Science topic

Gene Expression - Science topic

The phenotypic manifestation of a gene or genes by the processes of GENETIC TRANSCRIPTION and GENETIC TRANSLATION.
Questions related to Gene Expression
  • asked a question related to Gene Expression
Question
1 answer
HL-60 cells are cultured and expanded in RMPI medium containing 10-15% FBS and the cells are in single form and never form aggregates. However after multiple passages I observed that these cells made disordered aggregates although they are still being proliferated and expanded!!!
Now, I don't know exactly that they are being changed in their gene expression pattern thus being differentiated or they might have Mycoplasma contamination?
Relevant answer
Answer
Hi, My HL-60 cells also aggregates sometime...
Did you resolve that problem??
Was it really Mycoplasma contamination?
  • asked a question related to Gene Expression
Question
2 answers
I am currently planning a bacterial gene expression study and am seeking advice on selecting appropriate reference genes for normalization purposes. I aim to ensure the accuracy and reliability of my gene expression data across various experimental conditions.
Could you please suggest commonly used reference genes for bacterial studies? I am particularly interested in genes that exhibit stable expression levels across different bacterial species and experimental conditions.
Additionally, if you have any insights or recommendations on experimental validation techniques for assessing the stability of reference genes, I would greatly appreciate your input.
Relevant answer
Answer
In bacterial studies, certain genes are often used as references due to their stable presence and relatively conserved sequences across different bacterial species. These reference genes serve as internal controls for gene expression studies, aiding in normalizing data for accurate comparisons. Some commonly used reference genes in bacterial studies include: such as 16S rRNA, gyrB (Gyrase subunit B),
rpoB (RNA polymerase subunit B), recA (Recombinase A), and tuf (elongation factor Tu), These genes are not only used as references for gene expression studies but also for phylogenetic analyses, species identification, and other molecular biology applications in bacterial research.
  • asked a question related to Gene Expression
Question
8 answers
I am investigating changes in gene expression in cells after light irradiation at multiple wavelengths using ddCt method. I would like to compare the control and irradiated areas and describe the increase or decrease in gene expression. Usually the Dunnett method is used for comparison, but I believe the ddCt values are not normally distributed.
What is the best multiple comparison method?
Relevant answer
Answer
Hi
in a last paper I saw that 44 cell death genes were identified (based on array results) and I guess it could be more since it depends on lot of things, that's why I proposed this way.
all the best
fred
  • asked a question related to Gene Expression
Question
1 answer
How to download Colon cancer (COAD) gene expression data which includes tumor tissue samples and corresponding control tissue samples from TCGA(https://portal.gdc.cancer.gov/repository). Any information or resources you could provide would be immensely appreciated. I look forward to your guidance. Thank you very much for your time and assistance.
Relevant answer
Answer
Go to this site (https://xenabrowser.net/datapages/?cohort=TCGA%20Colon%20Cancer%20(COAD)&removeHub=https%3A%2F%2Fxena.treehouse.gi.ucsc.edu%3A443) and click on gene expression RNAseq -> IlluminaHiSeq* and then you can find download link.
  • asked a question related to Gene Expression
Question
1 answer
1. For the gene expression data (microarray dataset which is been extracted from the Gene Expression Omnibus (GEO) platform), which of the following normalisation techniques are suggested as the best in order to handle the outliers: quantile, log, z score,… As I was following articles where they were normalising by combining quantile and log, but when I check for the dataset I’m working on, there are outliers which are then negatively skewed after normalising. Is it normal to have skewness even when they are normalised? If not, are there any other ways where we can normalise them without any skewness?
2. I was using the Student t-test and Fold change values, to identify the DEG for two different cores, where I ended up getting 202 genes in total, where 44 are common between these two cores. Is it normal to get some common differentially expressed genes for two different conditions ? If not, what mistake probably would have occured?
3. Any precise formula to calculate the fold change values from the gene expression values? All over the internet, there are plenty of formulas. So, I'm confused about which formula to use.
Relevant answer
Answer
1. It is not uncommon to get skewness even after normalization of the data. Some degree of skewness is inherent to RNA-seq data due to the natural variation in gene expression.
2. It is normal to get some common differentially expressed genes between two different conditions. This might occur due to perturbation in common biological processes or pathways.
3. You do not require any sophisticated formula to calculate fold change. The simplest way is:
a. For each gene, you calculate the average expression value across all replicates in each condition (conditions 1 and 2).
b. Divide the mean expression in condition 2 by the mean expression in condition 1.
