Science method

Quantitative RT-PCR - Science method

Explore the latest questions and answers in Quantitative RT-PCR, and find Quantitative RT-PCR experts.
Questions related to Quantitative RT-PCR
  • asked a question related to Quantitative RT-PCR
Question
3 answers
I am analyzing the differential expression of splicing variants, through capillary electrophoresis, of a DNA repair gene after treatment with a DNA-damaging drug. I am using gene-specific primers for cDNA synthesis, which makes it challenging to identify a suitable internal control. Using another region of the same gene might also be affected by the drug, while choosing a housekeeping gene would require a separate cDNA synthesis step and introduce variability. What would be the most appropriate internal control to ensure accurate normalization in this setup?
Relevant answer
Answer
Why are you using capillary gel electrophoresis instead of qPCR? The detection limits of capillary tubes is very limited - easy to saturate & minimal signal for detection is also high.
You can use a 1-step qPCR for your splice variants and 1-step for a housekeeping gene.
  • asked a question related to Quantitative RT-PCR
Question
1 answer
Dear researchers,
I’m working on an Norovirus GI and GII qPCR assay with TaqMan probes. Recently, I experienced an issue with “multiplexing”. Briefly, the aim is to develop an assay for simultaneous detection of two targets (HuNoV GI and GII) using FAM and HEX reporters.
Single PCR, when the probes are used separately, works well. While in multiplex (when four primers, two probes are mixed per reaction), we get a low signal for one of the targets in several independent runs. On the other hand, in electrophoresis the DNA yield seems the same for different combinations. I tried to change probes concentrations and it seems that reducing probe amount for one target increases signal for the other target, but I guess that it may affect my results for competing target. I would appreciate any suggestion to optimize this assay, including good manulal about assay development. What to do in this kind of situation?
Thank you!
Technical details:
Concentrations of primers 400 nM, probes 200 nM, Mg 3 mM.
Cycling conditions: hot start at 95⁰C for 5 minutes, then 45 cycles of denaturation at 95⁰C for 15 seconds, annealing and elongation at 60⁰C for 1 minute.
Instrument: ABI StepOne Plus
Relevant answer
Answer
what kind of master mix did you use? Did you consider using a different master mix with a higher Mg concentration? What are the concentrations of the templates in multiplex format? Did you evaluate the effect of the concentration of each target on the amplification of the other? if the concentration of one target is high and the other is low, sometimes the high-concentration target hinders the amplification of the other.
though it might seem irrelevant, I have an experience I'd like to share. I was developing a multiplex qPCR assay to detect two viruses. I made a mixed serial dilution containing the two target standard plasmids, from 10^7 to 10^1. At low concentrations of one of the targets (10^3 to 10), the Cq values were equal. However, when I used separate standards and added each to the mix, the amplification at low concentrations was OK. I do not know what caused this.
Anyway, I think if you assess the effect of high concentration of one target on the amplification of the other, you might find something. Please let me know if you found the cause.
  • asked a question related to Quantitative RT-PCR
Question
7 answers
Hi, I recently did a thermal shift assay using the intrinsic fluorescence of tryptophan. Therefore, i use no dye whatsoever. I use Biorad CFX96 and use the available thermal shift assay protocol online. My problem is the high pre transition RFU. What could possibly cause this? I have centrifuged my samples to precipitate any possible aggregates. The y -axis is the fluorescence reading while the x axis is the temperature.
Relevant answer
Answer
Samuel Davis , 100% agree, if the machine doesn't work at wavelengths relevant to the intrinsic (trp) fluorescence, i.e. exc. 280--300nm, em. 330--360nm, then all other observations & troubleshooting steps are wasted.
In which case either change the protocol (external dyes), or the instrument; I've run thermal denaturation of proteins in a standard cuvette-based fluorescence spectrometer equipped with a temperature controlled sample holder. Mind you, that likely requires more sample and is quite slow.
  • asked a question related to Quantitative RT-PCR
Question
7 answers
Dear All,
mirna primer showing some problem in the melting curve? any idea why? As attached is the melting curve. The forward sequence is obtained from miRBase and reverse primer is universal.
Relevant answer
Answer
Hii Gayatri...I am facing the same issue with melting peaks in miRNA qPCR.....please help me if you could solve this issue.
  • asked a question related to Quantitative RT-PCR
Question
3 answers
I ask because biomolecular water or TE doesn’t maintain DNA integrity as expected.
Relevant answer
Answer
I'd add in that you should dilute & aliquot into "single use portions". That way you aren't doing repeated freeze-thaw cycles. And keep in a deep freeze or non-frost-free freezer.
  • asked a question related to Quantitative RT-PCR
Question
3 answers
Hello all,
I am working on a project of quantifying viral titer. I read some paper, and there are different units for viral titer. Among of them are TCID50 value, GC/ml and VP/ml. I do not understand much about the differences between them. Can anyone please give some explanation about this?
Relevant answer
Answer
Is there a formula to explain this relation between genome copy number and TCID50/ml. I did some AI search and it gave me this formula which I am not sure where it got from: Copy number/ml=TCID50/ml × Genome size (bp)​/Avogadro’s number (6.022 × 10²³).... is that right?
  • asked a question related to Quantitative RT-PCR
Question
5 answers
Hello, I've extracted RNA from A549 cells and I've done nanodrop to determin the RNA concentration. And then I meet the problem. I have no idea about the concentration and the quality of cDNA after doing reverse transcripataion.
I searched some information and know that I can make a stand curve to determin the concentration, but I don't have standard product.
I want to ask is there othre method to the concentration of cDNA with RT-qPCR. I have and idea about using internal control (like TBP), but I'm not very sure about how to connect the Ct value of TBP to the concentration of cDNA.
Hope someone give me advice!
THANK YOU
Relevant answer
Answer
The cDNA that you synthesized is for gene expression studies, so in such case, you should run comparative Ct protocol in RT-PCR for which you'll require house keeping genes as endogenous control. The Ct value of the endogenous control will be used to normalised your targetted gene, so no prior concentration of your cDNA is required.
Now if you want to analyze the yeild of your extracted RNA, you can run absolute quantification protocol using any known standards (now a days many standards are commercially available in some kits, like viral detection kit or NGS library quantification kits).
  • asked a question related to Quantitative RT-PCR
Question
3 answers
I am using three biological replicates. Their DNA concentrations measured by Qubit were different. I diluted all samples using an online calculator to ensure each sample had a concentration of 2.5 ng/μL. I used 2 μL of each sample in the qPCR, resulting in 5 ng/ul of DNA per well.
After performing qPCR, I noticed a slight difference in the Ct values of the biological replicates. I wondered why there was this difference. When I rechecked the DNA concentrations of my diluted biological replicates, I found slight variations i-e: 2.36 ng/μL, 1.98 ng/μL, 2.4 ng/μL, etc. This explained the observed differences in the Ct values during qPCR.
My question is: how can I ensure equal DNA concentrations in all samples?
Relevant answer
Answer
Measure the dna concentration well into the working range of the spectrometer....errors are large at low OD and high OD values.
In making dilutions use large volumes of dna.....the errors involved in pipetting low volumes ( less than 5ul) are very large.
Dilute the dna so that you use 5ul or even 10ul volumes added to the pcr reaction as once again the pipetting errors are greatest at small volumes. For instance pipetting 1ul could easily have an error of 10% using routine and seldom calibrated pipettes
  • asked a question related to Quantitative RT-PCR
Question
5 answers
Hi,
I found more monocytes in the heart in treated group accoring to the FACS data. And I did qpcr to validate this result.
First I checked CCL2 (or MCP-1) and other cytokines/chemokines because CCL2 plays an important role in monocytes recruitment in many research papers. But there's no significance among different groups.
Then I found the TGFb1 gene expression level significantly increased in treated group. But I couldn't find too much information about TGFb1 inducing the infiltrating of monocytes to the heart.
Does anyone have any ideas about TGFb1 and monocytes? And what else for monocytes recruitment?
Many thanks.
Relevant answer
Answer
Transforming growth factor beta 1 (TGFβ1) is a pleiotropic cytokine that plays a complex role in inflammation and immune cell regulation. While it is not typically characterized as a primary chemokine for monocyte recruitment, TGFβ1 can influence monocyte behavior and infiltration in several ways:
  1. Modulation of Monocyte Differentiation: TGFβ1 can promote the differentiation of monocytes into regulatory macrophages, which can have anti-inflammatory properties. However, the context and presence of other cytokines can lead to different outcomes.
  2. Induction of Chemokines: TGFβ1 can induce the expression of other chemokines and cytokines that are involved in monocyte recruitment. For example, it can upregulate the expression of CCL2 (MCP-1) indirectly, which is a known chemokine for monocyte recruitment.
  3. Regulation of Extracellular Matrix: TGFβ1 plays a key role in extracellular matrix (ECM) production and remodeling. Changes in the ECM can affect the migration of monocytes into tissues.
  4. Immune Suppression: TGFβ1 can suppress immune responses, which might create an environment where monocyte infiltration is less regulated.
Here are some considerations and additional factors that could be involved in monocyte recruitment to the heart:
  • Other Chemokines/Cytokines: Besides CCL2, consider looking at other chemokines such as CCL5 (RANTES), CCL7 (MCP-3), CCL8 (MCP-2), CXC chemokines like CXCL1 (GROα), and CXCL10 (IP-10), which can also be involved in monocyte recruitment.
  • Adhesion Molecules: Monocyte recruitment involves adhesion to the endothelium. Check the expression of adhesion molecules like VCAM-1, ICAM-1, and E-selectin, which are often upregulated in response to inflammatory signals and facilitate monocyte adhesion and transmigration.
  • Inflammatory Pathways: Look at other inflammatory pathways such as NF-κB, which can regulate the expression of various chemokines and adhesion molecules.
  • Cytokine Signaling Pathways: TGFβ1 can interact with other cytokines such as TNF-α and IL-1β. The combination of these cytokines can have synergistic effects on monocyte recruitment.
  • Cellular Cross-Talk: Consider the interaction between cardiac cells, endothelial cells, and immune cells. For example, cardiac myocytes can produce cytokines in response to stress, which can influence monocyte infiltration.
  • MicroRNA Regulation: MicroRNAs (miRNAs) can regulate the expression of cytokines and chemokines. Changes in miRNA expression could be contributing to monocyte recruitment.
  • Stromal Cells: Fibroblasts and other stromal cells in the heart can produce factors that influence monocyte recruitment.
To further explore the role of TGFβ1 in monocyte recruitment in your treated group, you could:
  • Perform in vitro experiments to see if TGFβ1 treatment directly affects monocyte chemotaxis.
  • Use neutralizing antibodies or inhibitors to block TGFβ1 signaling and observe changes in monocyte infiltration.
  • Investigate the signaling pathways downstream of TGFβ1 that might be involved in monocyte recruitment.
  • Look for correlations between TGFβ1 expression and other cytokines/chemokines that are known to be involved in monocyte recruitment.
Remember that monocyte recruitment is a complex process involving multiple factors and interactions. A holistic approach that considers the entire inflammatory and cellular environment will be key to understanding the mechanisms at play in your treated group.
