 cells with larger size plasmid around 8.5 kb.
Relevant answer
Answer
hello
Maybe you need to deal with amount of DNA and lipofectamine volume. Generally we use 1 DNA (µg) for 2 lipo (µL) but you can change and use 1:3 or mor. Because lipofectamine "create" drops with DNA so you need to recovert better the DNA and use more lipo. Be careful, more lipo can kill more cells.
If you have always problem, you can try with JetPrime. Its less rude for cells and you dont need to use Opti MEM. Moreover, SVF in opti MEM inactivate transfection by lipofectamine (i dont no if you do it like this)
Hope that can help you
  • asked a question related to Transfection
Question
1 answer
Hello! Can you give me some tips how to do CRISPR-Cas9 more effectively? Maybe you can give me some advice on this topic. This technology is new for me, I tried to conduct the experiments to knock-out genes, but they weren't successful. How do you detect knock-outs? What is the best combination of conditions to carry out CRISPR-Cas9?
Relevant answer
Answer
Use two gRNAs to create larger deletions. Consider knocking in a cassette with a selection marker. If here is a big deletion you can detect the knockout by PCR.
Also transfection is the generic process of getting DNA into cells and independent of the efficacy of your CRISPR. To improve transfection consider having a DNA nuclear targeting sequence on your plasmid.
  • asked a question related to Transfection
Question
1 answer
I have used miRNA-mimic transfect the HUVEC cell. and than RIPA lysis buffer to extract the cell protein. BUT the western blotting result is interesting. the mimic transfect cell express the lamin-B1, but the control group don't.
AS our previous study, the miRNA will induce the cell sencence. I don't know why.
primary antibody
lamin-B1 #365962
Relevant answer
Answer
Could it be that cells with different ages express different levels of laminin-1? On this paper DOI: 10.1002/aja.1002030404, I have found that "Different temporal patterns of laminin alpha 1, beta 1, and gamma 1 subunit chain expression were observed" ...
  • asked a question related to Transfection
Question
3 answers
Hello! I'm trying to create cell lines of HepG2 with separate knock-outs of mirna 101, 93, 30. I need to separately lower the level of mature miR 101, 93 and 30 in my cells. For this purpose I use CRISPR-Cas system utilising CRISPRMAX, TrueCut HiFi Cas9 protein and gRNAs. After transfection I conduct PCR from these genes and then cut these fragments with mismatch endonuclease I by BioLabs. I supposed to get smaller fragments after restriction, however I get only larger fragments in my agarose gel (they are above fragments on which restriction wasn't performed) . Does it mean that my transfection is unsuccessful? And how can I improve the result of my transfection? Last time I plated cells only the day before conducting the transfection and the cell density was approximately 40-50%.
Relevant answer
Answer
If you preform your PCR on untreated cells do you get the correct band size? If not it may be an issue of your primers not amplifying the correct locus. Do you know the copy number of your region of interest in HepG2 many cell lines have more than 2 copies of each chromosome making it harder to get a complete knockout.
  • asked a question related to Transfection
Question
1 answer
I am trying to transfect HEK293 cells with dCas9-KRAB-DNMT3A carrying plasmid and multiple IVT sgRNAs. RNAi max invitrogen and Hiperfect Qiagen are only good for small RNAs but not plasmids. Does anyone have experience with a reagent that allows uptake of both plasmid and RNA?
Relevant answer
Answer
Transfecting HEK293T cells with in vitro transcribed (IVT) sgRNA and a plasmid carrying a dCas9 backbone requires an efficient transfection reagent to ensure successful delivery of the genetic material into the cells. The choice of transfection reagent can significantly impact transfection efficiency and cell viability. Lipid-based transfection reagents are commonly used for this purpose. One of the most popular choices is Lipofectamine 2000, but there are other alternatives as well. Here are a few options:
  1. Lipofectamine 2000: Lipofectamine 2000 is a widely used cationic lipid-based transfection reagent that is known for its high transfection efficiency. It works well for a variety of cell types, including HEK293T cells. It's compatible with both DNA and RNA transfections.
  2. Lipofectamine CRISPRMAX: This is a specialized reagent from Thermo Fisher Scientific designed specifically for CRISPR/Cas9 applications, including delivering Cas9 plasmids and sgRNAs. It can be a good choice for this type of transfection.
  3. PEI (Polyethylenimine): Polyethylenimine is a cationic polymer-based transfection reagent. PEI can provide high transfection efficiency and is relatively cost-effective. However, it's essential to optimize the concentration and formulation to avoid cytotoxicity.
  4. JetPRIME: JetPRIME is another transfection reagent that works well for DNA and RNA transfections in HEK293T cells. It is known for its ease of use and good transfection efficiency.
  5. Fugene HD: Fugene HD is another cationic lipid-based transfection reagent with good transfection efficiency. It is compatible with a wide range of cell types, including HEK293T cells.
  6. Neon Transfection System: If you have access to electroporation equipment like the Thermo Fisher Neon Transfection System, you can consider electroporation as an alternative method. Electroporation can be very efficient for introducing genetic material into cells.
When choosing a transfection reagent, it's important to consider the specific requirements of your experiment, such as the cell type, the amount of genetic material, and the presence of any additives or modifications in your sgRNA or Cas9 plasmid. Additionally, perform transfection optimization experiments to determine the best conditions for your particular setup, as the ideal reagent and protocol may vary from one lab or experiment to another. Always follow the manufacturer's guidelines and recommended protocols for the transfection reagent you select.