  • asked a question related to Gene Expression
Question
3 answers
I am a PhD student working with human plasma looking to do miRNA-seq analysis (differential gene expression, isomer analysis and isolating the best housekeeping genes within a cohort).
I'm keen to do the analysis myself, but after doing a lot of literature searches (and YouTube tutorials) I am really struggling to do so.
Does anyone know of a useful contact, course or information pack that will help me successfully complete the pipelines? Thanks in advance!
Relevant answer
Answer
Hi Oenone Rodgers,
I have previously completed over 10 projects on miRNA analysis. If you require any assistance with analysis, I can provide support until your publication. I am available to work as a freelance support (my email is [email protected]). Alternatively, you can reach out to us at [email protected].
Good luck with your project. We can connect with you for better clarity and assistance.
Regards,
  • asked a question related to Gene Expression
Question
2 answers
Dear all,
I need a stable expression of the OVA gene in B16F10 cells. I've tried introducing the OVA gene via retrovirus transduction, but it doesn't seem to be working. Transduction on control L1.2 cells worked fine, suggesting that there was no issue with the viral titer and transduction protocol. 
Has anyone successfully done a retrovirus transduction on B16F10 cells and would like to share the protocol with me? Would it be easier for me to just move on to try lentivirus?
Thanks! Any suggestion will be greatly appreciated!
Regards,
HS
Relevant answer
Answer
Generating OVA (Ovalbumin)-expressing B16F10 melanoma cells involves introducing the gene encoding ovalbumin into B16F10 cells so that these cells express ovalbumin as a model antigen. This process is typically done to create a cancer model that presents a known antigen (OVA in this case), which can then be used in immunological experiments to study antitumor immune responses, vaccine efficacy, and T cell responses. Here's a simplified overview of how to generate such a cell line:
  1. Cloning the OVA Gene: The first step involves cloning the gene encoding ovalbumin into a suitable expression vector. This vector must have elements that allow for mammalian cell expression, including a strong promoter (such as CMV), and, if necessary, a selection marker gene (like neomycin resistance for G418 selection).
  2. Transfection: The plasmid containing the OVA gene is then introduced into B16F10 cells using a transfection method suitable for these cells. Common methods include lipid-based transfection, electroporation, or viral vector-mediated gene delivery. The choice of method depends on the efficiency of transfection and the potential toxicity to the cells.
  3. Selection of Stable Clones: After transfection, cells are usually placed under selection pressure using an antibiotic (if an antibiotic resistance gene is included in the vector), to select for cells that have stably integrated the plasmid into their genome. This step might take several days to weeks, and it's crucial for eliminating untransfected cells.
  4. Screening and Verification: The next step involves screening for cells that express ovalbumin. This can be done using PCR to confirm the presence of the OVA gene, Western blotting, or flow cytometry to confirm the expression of the ovalbumin protein on the cell surface or in the cells. It's important to verify both the presence of the gene and the expression of the protein.
  5. Expansion and Cryopreservation: Once positive clones are identified, they can be expanded and cryopreserved for future use. It's advisable to perform additional characterization, such as assessing the level of OVA expression over time and after freezing/thawing cycles, to ensure the stability of the cell line.
  6. Functional Testing: Finally, it's important to test the functionality of the OVA-expressing B16F10 cells in your specific application. This might involve immunization experiments, co-culture with T cells to assess immune response, or use in in vivo models.
This process requires a good understanding of molecular biology techniques, cell culture, and genetic manipulation. If you're planning to undertake this project, ensure you have access to the necessary facilities and regulatory approvals, especially if you're working with viral vectors or genetically modified organisms (GMOs).
l Perhaps this protocol list can give us more information to help solve the problem.
  • asked a question related to Gene Expression
Question
2 answers
I plan to use the kit from life technologies using CRISPR nuclease vector kit.
Relevant answer
Answer
Dear Colleague,
Identifying target sequences along with Protospacer Adjacent Motif (PAM) sites for CRISPR/Cas9-mediated genome editing involves a series of strategic steps. The CRISPR/Cas9 system requires the design of a guide RNA (gRNA) that is complementary to the target DNA sequence, adjacent to a PAM site, which is essential for Cas9 nuclease binding and DNA cleavage. Here is a methodical approach to identify suitable target sequences for your gene of interest:
  1. Understand PAM Requirements:First, familiarize yourself with the PAM sequence required by the Cas9 nuclease you plan to use. For Streptococcus pyogenes Cas9 (SpCas9), the most commonly used variant, the PAM sequence is NGG, where N can be any nucleotide.