  • asked a question related to Quantitative RT-PCR
Question
8 answers
Hi,
I have done 3 experiments (may be called biological replicates in this scenario) with the same cell line to measure gene expression of "XYZ" on days (D)4, 6, and 8.
For obtaining RNA, I have cultured cells in 6 wells (say replicates) for experiment-1. Pooled only 2 wells together at D4 and extracted RNA for qPCR analysis. qPCR experiment had three technical replicates. This way I got qPCR data for D4 for experiment-1. I did the same procedure for D6 and D8 to get qPCR data for D6 and D8 respectively for experiment-1.
In the same manner I repeated experiment-2 and experiment-3.
I wish to compare 3 time points D4 D6 D8 to see if increase or decrease of "gene A" expression is significant.
Is my data paired or unpaired? Could you please explain why so?
My assumption: My understanding is that the data is paired. Graphpad tests show some data sets to be distributed normally and some are not. Considering low sample number (n=3), I assume all the data to not follow gaussian distribution and therefore use non-parametric tests.
Can I compare with Friedman test with Dunn's post-hoc for XYZ at D4 D6 D8? Or should I use Wilcoxon testing two time points (like D4vsD6, D4vsD8 and D6vsD8)?
Relevant answer
Answer
Abhinav Kethiri, Gaussian and Normal distribution is the same. The model became famous after a description by Friedrich Gauss who derived it in the context of a system of normal-equations (a step to solve a system of linear equations). Therefor it's sometimes called the Gaussian and sometimes called the Normal distribution.
I know that GraphPad Prism offers "test for normality". However, these are almost useless for the purpose you used them. A hypothesis test checks if the observed data (the sample) already provides sufficient information about a particular aspect of a model. In the normal distribution test, the model is some continuous univariate distribution having a mean and a variance, and the aspect being tested is that the functional form of the distribution is Gaussian. This is a restriction of the more general model of "some continuous univariate distribution". The whole aim of the test is to reject this restriction, in which case one would have demonstrated that the observed data already provides sufficient information to "see" (clearly enough) that the hypothesized special case is not accounting for all aspects of the real world. If you are able for some sample to reject this hypothesis, you still don't know if the kind of deviation if of any relevance (irrelevant deviations will lead to rejection in large samples), and if you fail to reject it, also you don't have any use information, as the test may have overlooked an actually relevant deviation (relevant deviations will not lead to rejections in small samples).
  • asked a question related to Quantitative RT-PCR
Question
4 answers
Hi. I want to synthesize this blaTEM gene. This is the FASTA sequence, but I don't know which part is related to coding sequence, Can anyone help me?
(my major isn't related to microbiology or genetic, so I don't know the exact procedure of genetic basics,)
and one more thing, after synthesis how can I know how many copy numbers are there in my sample to generate qPCR standard curve based on that?
Relevant answer
Answer
The coding sequence start from the first "ATG" right after bacterial promoter sequence and end up at stop codon "TAA".
From 101 to 961.
  • asked a question related to Quantitative RT-PCR
Question
5 answers
Hello!
I am optimizing conditions of stem-loop RT-PCR to compare the expression of miRNAs in different cell lines.
Now I'm performing the reaction in one step: RT with stem-loop primer and subsequent real-time PCR with a pair of primers and SYBR Green - all in one reaction. So my reaction mix contains three types of primers, heat-inactivated reverse transcriptase, hot-start Taq polymerase, SYBR and all the nessesary components such as ddNTPs and Mg2+.
I use synthetic miRNA and total RNA from human cells as matrices.
When working with synthetic miRNA, everything goes well (Figures 1 and 2), amplification plot is ok and there is a single peak in the melting curve.
But with total RNA I observe a decline of fluorescence in the plateu phase of amplification (Figure 3) and 2 product peak in the melting curve (Figure 4).
Reference gene amplification plot doesn't have such issues.
What are the reasons of the decline of fluorescence after the 30th cycle?
What are the possible variants of the second product here? Primer-dimers observed in no-template control have Tm = 80 degrees C.
Relevant answer
Answer
I was a bit fast in my response... re-hybridization so to my knowledge the only known reason for hooking amplification curves - but this effect is understood only for hybridization probe formats. You seem to use SYBR Green I detection. Such effects have been observed for SYBR Green I detection, but the mechanisms here are not clear. It is hypothesized that photobleaching could play a role. However, the interesting finding in your case that the hooked curves occur together with a second amplicon, some triple-helix formation or the formation of other secondary structures during annealing may reduce the effective space for binding of SYBR Green molecules and the linear energy transfer trough the amplicon molecules, resulting in a lower net florescence.
Again, just a wild guess. It may help to have a look at a native and denaturing PAGE of the PCR products, or even at their sequences. If you are really interested in clarifying the mechanism here you might want to generate synthetic molecules of that sequence and analyze the net fluorescence and UV absorption for different mixtures.
For your qPCR of miRNAs, this effect is not relevant, as long as the wrong product is amplified late. You could stop the PCR a few cycles after the exponential phase and check a melt curve and gel there. If at this cycle there is only the correct product amplified, the Ct value is valid. Otherwise you will have to optimize the assay (vary MgCl2 and/or primer concentration; it sometimes helps just to use/test another brand [that often includes a different variant of the polymerase as well]).
  • asked a question related to Quantitative RT-PCR
Question
3 answers
I'm trying to detecte very small amounts of a mutant-type mtDNA using SYBR green qPCR. I can't really use a higher concentration of gDNA because my wild-type and housekeeping genes already have a low ct around 17. My negative controls show some primer dimers and are around a ct of 33. However, some samples show a specific melting curve peak identical to the one in my positive control, but with a Ct higher than my negative control (33,34,35) while some samples with the same Ct show a melting curve similar to the negative control, so all over the place or at the primer dimers temperature.
Should I consider aller samples with a higher Ct than my negative control as an absence of the target sequence or should I evaluate each melting curve separately and those showing a specific amplification peak are considered as containing the mutant-type, while others with not showing this peak are considered as homplasmic? If not, is there another way to rigorously discriminate my samples with absence/presence of my target gene?
Relevant answer
Answer
I mean, how much are the numbers really helping you here?
"Almost always either none, or below the level of quantification" is vaguely useful in the sense that it emphatically communicates "this target is extremely low abundance", but you can't actually put numbers on such low target levels. It's too low to quantify.
It's a PCR, though: it makes a product, and you can run that product on a gel. (this is effectively what the melt curve is pretending to do, really)
If you want to confirm that
A) it's definitely a real product, and
B) it's definitely your GOI
then take those positive and "primer dimer" samples and run them on a gel. If you see sharp single bands in the samples you deem to be positive, and diffuse primer dimer smears in the others, then: primer dimers vs specific amplicon confirmed.
If you then cut out the specific band and send it off for sequencing (cheapo sanger sequencing, using one of your primers) and it comes back with sequence that matches the expected amplicon/target, then "GOI successfully detected" is also confirmed.
Once you have that, you more or less have evidence that you can detect your target if it's there, but also, that even when present, it is present at vanishingly low levels.
I would sort of slightly question the chosen approach, though: if your housekeeping genes (which I assume are either genomic markers or other mtDNA markers) are giving Cqs of ~17, that's ~16-18 cycles earlier than your target of interest, i.e. ~60,000-240,000 greater abundance. That's...a lot, and by extension this implies that your target, even when present, is unlikely to be relevant.
What is your desired outcome measure for this experiment?
  • asked a question related to Quantitative RT-PCR
Question
3 answers
Hi, I am doing qPCR experiments. When the reaction finished, I obtain the Ct value of my samples is zero, but they have a peak with my expected Tm. Furthermore, I run the gel electrophoresis, the result also shows that these samples appear a band at my expected target size. But why the Ct value is zero?
Relevant answer
Answer
Look at the file containig information about the increase in the fluorescent signal as a function of Ct. Check how the Ct threshold is defined - it might be incorrectly positioned. Set it within the exponential phase of PCR.
  • asked a question related to Quantitative RT-PCR
Question
3 answers
Hi, I am designing primers to discriminate SNP alelle, which is 2 forward primer specific for each allele. To improved the discrimination of SNP, what is the acceptable delta G value of the primer?
Relevant answer
Answer
It's probably going to be cheaper and faster to PCR amplify and Sanger sequence to find SNPs.
Your assay is based on amplification from one primer (SNP) vs no amplification (other allele). And a second reaction where you get amplification from (other allele) and none from (SNP). Relying on negative results (e.g. not seeing amplification for homozygous for SNP with 2nd set of primers) is a risky design since it relies on negative data. PCR can fail for lots of reasons. If the SNP is rare, you can easily confused a failed amplification with presence of the SNP.
And it is common for the polymerase to be able to "read across" a mismatch. Yes, I know it's not supposed to, but certain mismatches are well-tolerated and easy to ignore.
RFLPs can work well, but can be really time consuming to optimize. And sometimes they just don't work reliably enough or the enzyme is expensive.
My vote is Sanger, back up the primers so they are at least 150 bp away from the SNP, make the product less than 1000 bp, and sequence from both directions.
  • asked a question related to Quantitative RT-PCR
Question
5 answers
The context of my qPCR experiment:
i) three individuals, one of which used as a control
ii) three replicates of each reaction (a total of 18 reactions, including the GOI and endogenous control, excluding the negative controls)
iii) calculation of relative quantification of gene of interest (GOI) transcripts with the Pfaffl (efficiency) method. (NOT the ΔΔCt method!)
iii) I get a fold difference number, and create a classic bar graph according to the ratios.
What do I do to add standard error bars to the graph? Are they even needed for such a small amount of data? Please help me, I cannot figure this one out.
Anna Vatzika, MSc Student
Relevant answer
Answer
I'm sorry I have missed your latest reply. When sample 1 & 2 have the same condition (which might be the same diagnosis) but have different clinical manifestations. I would prepare a mean of them with "proper" SD (might not really enough to have any scientific meaning). You might also think of super imposing on the bar graph the single measurements as dots (you should have seen these graphs in the literature).
Best wishes
Soenke
  • asked a question related to Quantitative RT-PCR
Question
10 answers
When we perform statistical analysis on qPCR data, do we use fold change or ΔCt?
Relevant answer
Answer
you mean -ΔΔCt?you could use either -ΔΔCt or the fold change ( 2 to the power of -ΔΔCt).
  • asked a question related to Quantitative RT-PCR
Question
5 answers
I have done 3 blind passages to concentrate the viral stock in the supernatant without adding TPCK Trypsin. After passage 1 my Ct value for INF A was 36.96. After P2 the value decreased to 33.36. However, I was expecting a much lower value in the mid-20s. I am freeze-thawing the flasks at -20 degrees to lyse the cells and release the virus into the medium. Is it the correct approach? and can I get a much higher titre without adding TPCK Trypsin?
Relevant answer
Answer
Dear Colleague,
I hope this message finds you well. Culturing Influenza A virus in MDCK cells typically involves the addition of TPCK trypsin to facilitate viral replication and ensure high viral titers. TPCK trypsin cleaves the hemagglutinin (HA) protein, enabling the virus to become infectious. However, you are inquiring about the possibility of obtaining high viral titers without the addition of TPCK trypsin. Below is a detailed and logical discussion on this topic.