Regenerate
  • asked a question related to Transfection
Question
3 answers
Currently, the localization study for my target protein because of another protein B has to be performed using confocal. It had been confirmed using Western Blot.
1. I wanted to know if it is necessary to perform, transfection after seeding the cells on the cover glass, followed by confocal microscopy, or perform the transfection in suppose a 12-well plate and then trypsinize it and seed the cells on the cover glass?
2. How much should be the concentration of siRNA or plasmid? Should it be the same as used in Western blot or less?
3. What must be the time of incubation after media change? 48 hours or 24 hours? Because when I seed around 10,000 cells on the cover glass and perform the transfection, cell clumping occurs after 48 hours on the cover glass.
Relevant answer
Answer
If you are validating a knock-down for another experiment, I would use the same lentivirus incubation times and concentrations for your ICC, WB, and other experiments.
As a side note, with my current lentivirus knockdown, I saw no significant depletion of protein at 48 and 72 hours post-transduction, but did see a significant depletion at 5 days.
Depending on the half-life of your protein of interest, you may want to leave it a bit longer following transduction.
All the best,
Sam
  • asked a question related to Transfection
Question
4 answers
Can anyone suggest a methodology for adsorbing mRNA onto hybrid lipid polymeric nanoparticles to achieve a monodisperse system capable of transfecting immune cells?
I am working with a 4:1 N:P ratio and have attempted to adsorb mCherry mRNA onto the nanoparticles using pipette mixing or vortex, either directly adding the mRNA concentrated solution (1 mg/ml) or by diluting the mRNA to match the volume of the nanoparticles. I have incubated the mRNA-LPN mixture for 2 hours at 4ºC or for 30 minutes to 1 hour at room temperature. However, in all cases, the nanoparticles significantly increased in diameter (from 200 nm to 700 nm) and polydispersity index (PDI above 0.3). Additionally, I have not observed any successful transfection rates with these trials. I am using PLGA nanoparticles, DOTAP, DOPE, and MC3 lipids, experimenting with different combinations and ratios, but none of them have yielded positive results.
If anyone is open to discussing this topic, I would be delighted to share and learn. I have read nearly all the papers on mRNA and lipid/PLGA nanoparticles but cannot identify where I am missing something, preventing me from achieving a stable system and successful transfection results.
Thank you very much.
Relevant answer
Answer
Developing an effective methodology for adsorbing mRNA onto hybrid lipid polymeric nanoparticles (LPNs) for transfecting immune cells is a complex task. Start by ensuring the quality and purity of all materials, including PLGA, DOTAP, DOPE, MC3 lipids, and mRNA. Prepare PLGA nanoparticles using a reliable method that yields monodisperse nanoparticles of the desired size (~200 nm). Optimize the formulation and parameters for nanoparticle synthesis to achieve consistent size and PDI. For mRNA preparation, ensure high-quality mRNA with minimal degradation.
When adsorbing mRNA onto nanoparticles, employ a controlled and gentle method to prevent size increase and PDI elevation. Experiment with different concentrations of mRNA and nanoparticles to determine the optimal ratio for adsorption (4:1 N:P ratio as you mentioned). Optimize incubation time and temperature for effective adsorption without causing aggregation. Regularly monitor nanoparticle size and PDI during the adsorption process, stopping if a significant increase is observed, and adjust conditions accordingly.
After adsorption, conduct transfection assays to assess the transfection efficiency of the mRNA-loaded LPNs in immune cells. Use appropriate controls and measure transfection using a suitable readout (e.g., flow cytometry, fluorescence microscopy). Continuously optimize the formulation, adsorption conditions, and transfection protocols based on the results obtained, making gradual changes and carefully monitoring effects on nanoparticle stability and transfection efficiency. Collaborate with experts in the field, attend conferences, and share your findings with the scientific community to gather feedback and insights that could help address any challenges you're facing. Additionally, conduct a thorough literature review to identify potential modifications or new approaches that may improve your nanoparticle-mRNA system. Discuss your challenges and findings with colleagues in your research group or other experts in the field, as they may provide valuable suggestions based on their experiences. Through meticulous optimization and seeking input from the scientific community, you can refine your methodology and achieve a stable, monodisperse nanoparticle-mRNA system capable of efficient transfection in immune cells.
  • asked a question related to Transfection
Question
2 answers
I want to co-transfection 3 DNAs with 3 different antibiotic resistance.
(three antibiotics : Hygromycin, Neomycin, blasticidin)
After transfection, is there no problem with adding three antibiotics (Hygromycin, Neomycin, blasticidin) to the media to grow cells and produce proteins?
because, I want all three DNAs to grow only transfected cells.
and I wonder if cells that are not antibiotic resistant give cell death signals to living cells as they die.
Relevant answer
Answer
The simultaneous application of three antibiotics may be tricky. I would suggest to first conduct kill curve/dose response experiments with each of the antibiotics on the non-transfected cells to determine the optimal/minimum concentration of each antibiotic, which will kill all cells over a specific period of time (e.g. 2-5 days). You might then need to apply all three antibiotics simultaneously to the non-transfected cells at the determined optimal concentrations to adapt the time for the actual selection experiment of the transfected cells.