  2. Select Your Gene of Interest:Identify the genomic region of your gene of interest where you aim to induce a modification. Consider whether you intend to knock out the gene function (targeting the coding sequence) or modulate its expression (targeting regulatory regions).
  3. Use Bioinformatics Tools:Leverage online bioinformatics tools designed for CRISPR target and gRNA design, such as CRISPOR, CHOPCHOP, or Benchling's CRISPR tool. These platforms allow you to input your gene of interest and will automatically identify potential target sequences adjacent to PAM sites, considering factors like off-target potential, efficiency, and specificity.
  4. Evaluate Off-Target Risks:A critical aspect of target sequence selection is evaluating the potential for off-target effects. The tools mentioned above typically provide scores or metrics indicating the likelihood of gRNA binding to unintended genomic locations. Select gRNAs with minimal off-target potential to ensure specificity.
  5. Check for GC Content and Secondary Structures:Ideal gRNA sequences generally have a GC content between 40% and 80% and lack significant secondary structures that could impede Cas9 binding. These parameters are often considered by automated design tools but should be reviewed manually if necessary.
  6. Experimental Validation:After selecting your target sequences and designing gRNAs, empirical validation is crucial. Clone your gRNA into an appropriate vector, introduce it into your target cells along with the Cas9 nuclease, and assess the efficiency of target gene editing using techniques such as T7 endonuclease I assay, Surveyor assay, or sequencing.
  7. Consider Ethical and Safety Guidelines:When working with CRISPR/Cas9 technology, it's essential to adhere to ethical guidelines and safety regulations, especially if your work involves human or animal subjects.
By following these steps, you can identify appropriate target sequences with PAM sites for CRISPR/Cas9-mediated gene editing. This technology offers unparalleled precision in genome engineering, but its success hinges on careful target selection, design, and validation.
Best regards.
Take a look at this protocol list; it could assist in understanding and solving the problem.
  • asked a question related to Gene Expression
Question
1 answer
I need to study the relationship between gene expression of GDF9 GENE in mice and its relationship to their exposure to heavy metals, AND please what about recommended me about this study
Relevant answer
Answer
Look up some pre-designed qRT-PCR assay available in the market (e.g. ThermoFisher, IDT etc) already. You can find if that has been used in publications.
  • asked a question related to Gene Expression
Question
2 answers
Genetic Engineering. Sequence. mRNA sequence.
Relevant answer
Answer
I think that if you have a good yield almost every RNA isolation technique may work (total RNA, micro RNA if you are interested). Do you use a kit or a classical trizol + cloroform/ethanol extraction?
Also it depends if you want to focus on one type of RNA: if you want to enrich in mRNA transcripts you can use oligo-dT Beads (as Lucia Hronska commented), if you want to focus on microRNAs or tRNAs you may use a micro-RNA isolation kit, if you want ribosomal RNA you can perform a gradient centrifuge (with sucrose for instance) and then isolate the enriched eluted fractions and later peform the RNA isolation (Ref: Liang S, Bellato HM, Lorent J, Lupinacci FCS, Oertlin C, van Hoef V, Andrade VP, Roffé M, Masvidal L, Hajj GNM, Larsson O. Polysome-profiling in small tissue samples. Nucleic Acids Res. 2018 Jan 9;46(1):e3. doi: 10.1093/nar/gkx940)
What sequencing technique will you use?? And in what kind of RNA are you interested?
Best regards!
Francesc
  • asked a question related to Gene Expression
Question
5 answers
In a gene expression analysis since RQ usig delta Cq method depends on reaction efficiency do I'll have to make a standard curve every reaction?
Relevant answer
Answer
Yordan Manasiev , thank you!!
  • asked a question related to Gene Expression
Question
1 answer
Hi researchers,
I plan to examine the effects of a natural product on the cervical softening. Cervical softening is controlled by the expression of 3 genes. I can determine the extent of softening by measuring the expression of these 3 genes. Can I use cervical epithelial cell lines as models to measure the gene expression of those 3 genes? How can gene expression be measured in cell lines? Also, how can I expose the cell lines to these natural products (raspberry tea leaf)?
Any suggestions/ alternate methods/ ideas are much appreciated!
Relevant answer
Answer
You can extract RNA from the cell lines (treated group and control) and then do qPCR for the expression of your three genes of interest. For your natural products use a chlorofom:methanol:water extraction and use a rotary evaporator to dry before resuspending in a small volume of DMSO. You can then add a volume of that to your cell culture media. As for how good your cell line will be for modelling cervical softening, I cannot tell. I advise using a positive control.
  • asked a question related to Gene Expression
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 Gene Expression
Question
1 answer
In relavtive gene expression analysis, we have to average the DCT values of the control gene and then subtact the DCT values of test genes with this.