Culturing Influenza A Virus Without TPCK Trypsin
Role of TPCK Trypsin
  1. Cleavage of Hemagglutinin (HA):TPCK trypsin cleaves the HA protein of the Influenza A virus, which is essential for viral entry into host cells and subsequent replication. Without this cleavage, the virus remains non-infectious, and the replication cycle is interrupted, leading to lower viral titers.
Alternative Approaches
  1. Trypsin-Free Culture Media:Using trypsin-free culture media generally results in significantly lower viral titers because the HA protein remains uncleaved. Some labs have experimented with alternative proteases or conditions that might support HA cleavage, but TPCK trypsin remains the most reliable and widely used method.
  2. Genetically Modified Virus:One potential approach is to use a genetically modified strain of Influenza A that can replicate without the need for HA cleavage by trypsin. However, such strains are not typically used for standard virology studies.
  3. Endogenous Proteases:Some cell lines express endogenous proteases that can partially cleave HA. However, MDCK cells typically require exogenous TPCK trypsin for optimal viral replication.
Experimental Considerations
  1. Optimization of Conditions:pH and Temperature: Ensure that the culture conditions (pH, temperature) are optimal for both MDCK cell growth and viral replication. Infection Dose: Use a higher multiplicity of infection (MOI) to compensate for the lower efficiency of viral spread without trypsin.
  2. Protease Activity Monitoring:Monitor the culture for signs of cytopathic effect (CPE) and viral replication using assays such as plaque assays, hemagglutination assays, or qPCR. Assess the presence of cleaved HA using Western blotting or similar protein analysis techniques.
  3. Alternative Proteases:Experiment with other proteases that might be able to substitute for TPCK trypsin. However, this requires careful validation to ensure they effectively cleave HA and do not adversely affect cell viability or viral integrity.
Practical Recommendations
While it is theoretically possible to culture Influenza A in MDCK cells without TPCK trypsin, achieving high viral titers under these conditions is challenging and often impractical. TPCK trypsin remains the gold standard for this purpose due to its reliability and effectiveness. If you decide to experiment with trypsin-free conditions, I recommend conducting parallel cultures with and without TPCK trypsin to directly compare the effects on viral titers.
Example Protocol with TPCK Trypsin
  1. Cell Preparation:Seed MDCK cells in culture flasks or plates and grow to 80-90% confluency.
  2. Virus Infection:Infect cells with Influenza A virus at the desired MOI. Allow the virus to adsorb for 1 hour at 37°C in a CO2 incubator.
  3. Media Change:Replace the inoculum with serum-free DMEM containing 1-2 µg/mL TPCK trypsin. Incubate the cells at 37°C in a CO2 incubator.
  4. Monitoring and Harvest:Monitor cells daily for CPE. Harvest the supernatant when CPE is observed (typically 2-4 days post-infection) and determine viral titers using a plaque assay or other appropriate method.
Conclusion
While culturing Influenza A virus in MDCK cells without TPCK trypsin is not impossible, it poses significant challenges and typically results in lower viral titers. TPCK trypsin facilitates efficient viral replication by cleaving the HA protein, making it a critical component for high-yield virus production. If you proceed with trypsin-free cultures, careful optimization and parallel comparison with TPCK trypsin-containing cultures are recommended.
With this protocol list, we might find more ways to solve this problem.
  • asked a question related to Quantitative RT-PCR
Question
9 answers
So I used CRISPR-Cas9 system to knock out the protein of interest. I sequenced the modified region, and then I analyzed and there are 2 KO clones according to my results (deletion-causing stop codons). Next step was western blot to detect the protein level, but when I did the chemiluminescence detection and applied long exposure time I got faint bands- 2 faint bands... I think my antibody is not perfect, because it is a polyclonal antibody.. might be same other protein in that band, beacuse my antibody is not so specific and it could recognise other proteins and bind to them?? What do you think? I checked my clones with RT-qPCR too, and I got amplification in the KO clones too, same as the wild type... what do you think about this? I am confused and try to find out a good answer to this phenomenom.. my ideai is the mRNA is transcribed and the (mutated) protein is traslated , but after that it is degradated, because the protein is truncated or the folding is not good.. but why I see faint bands? might be the antibody? What should I do?
I am looking forward your theories and answers.. Thank you so much :)
Loretta
Relevant answer
Answer
Basics of CRISPR-Cas9: How it works and its components.
Designing gRNAs: Tips for designing effective guide RNAs.
Applications: Different uses in research or therapy.
Experimental Setup: How to set up a CRISPR experiment.
Troubleshooting: Common issues and how to solve them.
Ethical Considerations: Discussions on the ethical use of CRISPR technology.
l Check out this protocol list; it might provide additional insights for resolving the issue.
  • asked a question related to Quantitative RT-PCR
Question
2 answers
Hi!
I notice that reverse transcriptases can be used for both cDNA library construction with template switching (eg. surescript II etc.), and downstream qPCR. To quality-control my library construction, I would like to do a qPCR of focused gene with the first strand cDNA, but reverse transcripted with template switching. However, even with the same reverse transcriptase, will the activation of template switching, as well as the addition of adaptor-UMI before poly T in primer?
Thank you!
Relevant answer
Answer
That's excatly what I mean. Thanks John!
  • asked a question related to Quantitative RT-PCR
Question
1 answer
I'm working on RT-qPCR optimization for gene expression. In melting curve analysis, the 5x primer negative control has a peak that resembles the samples peak but looks shorter. 10x and 2.5 have no curve, and when I applied the gel for verification, no product was seen. Why does 5x primer have this melting curve but no product in gel?
Relevant answer
Answer
Despite no visible gel product, the melting curve peak in the 5x primer negative control likely indicates non-specific amplification or primer-dimer formation, detectable by the sensitive melting curve analysis but not by the less sensitive gel electrophoresis. This could be due to incomplete PCR reactions, primer-dimer artifacts from high primer concentration, or fluorescent dye binding to small, non-specific products. To resolve this, re-evaluate primer design, optimize PCR conditions (including primer concentration and annealing temperature), use more sensitive detection methods, and include additional controls to distinguish specific amplification from artifacts.
Nucleic acid bands can fade or vanish due to diffusion or degradation when running an agarose gel for an extended time. To avoid this, it's crucial to monitor the progress of the gel electrophoresis and stop the run at the appropriate time.
  • asked a question related to Quantitative RT-PCR
Question
3 answers
I am treating human cells with an AAV vector encoding for my gene of interest tagged by FLAG. I want to quantify the mRNA expression of my gene of interest via qPCR to discriminate the endogenous and exogenous expression. I think that if I design primers spanning the junction between FLAG and my gene only the exogenous mRNA will be amplified. Am I correct in thinking this might work? Am I missing something?
I have never worked with FLAG tag so any recommendation would be highly appreciated
(I am also checking for the protein but wanted to combine mRNA and protein quantification)
Relevant answer
Answer
Hi Hellen,
I assume your AAV carries a gene that is also expressed endogenously by your human cells. Will you obtain cDNAs and permorm the qPCR?
I think your approach is correct (spanning the junction of the FLAG sequence and your gene), just a couple of considerations about it:
1- to ensure more specificity you can design one of your primers to hybridise directly into your FLAG epitope sequence (as your gene will be transcribed with it). As the FLAG sequence is only present in your exogenous transgenic AAV won't cause trouble mixing hibridising with other regions.
2- Your FLAG epitope is only FLAG or FLAGx3? (which is a bit longer) Does your transgene have any extra "linker" regions of nucleotides between the FLAG epitope and your gene sequence? Because if you have these "linker" you can also take advantage and span both the linker nucleotides and the FLAG sequence (some of their nucleotides at least) in your primer design.
Remember that in qPCR the amplicon must not have too much bases (not recommended more than 150 nucleotides) and also the general tips in primer designs (similar melting temperature (Tm) of both primers, avoid self and cross-hybridation between primers due to complementary regions...) that you can find in online tools like ThermoFisher.
If you have other doubts about the primer design do not hesitate to contact :)
Also I would recommend testing more than one viral dose of the AAV (I work with Adenovirus, but not with AAV so surely you are way more skilled with those) and also take into account the cells that are going to express your gene of interest (I faced some differences when I transfected a gene with a FLAG epitope between cell types).
What cells will you use for your experiments?
Best regards! I hope your assays go well
Francesc Estrany
  • asked a question related to Quantitative RT-PCR
Question
2 answers
I performed a nucleic acid extraction using 2 protocols for RNA extraction (Dengue Virus) and one parameter I need to evaluate is the integrity of my RNA samples. In this case, I can't use a RNA ladder. Also, I recently performed a RT-qPCR for the same RNA samples and I have the cDNA.
Relevant answer
Answer
Hello Gabriel,
You cannot use DNA ladder for RNA samples. Though RNA and DNA ladders may serve the same purpose, they are specific to their respective nucleic acids.
There are differences between them.
1. RNA ladders will contain RNA fragments of varying sizes.
2. You will have to run RNA samples in a denaturing electrophoresis gel in order to determine fragment sizes.
3. RNA migrates faster than DNA through electrophoresis gel.
You need to consider these differences before you plan your experiment.
However, if you are interested in checking the integrity of your RNA samples, you need not use the ladder. Run RNA samples in a denaturing electrophoresis gel (a denaturing agarose gel containing formaldehyde).
If your RNA samples are intact, you will observe a sharp and clear 28S and 18S rRNA bands. The 28S rRNA band should be approximately twice as intense as the 18S rRNA band. This 2:1 ratio for 28S:18S is a good indication that your RNA sample is intact.
Best.
  • asked a question related to Quantitative RT-PCR
Question
3 answers
What is the best way to get the qPCR software, REST 2009, to work with Windows 10/11? The software was originally designed to run on a 32bit operating system (Windows XP) and will not open in Windows 11. Other than finding an older computer that still has Windows XP, does anyone have suggestions to make the software compatible? I already tried running the troubleshooter and changing the compatibility settings in the properties window. Any other suggestions? Thank you https://www.gene-quantification.de/rest-2009.html
Relevant answer
Answer
hello, I made a tutorial on how to install REST2009 on Windows 11, I hope it helps
  • asked a question related to Quantitative RT-PCR
Question
1 answer
I'm doing a 3C analysis shown in the picture linked and attached below with the Bait fragments labeled and the target fragments that will be tested for interaction with the bait labeled with numbers. The picture shows two possible 3c maps based on two different restriction enzyme digests of the same region.
A, B, C, and D are regions we are considering for analysis in the future to determine if they interact with each other and with the target fragments.
Assuming we can find restriction enzymes that can digest and separate the regions shown is it possible to test between the fragments as shown for interaction or are some of them too small/too close together?
Relevant answer
Answer
In short, I'd suspect the target regions are simply too close to the baits to detect an enrichment of interaction over background. Anything less than 10kb away from the bait region is likely to interact with bait frequently enough by chance that you probably won't be able to discern a specific interaction peak. Ideally, you would like to see the 3C signal drop down to some background level as you get farther away from the bait before you get to the specific interaction target region. Take a look at Fig 2 of this paper (https://www.nature.com/articles/nmeth823) for an example of what this would look like and note that they don't see the interaction signal get down to background until 20kb-40kb away from the bait. I suspose you could try to see how quickly the random interaction signal drops away for your region but you should probably do a quick cost-benefit analysis for that in your head before taking that plunge. Good luck!
  • asked a question related to Quantitative RT-PCR
Question
1 answer
Hello everyone! My PI would like me to measure the amount of mitochondrial DNA in multiple cell lines using qPCR targeting COXI and COXIV. However, I have never done a qPCR that measures mitochondrial genes. Does anyone have any primer recommendations for these genes?
Also, should I use cDNA or gDNA for this experiment?
Relevant answer
Answer
Since you are wanting to measure mitochondrial genome copy number, you want a DNA extraction that includes mtDNA.
cDNA is made from mRNA, which will give you transcript abundance, not genome copy number.
Start by reading the published literature & also the MIQE guidelines.
Good luck!
  • asked a question related to Quantitative RT-PCR
Question
4 answers
I have a question regarding target miRNA expression after transfection with antisense miRNA (miRNA inhibitor). I understand that the expression of the target miRNA will increase after mimic transfection using qPCR. However, should the level decrease after inhibitor transfection? Since the antisense will bind to the target miRNA and reduce its function but not its expression level, I think the expression would be the same between the negative control and the inhibitor-transfected samples. However, some papers show reduced miRNA levels after inhibitor transfection. Which one is correct?
Relevant answer
Answer
Hi Minseon, the gene expression of a given mature miR can be measured by qPCR through estimation of its quantity in the cytoplasm. As the transfection of the miRNA inhibitor will result in the act of this inhibitor over the mature miRNA, when the sample that was transfected with the miRNA will be quantified to that mature miRNA is expected that the gene expression for that miRNA would be decreased, exactly because the action of the inhibitor. To guarantee that this is the case, you can try to quantify the pre-miRNA and the pri-miRNA that gave origin to that inhibited mature miRNA. The case of the negative control it not occur the same level of gene expression than seen in the transfected inhibited one due it was used only the lipofectamine, no inhibitor was present, so basically the gene expression of the miRNA will be the same of the control.
At least this was the case I found in my experiments that I done during my doctorate.
  • asked a question related to Quantitative RT-PCR
Question
9 answers
So I'm using TRIzol for RNA extraction, but suddenly I'm getting no pellet during the isopropanol step *even though I added glycogen.*
A few weeks ago I:
1. Took a large quantity of bacteria
2. Resuspended in TRIzol
3. Made a bunch of aliquots
4. Stored them all at -80
And those couple weeks ago, I was getting~50 ug of RNA per aliquot (with good RINs, and they looked great on RNA gels).
Yesterday, I took another aliquot and tried to prep it using the exact same process, and got nothing. No pellet appeared during the isopropanol precipitation step even though I added GlycoBlue.
I continued the purification to see if the pellet was just hard to see: ~0 ng/uL by nanodrop
Did I just make a pipetting error? Was my isopropanol bad? No: I repeated the repeated the process *again* with completely new sample, completely new isopropanol, and still no pellet at all.
I'm confident I'm lysing my samples well. I optimized the lysis a while ago, but even before optimization, my yields were never this bad.
I'm sure I added the GlycoBlue. I watched it diffuse into my sample.
I'm sure I added isopropanol. I watched the alcohol/water mix and used brand new isopropanol.
I'm sure I mixed the isopropanol/aqueous phase. I watched it carefully.
I'm sure I actually spun my samples. I tried it over and over.
Protocol:
1. Take bacteria+TRIzol aliquot from -80 (which used to give ~50 ug)
2. Lyse via bead beating (same beads, same bead beater, same settings, same duration that gave 50 ug in the past)
3. Add 0.2 V chloroform
4. Centrifuge 12k xg for 15 min
5. Take upper, colorless aqueous phase
6. Add 1 V of RT isopropanol
7. Add ~30 ug GlycoBlue
8. Invert a bunch of times to mix well
9. Incubate 10 min RT
10. Centrifuge ~20k xg for 10 min
Nothing. No pellet at all. Even with glycogen.
I tried spinning again: No pellet
How is this possible?
Relevant answer
Answer
I’ve had similar issues recently isolating small quantities of RNA (ribosome protected fragments). I use 300 mM sodium acetate pH 5.2 and 5 mM magnesium chloride followed by ethanol precipitation (2.5 volumes) overnight at -20 C with addition of 1.5 uL GlycoBlue. Spin at 20,000 x g for 1 h at 4 C in Eppendorf low bind tubes. I see good pellets in some samples but not for others. I do invert tubes sufficiently after addition of ethanol. I wonder if vortexing and centrifuging again might help. In some samples in looks like several particles precipitated on the side of the tube and not the bottom. Perhaps such multi-localized pelleting is the issue. Otherwise my best guess is RNase contamination. Although I am very careful with using fresh gloves and RNase Away on pipettes and gloves.
  • asked a question related to Quantitative RT-PCR
Question
3 answers
Hi,
I am growing Caco2 cells in 24 Transwell cell inserts. I am planning to induce gut leakage by LPS and want to study gene expressions. My concern is to isolate the RNA from transwell inserts since those are very tiny and hard to use cell scrapper. Any protocol or suggestions would be greatly appreciated
Relevant answer
Answer
Hi,
I hope the attachment is useful for your reference.
According to the protocol provided in the reference, you can harvest and lysis cells using Buffer RLT directly without any scraping, followed by RNA purification as instructed.
Good luck!
  • asked a question related to Quantitative RT-PCR
Question
8 answers
Hi all,
I am planning to do a RT-PCR followed by qPCR starting from 100 ng of total RNA. The way we do it in our lab is we dilute the 20 uL of RT reaction containing 1 ug of total RNA 1:4 and use 2 uL of it per 12.5 uL of reaction per well for a 96-well plate for qPCR. Will this protocol work for 100 ng of starting RNA conc.? Will it be too low to detect? We use myScript Sybr Green kits from Bio-Rad. Also, I am starting with low amount of total RNA conc, (in the range of 6-30 ng/uL).
Please tell me your opinions! Thank you!
Relevant answer
Answer
Hello! I have 2ug of cDNA concentration with 20ul after the reverse transcription process. Now I am planning to do the qpcr, how much or ul of cDNA is needed for testing the housekeeping gene Beta Actin desired 20-25 cycles Ct value.
  • asked a question related to Quantitative RT-PCR
Question
2 answers
Hi.
I’ve been unsuccessfully trying to isolate DNA from neurons extracted from adult mouse brains for further downstream analysis.
First I’ve tried sorting neuronal nuclei (Approx. 105 NeuN+ nuclei/sample) (protocol by Nott et al Nat Protoc 2021), followed by DNA isolation and qPCR analysis. However, my PTHrP levels were always undetectable.
Then I moved to commercial kits, and bought the adult neuron isolation kit from Miltenyi. As I read, one would expect approx. 105 isolated neurons per adult brain. I did the whole trinity from Miltenyi: 1. Adult Brain Dissociation Kit, 2. Myelin Removal Beads II, 3. Adult Neuron Isolation Kit. However, again after following the protocols, and immediately isolating the DNA (Nucleospin tissue from MN) I did qPCR (sybr green) and again I did not detect PTHrP in my samples!
I’ve preformed DNA isolation for high yield and concentration in a final 60ul elution volume. Following DNA isolation, the samples were stored at +4C, and the qPCR was done the next morning.
In my control samples from other tissue, I can detect PTHrP which means the qPCR protocol and primers are working fine.
Am I doing something wrong with the DNA isolation and therefore losing my DNA? Is there an alternative method to get clean neuron populations for DNA isolation?
I would be grateful for any help!
Best,
Relevant answer
Answer
Dear Abdus,
Yes. I've used the nanodrop spectrophotometer to quantify the DNA concentration in each sample. But each time it is undetectable.
  • asked a question related to Quantitative RT-PCR
Question
7 answers
Hi ResearchGate community,
I have been trying to learn more about the optical differences between block-based real-time PCR machines like ABI StepOne versus rotor-based machines such as MIC or RotorGene systems.
I understand that some systems rely on ROX as a passive reference dye while others state that it is optional to incorporate it and others do not need such a factor at all.
My question is if you add this fluorescent dye to your master mix, would it interfere with the detection when it is being amplified using one of the systems that do not need such normalization?
Highly appreciate any insight in this regard.
Best,
Negar
Relevant answer
Answer
Dear ResearchGate community,
I'm somehow referring to the same question, I was wondering if it is fine to use a Sybr green master mix containing ROX for a machine that does not require ROX addition, The CFX96 C1000 touch from Biorad will this addition affect the signal detection and if yes are they any ways to subtract it? looking forward to your insights.
  • asked a question related to Quantitative RT-PCR
Question
1 answer
Greetings. Probably the question is not complex at all, but can't find an answer.
If I have RT-qPCR data of gene expression in a sample with multiple analitycal replicates - to compare it to data obtained in other experiments I need to normalize the expression of genes of interest to the expression of reference gene (which is constitutievely expressed)..
How to perform it if there are replicates and expression of both genes of interest and reference gene are in a form of Expression and Standard Error of the Mean?
Is there a formula to adjust GOI SEM using RG SEM?
Relevant answer
Answer
Just think how you'd do it for, say...a western blot. You'd have all your densitometry for your protein of interest, all your densitometry for your loading control, and you'd...divide the former by the latter to get your per-sample protein expression. Then you'd look at your (now normalized) protein levels and see if anything interesting is happening.
You would never work out the SEM of your loading control and the SEM of your GOI and then...do stuff with those SEMs: that's madness. You normalize each sample first, THEN do your comparisons.
qPCR is no different: you take exactly the same approach and do exactly the same thing, with the exception that qPCR data is in log-space, so divisions become subtractions.
So: let's say you have five samples of treated cells, and five control samples.
Isolate RNA, spec it, QC it, make cDNA.
Now, run qPCR, using these ten samples, in triplicate (so three wells per gene, per sample) for two (or three) validated reference genes and your GOI.
This will give you replicate Cq values for everything.
First thing: manual QC.
Look at the replicate Cq values for each sample, and see if they agree. This is basically "did you pipette the same way three times, and also did anything weird happen": you might see something like 22.2, 21.9, 22.1 as replicate Cq values for GAPDH, and this is great. The final GAPDH Cq for that sample is the arithmetic average of those three Cqs.
If instead you have 22.2, 21.9, 26.8, then you can pretty safely assume well #3 just went weird, and discard that data. Use the mean of the two remaining (non-weird) Cqs.
So eventually you should have, for each sample, mean Cq values for your GOI, and mean Cq values for each reference.
Make your two reference gene Cqs into a "normalisation factor": the average of the two.
Then subtract this from your GOI Cq for that sample. This is the dCt value for that sample, for that GOI: that sample is now normalized.