But my control gene DCT values among replicates are both positive and negative. So when I average them, do they actually hinder the result.
Relevant answer
Answer
Of course it can. Why should this be a problem?
  • asked a question related to Gene Expression
Question
4 answers
I would like to confirm PCR Results, so i thought that "knock out" of gene of interest will work. Who support this? If someone has different aspect would you please share?
Relevant answer
Answer
If you have a knockout-animal, you could try it.
However, in knockout animals you usually don't remove the entire gene, but have a transcript that cannot be translated into a full protein.
I am also wondering:
Why would you want to confirm a PCR with a knockout-animal? You could just use a different set of primers for a different region of the cDNA and/or perform Western Blot with an appropriate antibody...
  • asked a question related to Gene Expression
Question
4 answers
Dear researcher,
Please can you advise any software that can be use for comparison of gene expression of RT-PCR result?
Relevant answer
Answer
Thanks for your response. I have sent you a message. Regards
  • asked a question related to Gene Expression
Question
3 answers
We have extracted RNA from our fungal strain, growing in three different carbon source growth conditions. We have received the RNA-Seq data and have carried out different gene expression analysis (DESeq) between either two of the growth conditions.
Now we are interested in the absolute gene expression levels across all the growth conditions apart from DESeq.
I have the raw hit-counts files ready in a table, first column is gene ID, 2-10 columns are the three replicates of condition 1, condition2 and condition3, respectively.
The next step would be normalization of the read counts and generate the absolute gene expression levels. However, I have limited knowledge of R, in this case, can I do it manually or have to use R to do that? Is there any package (including normalization) I can use? How can I generate a table or even a plot such as heatmap of the top 10 genes?
Thank you very much. Any hint or guide will be very much appreciated.
Relevant answer
Answer
Hello Tiantian Fu,
DESeq2 can generate such files, usually the best one is VST normalized table, where you plot each gene alone with very little variations among samples.
Regarding the top 10, they are the 10 genes with the lowest FDR values from the DESeq2 results table, then you can go back to the VST normalized table and calculate the Z scores of the top 10 genes. After that you can use GraphPad prism to plot a heatmap.
Hope this helps
  • asked a question related to Gene Expression
Question
4 answers
I want a gene to express in a pulsed pattern, not continuously, resulting in stimulation intervals. Any expression system can be applied to facilitate this pattern? Thank you for your suggestions!
Relevant answer
Answer
Use a promoter from one of the circadian rhythm genes? Or you could try a quorum sensing based repressilator.
  • asked a question related to Gene Expression
Question
1 answer
Please in this type of scenario how is the discussion of my result going to be?
Is it in favour of downregulation or upregulation
Relevant answer
Answer
Certainly! Here's a more human-friendly version:
When we talk about gene expression, we often use a measure called the Ct value in a technique called quantitative PCR. This value tells us at which cycle a sample's reaction shows the detection of a specific piece of genetic material. A lower Ct value means there's more of the target genetic material, and a higher Ct value suggests less or some issues.
For something like SARS-CoV-2, the virus responsible for COVID-19, the Ct value can be linked to the amount of viral genetic material present. So, if both a control group and a treated group show an increase in gene expression based on their Ct values, it seems like there's more viral genetic material in both.
However, the fold change, which compares the gene expression of the treated group to the control group, might show a decrease. This could mean that, relatively speaking, the gene expression in the treated group is lower when compared to the control group or a reference gene. There are several reasons why this might happen, including various biological factors or issues in the PCR process.
In simpler terms, even if the Ct values go up in both groups (indicating more viral genetic material), the fold change could go down, suggesting a relative decrease in gene expression. This could be due to a range of factors that affect the PCR process.
Citations:
  • asked a question related to Gene Expression
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 Gene Expression
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 Gene Expression
Question
2 answers
Hello everyone,
We aim to study the effect of certain extract on immunity. In other hand, we want to know whether our extract can boost immunity in human?
So, in first step, we should choose the suitable human cell line and then study immune activity and also immune-related gene expression.
I would be grateful if anyone could guide me or help me with other suggestions for the study.
Relevant answer
Answer
Dear Malcolm Nobre,
Thank you very much for your time and valuable answer.
Are there other suitable cell lines for this study? I found a cell line named NK-92. Do you think it is common to study this type of cell line, especially for my goal?
The best,
  • asked a question related to Gene Expression
Question
1 answer
I would like to know more on their advantages, disadvantage and their use in gene expression analysis.
Relevant answer
Answer
Describe different methods of RNA interference.