Do this for all your samples. Now all your data is normalized, and is in the form of dCt values.
Note that dCt is somewhat counter-intuitive, as low numbers are high expression, and vice versa. You can just multiply everything by -1, because the statistical comparisons are exactly the same. "-dCt" is a perfectly valid metric.
dCt values are approximately normally distributed, and entirely statistically-tractable. Use these for your stats. From the example above, you will have, for your GOI, five treated dCt values, and five control dCt values: this can be a straight T-test. If paired data, paired T-test.
There are other ways to do this that are mathematically identical but less "OMG LOG SPACE SCARY", which I typically use because log space scares me, but this basic method works and is easy to work through.
So, TL:DR, ignore SEMs, they're not useful here. Go back to the raw data.
If you have data you're willing to share, I could workshop up a basic spreadsheet for you.
  • asked a question related to Quantitative RT-PCR
Question
6 answers
We would like to purchase around 10 thousand DNA oligos in a 96 well format (25 nmol). The cost per base is coming to around Rs 14-15. We wonder if there is any economical option available in the market.
Thank you
Relevant answer
Answer
Dear Colleague,
I trust you are doing well. In response to your request for suggestions on reasonably priced oligonucleotide synthesis services, both within India and internationally, I am pleased to provide a comprehensive overview aimed at facilitating your decision-making process.
Oligonucleotide Synthesis Services in India:
  1. Eurofins Genomics India Pvt Ltd: Eurofins is renowned for its high-quality sequencing and synthesis services. They offer competitive pricing for custom oligonucleotides, catering to various research needs, including standard, modified, and high-throughput oligo synthesis.
  2. Xcelris Labs Ltd: Xcelris is another prominent player in the field, offering a range of genomic services including oligonucleotide synthesis. Their services are known for being cost-effective and reliable, making them a popular choice among researchers in India.
International Oligonucleotide Synthesis Services:
  1. Integrated DNA Technologies (IDT): IDT is a global leader in the area of custom oligo synthesis, renowned for its high-quality products and services. They offer competitive pricing and have facilities in the United States, Europe, and Asia, ensuring timely delivery worldwide.
  2. Sigma-Aldrich (now Merck): Sigma-Aldrich provides a wide range of oligonucleotides through its custom DNA synthesis service. They are known for their reliable quality and extensive options for modifications, catering to diverse research requirements.
  3. GenScript: Offering both standard and customized oligonucleotide synthesis services, GenScript has a strong presence worldwide. Their services are competitively priced and are backed by excellent customer support and fast turnaround times.
Selection Criteria:
When selecting an oligonucleotide synthesis service, consider the following criteria to ensure you receive the best value and quality for your research needs:
  • Quality and Accuracy: High-quality oligos are crucial for the success of your experiments. Look for services with positive reviews regarding the accuracy and purity of their products.
  • Pricing: Compare prices among different providers, but also consider the cost-effectiveness in terms of quality and additional services provided.
  • Turnaround Time: Ensure the provider can meet your timeline requirements, especially if you are working on time-sensitive projects.
  • Customer Support: Efficient and responsive customer service can significantly enhance your experience, especially when customizations or modifications are involved.
  • Shipping and Handling: For international orders, consider the logistics of shipping and handling, including costs and the potential for delays or customs issues.
Recommendation:
Before finalizing your decision, it may be beneficial to request quotes from multiple providers and evaluate any bulk order discounts or promotional offers that could further optimize your investment. Additionally, reaching out to your professional network for firsthand reviews and experiences can provide valuable insights into the reliability and quality of the services you are considering.
Should you have any further inquiries or require assistance in contacting these services, please feel free to reach out.
Best regards,
With this protocol list, we might find more ways to solve this problem.
  • asked a question related to Quantitative RT-PCR
Question
2 answers
qPCR was performed on the same environmental DNA samples, first using a primer pair targeting the archaeal 16S gene, and subsequently, another qPCR was performed using primer pairs targeting the 16S gene of Lokiarchaeota, Bathyarchaeota, and Woesearchaeota, respectively. BLAST confirmed that the primer designed to target the common archaeal 16S gene also indeed binds to the 16S site for Loki-/ Bathy-/ Woese- archaeota. I want to know if it is okay to process this data as follows:
Total Remaining Archaeal gene copies = Archaeal gene copies - Lokiarchaeota gene copies - Bathyarchaeota gene copies - Woesearchaeota gene copies
Relevant answer
Answer
Robert Adolf Brinzer at the very least, I would assume any biases in amplification to be conserved for the specific taxa. But yes, in theory, I am "assuming equal amplification". There were adequate technical replicates, but unfortunately not adequate biological replicates. Even if I am not getting equal amplification for the remaining taxa, the general point that I am trying to convey has not much to do with the remaining taxa, but more to do with the taxa that I am subtracting. What I want to show is that one of the archaeal taxa is selectively enriched in a particular sample. This means that if the relative abundance of Total Remaining Archaeal is rather low, and the relative abundance of one of the specific taxa (Loki, Bathy, Woese) is quite high, then one can conclude that of the total observable archaeal abundance, the composition is skewed towards one of the taxa.
  • asked a question related to Quantitative RT-PCR
Question
4 answers
Hello Researchgate community,
I recently ran into a bizarre (at least I've never thought it could happen). We selected a few DEGs from the scRNA seq dataset and run q-PCR to validate the results (heart tissue).
One particular DEG of interest was DOWNregulated in cardiomyocytes only, but no change in other cardiac cell types, and no change as a whole after combining data from all cells. However the q-PCR data suggested there is a big UPregulation in the whole heart under the exact same experimental condition.
What could be the causes of near-opposite data from RNAseq vs. q-PCR? How should I interpret it, and what's a logical next step here?
Thank you very much!! Any input is much appreciated!!!
Relevant answer
Answer
Interesting indeed, are you confident your qPCR primer is specific to your gene target? It is also possible that when you use LV tissue for the qPCR analysis, you're getting a signal from other cell types but perhaps this signal isn't present in the single cell analysis as you're sampling much less cell counts? A speculative idea.
  • asked a question related to Quantitative RT-PCR
Question
5 answers
I have raw qPCR data. 2 samples "1 control and 1 with gene knock out". I made a serial dilution of each sample and performed a qPCR using 2 reference genes and 3 genes of interest. Now I have the raw data “the CT and average CT of each sample. I want to present the data as a chart. What exactly do I put in the chart. The delta delta CT? Or the 2^-delta delta CT? Or something else?
and do I put all the dilutions in the chart? or just the undiluted original sample? or calculate an average or a geomean of the sample and the diluted samples?
Another question. When I have more than one house keeping gene or reference gene, can I take the geomean of the average CT of both genes to calculate the delta CT?
Relevant answer
Answer
Yep, your math checks out!
It's...a bit convoluted, but it gets you to the right place. You can save time after the dCt step by just directly subtracting:
3.7 - 5.11 = -1.415
2^-1.415 = 0.37
But yeah: your method in principle is solid.
I would report this as "~40% of WT levels", because excessive precision is risky.
Having said all that, comparing two samples is even more risky: even if you only have "one control, one knockout" (assuming, say, this is a cell line), I would still recommend you do at least 3, and ideally 5, biological replicates, such that you can plot replicate dCt values and get a better handle of the range of that "40%".
  • asked a question related to Quantitative RT-PCR
Question
4 answers
Hi ALL,
I am using a pair of primers to amplify a region in my gene of interest from cDNA samples. The cDNA samples are extracted from tissues of mouse of different ages. The gene is known to have decerased expression level when mouse ages. However, I did not see any change of the RT-PCR amplicon band intensities on agarose gel, indicating no change for the transcript level. I did not saturate the PCR products as I tried different cycle numbers (from 23 to 30 cycles). What could be the possible reasons? Should I design new primers targeting a different regions in my gene? Thank you for the help!
Relevant answer
Answer
Sequencing amplification product is always a good option to determine the specificity of the PCR. In addition, you should at least run an end-point PCR with a low number of cycles for an appropriate house-keeping gene as control for your input.
Could it be possible that your primers also amplify genomic DNA, which basically always contaminates your RNA unless you conduct an DNAse treatment step. If so, design exon-exon spanning primers.
  • asked a question related to Quantitative RT-PCR
Question
1 answer
I'm conducting a time course study on gene expression using RT-q PCR for samples treated with 4 conditions: vehicle, RA agonist, Calcitriol agonist or a combination of both agonists over 6 hours. I'm expecting to observe a gradual increase in expression over time for the combined treatment condition due to an additive effect of the ligands. Indeed, I have observed that for all of the time points except for the last one where the Ct value for my combined treatment is 30 while my untreated control at zero hour has a Ct value of around 28.85. Even the Ct value for the vehicle condition for my last time point is around 28.65 so, why am I getting such Ct for the combined treatment?
Relevant answer
Answer
The Ct value depends not only on the gene expression but also on the amount of biological material used, its quality (integrity), assay parameters (e.g. background correction, selection of a threshold value) and the assay performance, all of which can differ between samples and/or runs. Only after somehow controlling all these possible influences you may interpret differences in observed Ct values as differences in gene expression. This is typically achieved (at least in part) by measuring the CT values of some internal control with presumably constant expression under all conditions and in all samples and using plate calibrators where measurements should be compared across plates or runs. And further, Ct values can be quite variable between samples, what mean that it needs some statistics check the statistical significance of observed differences.
Given all this was done but just not communicate by you, then the observed result may indicate a counter-regulation. If so, it should also dampen at even later time-points.
  • asked a question related to Quantitative RT-PCR
Question
2 answers
Basically, I would like to quantitatively detect total bacteria in mice feces. How can I obtain a standard curve to reveal total bacteria quantitatively?
By the way, I have one bacterial species that I grew in a suitable medium, and I obtained a standard curve by making serial dilutions, and I found that bacteria in the DNA whose amount I did not know by substituting it in the Ct equation (obtained from the standard curve). But I don't know how to quantify total bacteria. I would be glad if you help.
Relevant answer
Answer
I understand your challenge in quantifying total bacteria in mice feces. Obtaining a standard curve for this purpose can be tricky, as it's not feasible to isolate and culture all the diverse bacterial species present in the gut. However, there are alternative approaches you can consider:
1. Universal 16S rRNA gene amplification:
  • This method targets a conserved region of the 16S rRNA gene present in all bacteria. By amplifying this region using specific primers and quantifying the amplicons (e.g., using qPCR), you can estimate the total bacterial abundance.
  • Standard curve options:Genomic DNA from a single bacterial species: Similar to your approach, you can use genomic DNA from a single bacterial species (e.g., E. coli) to generate a standard curve. However, this will only provide an estimate relative to the chosen species and won't reflect the true diversity of gut bacteria. Mock community DNA: A more accurate option is to use commercially available mock community DNA containing known amounts of various bacterial species. This provides a more representative standard curve for diverse gut microbiota.
2. Fluorescence-based methods:
  • These methods stain bacterial cells in the fecal sample with fluorescent dyes and then measure the fluorescence intensity to estimate total bacterial abundance.
  • Examples:SYBR Green: This dye binds to double-stranded DNA in all bacteria, providing a direct measure of total bacterial biomass. Propidium iodide: This dye stains only bacteria with compromised cell membranes, potentially underestimating total bacterial abundance.
3. Flow cytometry:
  • This technique uses fluorescence-labeled antibodies to target specific bacterial groups or total bacteria, allowing for quantification and characterization of the gut microbiota.
Choosing the best approach:
The best method for your study will depend on your specific research question, budget, and available resources. Here are some factors to consider:
  • Sensitivity: Some methods are more sensitive than others, which may be important if you are expecting low bacterial abundance in your samples.
  • Specificity: If you are interested in quantifying specific bacterial groups, you will need to choose a method that targets those groups.
  • Cost: Some methods, such as flow cytometry, require specialized equipment and can be expensive.
  • asked a question related to Quantitative RT-PCR
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 Quantitative RT-PCR
Question
2 answers
Hello,
I am trying to quantify the expression level of different snoRNAs between control and CRISPR samples, to know if the knock-out of my gene has an effect on these small RNAs.
A colleague did a first RTqPCR, one-step, and found that they were mostly overexpressed in the CRISPR samples.
I repeated the RTqPCR several times but as a two-step RTqPCR using random primers, and found everytime that they were mostly downregulated in the CRISPR samples which was quite surprising.
I am a bit puzzled that there is so much different between the results, in my opinion if the random primers are less efficient for RT they should be equally less efficient in the control and CRISPR samples so the dCt should not change.
The experiments have been repeated multiple times, I attached the file summarizing my results for 2 replicates of the CRISPR experiment (sample 1 and 2) for 4 different snoRNAs (A to D), for which we have the most technical replicates. The variation between replicates is not really high so it cannot explain such differences between the conditions.
dCt is the average Ct value of the control sample - the average Ct value of the CRISPR sample.
If anybody has a suggestion why we see such results I would be happy to hear it.
Thanks in advance for your help :)
Best regards,
Violette Charteau
Relevant answer
Answer
Hi Violette
May I know how you modified your RT-qPCR protocol targeting snoRNAs? I also have a plan to quantify snoRNA and found several complicating protocols. It would be really appreciated if you could share some experience.
  • asked a question related to Quantitative RT-PCR
Question
3 answers
I have performed 3 RNA isolations on Jurkat cells using the phenol-chloroform principle, using Omega Bio-Tek RNA Solv® & following the protocol. I took care in not disturbing the separated phases when removing two thirds of the aqeous phases but the data I get when I measure the 260/280 & 260'/230nm absorbance ratios lead me to suspect contamination of the samples with phenol or other reagents.
Most of the samples have a 260/280 ratio below 1,6 and only one sample has a 260/230 ratio that comes close to 2 (1,84 to be exact). Subsequent cDNA synthesis & RT-qPCR have not resulted in genes of interest to show any fluoresence at all, only the housekeeping genes in 2 of 7 samples showed up. Problems with primers & RT-qPCR were ruled out as the same setup yielded viable results previously.
Thanks in advance!
Relevant answer
Answer
I agree with Julie Ann Dougherty . RNA quality can be affected by impurities like phenol which can be carried over to samples. Residual phenol is assumed to inhibit PCR reactions by denaturation of enzymes such as polymerases and reverse transcriptase in a concentration dependent manner. So, try to use lower concentration of RNA than what you would be presently using.
You may want to refer to the articles attached below for more information.
Best.
  • asked a question related to Quantitative RT-PCR
Question
3 answers
When I ran qPCR on my samples, some of them returned N/A results (I replicated it twice, and the replicated sample returned Cq value results), so I decided to run qPCR on the same samples again. The sample that previously gave N/A results turned out to have Cq values. What does this mean? And what are the possible issues that cause the first run to return N/A results in some samples?
 
 
Example (because I am not fluent in English, using the example will make it easier to understand that we are on the same page):
 
Sample A:
Gene A -> Cq: N/A
Gene B -> Cq: 36.55
Gene C -> Cq: 35.75
Sample A (replicated):
Gene A -> Cq: 34.78
Gene B -> Cq: 35.97
Gene C -> Cq: 36.12
 
And because gene A of sample A gave N/A as a result, I decided to run qPCR with the same sample again (to see if it could give me a Cq value).
Sample A:
Gene A -> Cq: 35.04
Gene B -> Cq: N/A
Gene C -> Cq: N/A
 
Sample A (replicated):
Gene A -> Cq: N/A
Gene B -> Cq: N/A
Gene C -> Cq: N/A
Relevant answer
Answer
Cq of 35-36 is basically "there was a single target molecule in this well". It's usually the highest Cq value you can get before you hit "N/A".
So exactly like Katie A S Burnette says, what you have here is things like "one, none, none" or "none, one, none".
Your target is there, maybe.
Now you can use more cDNA, potentially, but PCR is exponential: you'd need to add a LOT more to substantially change matters. Twice as much would really only give you results like "none, two, one", at best.
Unless you're already using cDNA at incredibly high dilution, I don't think "adding more" will really change the conclusion that your target is there, maybe.
So you would then need to consider whether "is there, maybe" really represents a finding of biological consequence: it might be detectable, but only just, and only because PCR is incredibly sensitive. If you can barely detect it, can it really be contributing, meaningfully, to your biological scenario?
  • asked a question related to Quantitative RT-PCR
Question
8 answers
We are working on the expression of resistance genes in potatoes using the LightCycler ® 480. There are two ways to evaluation of the results: basic (that use ΔΔCT method) and
advanced (that use E-Method). I know that the second one is more precise, but I can't find an explanation for the differences in the size of units: for the same data we get values 1,57E-06 (basic) and 0,706 (advanced). The reaction efficiency is very close to 2.
How can such differences in the size of the results be explained?
Relevant answer
Answer
Hi Dorota Milczarek : this is all very helpful.
A few tips: quantifying cDNA is not (in most circumstances) meaningful.
Remember, to make cDNA you add RNA, you add primers (lots of primers), and you add dNTPs. At the end of the reaction, all of those are still there, and all of those absorb at 260.
Consider, you are adding (by my calculations) ~2.5ug of RNA to your reaction, using 5ul of RNA. I thus assume the total reaction volume is going to be greater than 5ul, and is probably 20ul (as this seems to be standard for cDNA). If you have 1900ng/ul cDNA, that means you have somehow converted 2.5ug of RNA to 38ug of cDNA: a fifteen fold amplification, somehow.
This isn't really how cDNA synthesis works.
Instead you should assume your cDNA synthesis is approximately 1:1, so if you add 2.5ug of RNA, you get 2.5ug of cDNA.
Assuming 20ul reaction vol, this means your actual cDNA concentration is closer to 125ng/ul.
Conveniently, though, you're still ok: if you're diluting your cDNA the way it sounds, you're basically diluting it 1:20 and using 1ul, so are using about 6ng of cDNA per well.
This is an entirely acceptable amount of cDNA to use (I typically use 8ng, so you're easily within the right ballpark).
So you can probably trust your numbers.
Now, for normalisation, yes: 18S is not necessarily the best choice, both because of abundance, and also because rRNAs are not subject to the exact same degradation/synthesis pathways as mRNAs, so might respond differently to your experimental conditions.
Consider: if 85% of your sample is ribosomes, and 5-10% is tRNAs, mRNAs are only a tiny fraction (5-10%) of your sample. Huge changes in total mRNA content might be masked entirely if you use 18S, because these huge changes are still only a tiny fraction of total RNA.
I would recommend using a couple of stable mRNAs rather than 18S.
Finally, all this said and done, what you are really interested in is the difference in gene expression, and ddCt methods seem almost entirely designed to hide this from you behind a wall of inexplicable numbers and log transformations.
What I like to do (because I can factor in efficiencies this way) is convert all numbers from log values (Cq) to linear relative quantities (RQ).
For this, for each gene, you take the lowest Cq value (i.e. the sample with the highest expression of that gene), and then for each sample:
RQ = efficiency (lowest Cq-sample Cq)
In other words, your highest expressing sample will now be 1 (lowest Cq - lowest Cq equals 0, and anything to the power of 0 is 1), and everything else will be linearly related to that (so a sample with half as much will be 0.5).
This will factor in your efficiency differences, so here you would use 2.16 as efficiency, rather than 2.
You can do the same for your references. Your normalisation factor is the geometric mean of these.
Then you normalise your data by simple division, because we're now in linear space.
Your final (normalised) data should then be log transformed again for stats and plotting, because qPCR data is typically log-normally distributed, but it will now be normalised.
NOTE:
If you know your efficiencies are equal between reactions, you can short cut this all and simply generate dCt values: sample Cq - refgene Cq (use the arithmetic average of reference Cqs if using multiple references).
dCT values are mathematically identical to log transformed normalised RQ values obtained via all the faff described above, so if you can use them, it's much quicker.
For all stats, just compare these numbers. Don't generate ddCt values, because all these tell you is the fold change, which generally you can just determine by looking at the plots.
Stats will tell you whether your expression change is statistically significant.
Your own understanding of your biological system will tell you if the magnitude of this change is biologically meaningful.
  • asked a question related to Quantitative RT-PCR
Question
2 answers
Hi everyone,
i was running a real-time PCR in two different machine but using the same protocol and kit. How is it possible? Can anyone explain this to me
Relevant answer
Answer
It might be that the two machines are set for different volumes of pcr reaction. A higher volume would mean undershoot of temperatures and different times between temperatures
  • asked a question related to Quantitative RT-PCR
Question
1 answer
Something went wrong with our cell incubator over the weekend so to my knowledge the cells inside were deprived of a sufficient level of Carbon Dioxide for about 24 hours. The photo attached shows the state of the cells.
The cells are NTERA-D2 cells. Passage number 16.
Magnification is 10X.
Cells were passaged 3 days ago into a T-25 flask.
My question is; Can I salvage these cells so they can be used for rtq-PCR and colony formation assays? Or should I just thaw a new vial of NTERA cells?
Relevant answer
Answer
Without CO2 supply, cells are adversely affected within five minutes. Usually, when there is a technical fault in the CO2 incubator and CO2 supply is shut, one can minimize pH change by supplementing the media with 25 mM HEPES buffer. Cells can proliferate without CO2 if the medium is buffered with 25 mM HEPES at pH 7.4, but this environment is viable for no more than 10 hours and is highly dependent upon type of cell line and cell concentration.
Cells deprived of sufficient level of CO2 for about 24 hours may be stressful to the cells, and may result in irreversible damage. I would recommend that you should not waste time salvaging these cells as they would not be healthy as before and therefore unsuitable for any cell-based assay. Since you have NTERA-D2 cells in stock, you should go ahead and thaw a new vial.
Best.
  • asked a question related to Quantitative RT-PCR
Question
2 answers
I'm self-studying RNA expression, and I'm quite confused with the difference between Reference gene and Calibrator gene in calculating the relative quantification of RNA expression.
Furthermore, if a GAPDH (housekeeping gene) is used as a reference gene. Would the value of the reference gene be taken from the sample of the treated groups or the control groups?
Example:
if I were to study the expression of IL-8 in fibroblasts, would the value of the GAPDH as the reference gene, be taken from treated groups or negative control groups?
Relevant answer
Answer
I agree with Mohamed Khashan . The calibrator is used to correct for technical variances between different runs. You will have to normalize each sample with its reference gene (GAPDH) of the treated group.
The untreated group may be used as a baseline relative to the expression/detection level of target gene in the treated group. The relative quantity of a target gene in a treated group could be expressed as the fold change relative to an untreated group, using GAPDH, a reference gene, as a normalizer.
You may understand the calculations much better if you refer to the link below. The link below presents data from an experiment where the expression levels of a target gene(c- myc) and an endogenous control (GAPDH) are evaluated. The levels of these amplicons in a series of drug-treated samples are compared to an untreated sample.
Best.
  • asked a question related to Quantitative RT-PCR
Question
3 answers
During work for my undergrad thesis, I've examined and compared gene expression of stress-induced genes in plants challenged with a fungal infection.
I have calculated the relative fold change and wondered: How high does my fold change have to be for it to actually make a difference?
For example:
The log2 RFC between Group A and Group C is 0.63.
According to ANOVA, this difference is significant.
I'm wondering if this difference is enough to change the plants' stress response.
Is there a certain value that I can see as a "threshold" or is mere statistical significance enough to confirm the change in the plant?
Thank you in advance .)
Relevant answer
Answer
There is no cut-off for judging a fold-change (btw: fold-change already is a relative measure; it is superfluous to stress that you your fold-change is relative) being biologically relevant, as this depends on too many factors (what gene, involved in what signally/metabolic processes, in what cells, what is the proportion of relevant cells in the sample, at what time, are the cells synchronized, is the mean change considered or are expression bursts relevant, what compensatory effects can exist, can it be an unspecific side-effect without practical relevance in the cell, it the gene expression change counter-balanced by post-transcriptional or post-translational regulation, etc etc etc). It's your job as expert biologists to make such a judgement.
As Can wrote, the significance test only judges the information from your sample data to compare the sample estimate (b) to a hypothetical population value (h): d = b-h. If the information is considered sufficient, then b can be believed to be "on the correct side" of h, so the test tells us if we may have confidence in the sign of d, or that we can statistically distinguish b from h. This hypothetical value if often zero (h=0, so d = b), it is about interpreting sign of b (here: the sign of the log fold-change calculated from your sample).
Note that the point estimate b is associated with uncertainty. Consider the typical case that h=0. A test tells then that you can have confidence that the sign of b is correct, but not that b itself is correct. It might be that b is rather large and the p-value is small, but a irrelevantly small hypothetical value close to zero would not be statistically distinguishable from this h. So the data may be compatible with irrelevant values of h. If relevance (not statistical significance) is of interest, it's more useful to interpret the confidence interval (CI), which is the range of all possible values of h that would be statistically distinguishable from b (giving low p-values in a test). Only if the limit closest to zero is large enough to be considered relevant, then the information from the sample is sufficient to exclude irrelevantly small values of h and claim a "relevant effect".
But these are all technicalities. How large a relevant effect has to be is an expert judgement and cannot be answered statistically. Very often, biologists don't have much of a clue, so the best one can do to claim that one identified the direction of regulation (and avoid to say that the amount of regulation may not be of any biological relevance).
  • asked a question related to Quantitative RT-PCR
Question
6 answers
Hi y'all,
I am running into some difficulties with qPCR (SYBR). To give y'all a brief background summary, I transfected some cells (ASO) and am running qPCR to see its effect on known genes using qRT-PCR. The first time I ran a qPCR, I saw little to no amplification. I re-ran it again and this time I went back to the beginning (RT-PCR from the original RNA) and was able to get some data. However, I am running another qRT-PCR but this time it's not working at all, as in I am seeing zero amplification, which is weird because, again, we know for a fact that some of the genes are supposed to show up in abundance. I was very careful throughout the whole process, so I don't think there was any human error. I haven't run a gel yet, so that's something that I have in my mind. I just wanted to see if y'all have faced similar problems.
Thanks in advance!
Relevant answer
Answer
I agree with the suggestions above. Also, is anyone else having the same issue and if so, what are any commonalities? Same SYBR? Same primers? Same QPCR machine?
My best guess would be your cDNA is not good. But, I did have a QPCR machine itself stop working one time (and I was the only user at the time). Double check your program too (sample setup and cycle parameters).
  • asked a question related to Quantitative RT-PCR
Question
2 answers
What are the validation timelines for quantative real time PCR and western blotting results, is it 24 hours for qpcr and 48 hours for western blotting if so! Why then!?
Relevant answer
Answer
Hi
It depends on your treatment efficiency. Researchers conducts 24, 48, and 72 hours of treatment and if required do it for a long time. Technically, treatment conditions for both PCR and Western experiments should be of the same time period. However, sometimes impact on protein expression may need more time.
Regards
Saurabh
  • asked a question related to Quantitative RT-PCR
Question
4 answers
After trying different protocols I was able to extract RNA from old olive tree leaves with a phenol / chloroform : IAA protocol. Nanodrop measurments of the samples gave 260/280 rate ranging between 1.25-1.42 and 260/230 from 1.04-1.55 and a content of 100 - 200 ng/ul. I proceeded creating cDNA using 200ng of RNA template for a 20 ul cDNA volume. Then a qPCR was performed with the Kapa SYBR FAST qPCR Master MIx (2x) with the use of 1ul of the created cDNA for a 10ul reaction. The amplification plot looked like the attached. I am actually inexperienced but i was told that the refrence gene i am using (actin) is showing up late (30-35+ cycles)
I think that there is probably a phenol contamination in my samples and that is the cause for this bad qPCR. But i am also thinking about the quantity of the RNA that was used. Maybe it was too little, explaining the late amplification or too much taken the fact i didn't dilute my cDNA. Thank you very much in advance!
Relevant answer
Answer
Dear Vasiliki Kavoura per se the amount you used per qPCR shouldn't be a problem imo, in facts it is quite a lot (you may have to dilute it more). That is assuming the concentration measurement is ok and RNA intact; did u heck that there is no DNA and the RNA is not degraded?
Otherwise, the most likely is purity as you say: Your 260/280 260/230 are quite low, so as you mention there could be contamination of possibly phenol and that would be a problem (not just for amplification but also Reverse Transcription. So your problem can be both cDNA synthesis and qPCR.) If you can try to cleanup the RNA more or use a different protocol maybe it would be better (check this recent study for difficult plant samples:
Maybe there is something helpful in here for your case.
Apart from that, for the qPCR step I think it is good to dilute the cDNA more in general, and you can make a dilution curve to see if you have something that inhibits or just generally low levels. You are using a lot, for qPCR you can use a dilution of 1/100 or more depending on expression level and how much you have.
So you could make serial dilutions and use e.g. 1/20 1/40 1/80 etc etc and from the curve you can see where the amplification efficiency is ok (in this case e.g. 1/2 dilutions should correspond to 1 Ct difference for 100% efficiency. This might be a bit difficult with your Ct values being very high, though, which will give a lot of variation. Either way, best recommendation is to try and have better quality RNA.
Last, maybe you can include a positive control (some easier sample RNA, something you are sure will work and give good RT and amplification for actin, to exclude that there is any other problem in the process.)
Good luck.
  • asked a question related to Quantitative RT-PCR
Question
3 answers
I have nhe6 KO mice, confirmed via genotyping from their tail clips. I isolated BAT, iWAT, and gWAT from three controls and three KO mice. RNA isolation followed by cDNA preparation was performed from these isolated tissues. On running qPCR, I expected low ct values (high nhe6 expression) in controls and high ct values or undetermined ct values (may be?) for KO samples. However, shockingly! I got similar ct values (30-32) for both- controls and KO.
The questions here is-
Is it correct to perform qPCR on genomic knockouts? AND WHY?
Relevant answer
Answer
Alternatively, you might've included _too much_ cDNA in the reaction (cDNA synthesis buffer is inhibitory to PCR, so you want to dilute your cDNA after synthesis, ideally).
But like Ciaran Daly says, Cq values of 30-32 are far too high to be trustworthy: these correspond to countable numbers of target molecules (like, 5-20 targets per well) and are thus very susceptible to stochastic noise, mispriming and just general PCR shenanigans.
It would be helpful if you could provide more detail about your entire experimental set-up, since there are lots of places where things can go wrong.
How are you isolating the RNA?
What are your yields?
Are you QCing your RNA afterwards, and how?
What are your 260/280 values, and more importantly, your 260/230 ratios?
How much RNA are you using for cDNA synthesis?
How are you priming (oligodT, random priming, or both)?
Are you diluting your cDNA afterward?
How much cDNA are you using per well, assuming 1:1 conversion?
  • asked a question related to Quantitative RT-PCR
Question
3 answers
Hello fellow researchers,
I'm currently conducting a qPCR experiment and have encountered an issue with varying PCR efficiency between my reference and target genes. Some of the reactions are showing efficiencies above 110%. Initially, I had planned to employ the ΔΔCT method, but these efficiency differences have raised concerns.
I've taken steps to troubleshoot the issue by thoroughly checking melt curves and running agarose gels to confirm the quality of my samples, and everything appears to be in order.
To provide a better understanding of my situation, I've attached a picture of one of the efficiency calculations for your reference.
Now, I'm considering switching to the relative standard curve method to analyze my qPCR data. However, I'm curious to know if the efficiency of the reaction still plays a critical role in this approach. Are there any potential pitfalls or considerations when using the relative standard curve method, especially when dealing with varying PCR efficiencies?
Thank you for your help!
Relevant answer
Answer
For your data, the 95% confidence interval for the efficiency is (2.0, 2.5), so an efficiency of 2.0 is still statistically compatible with your data.
  • asked a question related to Quantitative RT-PCR
Question
6 answers
Hellow fellow academics
I am currently in a dilemma and I would really appreciate some suggestions/guidance on the matter.
Situation:
I have overexpressed my gene of interest (GOI) from wheat in Arabidopsis using the floral dip method and with strict screening on MS-Hygromycin, obtained my T3 transgenics. Now the problem is that while the selection, on the media has been successful, I am not able to get a band of my GOI on agarose gel after doing semi- RT-PCR. Initially, I thought that maybe my overexpression was unsuccessful so I took the T3 seeds to screen again on the media, but, the result was the same; the overexpression was successful and met the segregation ratio requirement of 100% germination. As this is my first time working with transgenics, please enlighten me on where I could be going wrong.
Please advise. Thank you for your time in advance.
Dee
Relevant answer
Answer
Ok, that helps a lot. So, if your GOI is controlled by the 35s promoter there are a few possible explanations for the lack of expression in your assay.
It is possible (but not common) for an overexpressed gene to be toxic and thus silenced by your plant.
It is likely that your expression assay is not working properly and could use more controls. Here is what I suggest.
1. genotype your T3 plants for the 35S:GOI fusion construct (F primer in 35S, R primer in GOI), this is standard end-point PCR on gDNA
2. use a housekeeping gene as a control for your RT assay (actin is a good choice) in general
3. re-try the RT of your GOI, use a kit for the RNA extraction, be sure to DNAse treat your RNA, and use oligo-dT for the cDNA synthesis reaction.
hope this helps!
  • asked a question related to Quantitative RT-PCR
Question
3 answers
Is there anyone who has done TaqMan assays using average regular use PCR mastermix (not the TaqMan assay specific mastermixe) using cDNA as template for the qPCR test? I wanted to know the ins and outs of the procedure and the optimization you did to get accurate results.
Thanks in advance.
Relevant answer
Answer
No, you cannot perform TaqMan assay using average regular use PCR master mix.
If you wish to perform PCR using the regular PCR mix, the assay is no longer called TaqMan Assay because the defining feature of a TaqMan Assay is the probe. This small piece of DNA matched to the DNA template being measured has two special molecules attached, a fluorescent reporter dye (R) and a quencher (Q). While both molecules are attached to the probe, the fluorescence of the dye is suppressed by the quencher. These probes bind to the template DNA after it has been denatured into single strands but before it has begun duplicating, making sure that all duplications of the template interact with the probe.
During the PCR reaction, Taq DNA polymerase extends the primer through the polymerase activity, as it approaches the probe it displaces the probe and cleaves it through the 5′ to 3′ exonuclease activity. This separates the reporter dye and the quencher dye from the probe, which results in increased fluorescence of the reporter. Accumulation of PCR products is detected in “real-time” directly by monitoring the increase in fluorescence of the reporter dye with an automated PCR system.
The assay which you would wish to perform is called two-step reverse transcription-polymerase chain reaction. In this assay, two enzymes are used namely, reverse transcriptase to produce single-stranded cDNA copies, which are then used as templates in an amplification reaction catalyzed by a thermostable DNA polymerase. This assay is the traditional method of RT-PCR in which the two synthetic reactions are performed separately and sequentially.
The TaqMan Assay is a real-Time PCR assay which detects the accumulation of amplicon during the reaction. The data is then measured at the exponential phase of the PCR reaction. The assay which you may plan to perform using average regular use PCR master mix is a type of conventional PCR using agarose gel which is not as precise as qPCR. By using the regular use PCR master mix, you cannot perform qPCR because for qPCR one requires the fluorescent reporter molecule such as fluorescent dye, a labeled oligonucleotide primer or probe such as (TaqMan Probe) for fluorescent detection which is monitored by the automated PCR system. Real-Time PCR makes quantitation of DNA and RNA easier and more precise than conventional PCR.
So, if you wish to use the average regular use PCR master mix, you need to perform the two-step reverse transcription-polymerase chain reaction and not qPCR.
Best.
  • asked a question related to Quantitative RT-PCR
Question
1 answer
I have an unusual question: I am working on a Erasmus internship project with Drosophila mutants at 2 different timepoints and with WT, KO and KI condition. A company analyzed the data using DESeq2 and I have only got loads of PDFs and the results_apeglm.xlsx file.
This contains: Transcripts per million for each gene, replicate and timepoint with the comparison for looking at DEGs - so I have a padj and log2FC value. A snippet is attached as an example.
I now want to construct a graph and clustering where genes that are going in changing directions between WT and KO over time become visible out of the hundreds of candidate DEGs. With this I want to narrow down the long list to make it verifiable with qPCR and serve as a marker for transformation from presymptomatic to symptomatic.
I am setting up my analysis in R and want to use the degPatterns() function from DEGReport, as it gives a nice visual output and clusters the genes for me.
How can I now transform my Excel sheets, to a matrix format that I can use with degPatterns()? The example with the Summarized Experiment given in the vignette is not really helpful to me, sadly.
Thank you all for reading, pondering and helping with my question! I would be very happy if there´s a way to solve my data wrangling issue.
All the best,
Paul
Relevant answer
Answer
Hi, what exactly do you need? A matrix from excel sheet. Then, simply read excel file using
as.matrix(readxl::read_excel(your_file_location))
function. You need to remove few columns and then matched the columns to meta data.
and IF degPatterns() function is not working properly, then you may need to clean and re-transform your data.
  • asked a question related to Quantitative RT-PCR
Question
6 answers
I am optimizing qPCR assay using a pooled cDNA sample. I have several target genes (100-200bp).
current template dilution: 1:30 (30x)
final primer concentrationn: 0.27 uM
annealing temp: 60C
extension temp: 72C
Ct values I get using this template dilution range from 32 to 35, which I think are too high (aren't they?). Increasing the template to 1:10 (10x) and decreasing the annealing temp close to primer Tm (55C) didn't do much.
All melt curves show single peaks at expected Tm. No problem with primer dimers and specificity.
Do you think I should increase the primer concentration to 0.5 uM to lower the Cts?
Relevant answer
Answer
Nothing wrong with a Ct in the 30's but if you're expecting even lower target DNA content in other samples you'll be struggling to detect it
The primer concentration should be determined by the kit/ mastermix you're using. For example, the QuantiTect SYBR green from Qiagen recommends 0.3uM primers. You can titrate these upwards but it's probably not the primary issue.
Is there any reason you've diluted your samples so much and when you say the 1:10 didn't help much, what Ct did this decrease to? As Can said, Ct should decrease by 1 with a doubling of DNA.
Possibilities to consider:
1. Is your template input just really low. You can quantify total DNA input using the Qubit or Tapestation. You can use a Nanodrop as last resort. Given you're pooling samples, is there any reason you would expect higher Cts?
2. Have you introduce contaminates during sample prep. Ie residual ethanol is your samples or nucleases.
3. Have the samples and/ or reagents been freeze-thawed too many times.
Things to list for future help:
1. DNA/ RNA input.
2. Kit you're using.
  • asked a question related to Quantitative RT-PCR
Question
2 answers
Trying to design my first set of methylated primers. Ran a temperature gradient and think I’ve narrowed down a functional range of annealing temperatures across primer concentrations.
Now, I wanted to finally test efficiency across a few annealing more select temperatures. However, I’m realizing that in my labs qPCR protocols, most of them don’t carry an extension step, only the annealing and denature step.
My understanding is that since the templates and products are small (100bp) the copy is usually completed during the ramp up to the denature step. I’ll add that my Tms are approximately 64C, and my estimated annealing is consequently in the 55-61C range. We are using IQ sybr green super mix (iTAQ polymerase).
Just wanted to inquire if this was indeed the case, and if I should rerun my temperature gradient with an added extension step or just proceed with piloting.
On an agarose gel, I didn’t see any double banding across temperatures (suggesting high primer specificity?) albeit brighter bands were detected at specific temperatures, within 2C-6C of the Tm, so I wanted to run sample dilutions across a few degrees to maximize efficiency.
thanks for any help!
Relevant answer
Answer
Efficient amplification of such short products does not need any extension time; it also needs no annealing time and no denaturing time (some denaturing time is needed for efficient separation of high-molecular template DNA during the first few cycles to avoid excessive re-annealing).
The critical part is just to actually reach the target temperatures in the whole volume of your PCR. If the volume is small, waiting for more than a few seconds is simply a waste of time. In rapid-cycle PCR, where the surface-to-volume ratio of the reaction tubes is typically large and the instrument allows steep heating and cooling rates, no waiting time at all is required in any step. In my personal experience, shorter cycling times typically increased the specificity of the amplification, too.
Adding to Pauls excellent answer I'd like to recommend using a 10-15% PAGE which separates these short products and primer dimers well:
4.5 % (w/v) Acrylamide
0.5 % (w/v) Bisacrylamide
0.08 % (w/v) APS
0.2 % (v/v) TEMED
in (1x)-TPE-Buffer
  • asked a question related to Quantitative RT-PCR
Question
2 answers
Hello, I am working on microRNA expression studies on Regeneration. I isolated RNA from my sample and converted it into cDNA using Poly A polymerase and Reverse transcriptase enzyme. Previously I designed a miRNA-specific Forward primer at the melting temperature of 60°C. For Reverse primer, I used Universal Reverse primer from a commercial kit. But now I need to design a miRNA reverse primer for myself. Kindly suggest me method to design a reverse primer for Poly A-tailed miRNA. Thanks in advance.
Relevant answer
Answer
As it is a poly-A that means the end of your sequence is several A codons in a row. Therefore your reverse primer needs to be poly-T.
  • asked a question related to Quantitative RT-PCR
Question
3 answers
Is it possible that the amplification failure products can be visualized in electrophoresis? Due to the failed amplification results it shows bands in my electrophoresis with bands that are quite clear. My amplification curve clearly shows amplification failure, but when I look back at it with electrophoresis there are some obvious bands, how is that possible?
Relevant answer
Answer
I think this band is because of the primer bind with itself we call it dimer
  • asked a question related to Quantitative RT-PCR
Question
3 answers
Hi everyone. I have searched all around the internet and literature for the answer to this question but haven’t been able to find any info regarding my specific situation.
I have multiple experiments consisting of qPCR data but can’t figure out how to best analyse it. I have WT and KO cells which I apply 3 treatments to and I have a control (no treatment) for both genotypes, and I check 15 genes. What I really want to show is if the genes are up/downregulated when I add a treatment in ko vs wt, so I want to make my comparison between the genotypes. But I can’t compare them directly, because at baseline they have quite different expression levels already, so I want to take the control for each into consideration. Before I was plotting -Dct (normalised to housekeeping gene only) and would compare each treatment in each genotype to its own control. But my group didn’t like this, which I understand, because the graphs are cluttered and I don’t show the comparison I’m really trying to make. I worked with a bioinformatician with my idea to normalise the Dct for each genotype/treatment to its own control and in that way make DDct, and then I compare the -DDct between genotypes for each treatment using an unpaired t-test. I don’t do fold change. These graphs are much nicer to look at, but my supervisor says it doesn’t make statistical sense this way, and wants to keep the graphs the original way.
can anyone help me out? What is the best way to analyse and graph my data?
Relevant answer
Answer
Ensuring that your data analysis and visualization methods are scientifically ⁠ valid and effectively convey the information is crucial. Here's a suggestion for a statistical analysis ⁠ that might better suit your needs: ⁠
Relative Expression Analysis: ​
Instead of using -Dct or -DDct, Take into account ⁠ utilizing the approach referred to as 'Relative Expression'. The fold change in gene expression between treated samples and their ⁠ corresponding controls is calculated within each genotype using this method. This way, you'll be comparing the change in expression ⁠ due to treatment in both genotypes separately. ‌
Relative Expression (RE) = 2^(-Dct) [where Dct ⁠ = Ct(target gene) - Ct(housekeeping gene)] ​
Fold Change Calculation: ‍
Calculate the fold change for each gene between treated samples and their controls ⁠ within each genotype using the relative expression values obtained in step 1. ‍
Fold Change Division of RE(treatment) by ⁠ RE(control) RE(treatment) / RE(control) ​
Comparison Between Genotypes: ‍
Having the fold change values for each gene within both genotypes, examine and contrast the fold changes between KO and WT genotypes across all treatments An adequate statistical test ⁠ can be employed to perform this task, like employing either an unpaired t-test or a non-parametric alternative when the data doesn't satisfy the assumptions of a t-test. ‌