Introduction
RNA interference (RNAi) is a biological process in which RNA molecules are involved in sequence-specific suppression of gene expression by double-stranded RNA (dsRNA), through translational or transcriptional repression. RNAi is a powerful tool for gene silencing and can be used to study gene function and develop new therapies.
Different methods of RNA interference
There are a number of different methods of RNAi, but they can be broadly divided into two categories:
  • RNA-mediated RNAi
  • DNA-mediated RNAi
RNA-mediated RNAi
RNA-mediated RNAi involves the introduction of dsRNA into a cell. The dsRNA is then processed by enzymes into small interfering RNAs (siRNAs). siRNAs are short, double-stranded RNA molecules that are about 21-25 nucleotides long. siRNAs bind to messenger RNA (mRNA) molecules that are complementary to their sequence and cause them to be degraded. This prevents the mRNA molecules from being translated into proteins.
There are a number of different ways to introduce dsRNA into cells. Some of the most common methods include:
  • Transfection: Transfection is a process in which dsRNA is introduced into cells using a variety of methods, such as lipofection, electroporation, or calcium phosphate precipitation.
  • Viral vectors: Viral vectors can be used to deliver dsRNA into cells. Viral vectors are modified viruses that can infect cells and deliver their genetic material.
  • Naked dsRNA: Naked dsRNA can be introduced into cells by injecting it directly into the cytoplasm.
DNA-mediated RNAi
DNA-mediated RNAi involves the introduction of a DNA sequence into a cell that encodes a short hairpin RNA (shRNA). shRNAs are double-stranded RNA molecules that are about 21-25 nucleotides long. shRNAs are processed by enzymes into siRNAs, which can then silence gene expression.
DNA-mediated RNAi is a more stable and long-lasting method of RNAi than RNA-mediated RNAi. This is because the DNA sequence can be stably integrated into the genome of the cell.
There are a number of different ways to introduce DNA sequences into cells. Some of the most common methods include:
  • Transfection: Transfection is a process in which DNA is introduced into cells using a variety of methods, such as lipofection, electroporation, or calcium phosphate precipitation.
  • Viral vectors: Viral vectors can be used to deliver DNA into cells. Viral vectors are modified viruses that can infect cells and deliver their genetic material.
  • Retroviruses: Retroviruses are a type of virus that can integrate their genetic material into the genome of the cell. Retroviruses can be used to deliver DNA sequences into cells to create stable RNAi constructs.
Applications of RNAi
RNAi has a wide range of applications in research and medicine. Some of the most common applications of RNAi include:
  • Gene silencing: RNAi can be used to silence gene expression in cells. This can be used to study gene function and develop new therapies.
  • Functional genomics: RNAi can be used to study the function of genes on a large scale. This is called functional genomics.
  • Drug discovery: RNAi can be used to identify new drug targets. This can be done by silencing genes and observing the effects on the cell or organism.
  • Gene therapy: RNAi can be used to deliver gene therapy to cells. This is a type of therapy that uses genes to treat diseases.
Conclusion
RNAi is a powerful tool for gene silencing and has a wide range of applications in research and medicine. RNAi is still a relatively new technology, but it is rapidly evolving and has the potential to revolutionize the way we study and treat diseases.
  • asked a question related to Gene Expression
Question
3 answers
I want to prove 3 transcription factors from a complex to regulated a gene's expression. How can I prove these three proteins interaction.
Relevant answer
Answer
Proximity labeling (BioID-TurboID biotin labeling) in an expression system or co-IP proteomic would be helpful...
  • asked a question related to Gene Expression
Question
5 answers
Neurons were treated with four different types of drugs, and then a full transcriptome was produced. I am interested in looking at the effects of these drugs on two specific pathways, each with around 20 genes. Would it be appropriate for me to just set up a simple comparative test (like a t-test) and run it for each gene? Or should I still use a differential gene expression package like DESeq2, even though only a few genes are going to be analysed? The aim of my experiment is a very targeted analysis, with the hopes that I may be able to uncover interesting relationships by cutting out the noise (i.e., the rest of the genes that are not of interest).
Relevant answer
Answer
Heather Macpherson oh yay that is much better. I think edgeR or limma would be highly appropriate to process your data. The edgeR and limma user guide is an excellent resource and has many tutorials on its proper use. as Jochen Wilhelm explained very well you will not want to subset. In edgeR and limma you can filter by experiment which would require a design matrix. i would also generate a contrast matrix for the group comparisons. After your groupwise comparisons you can subset as you like. highlight those genes in a volcano plot or smear plot. If you have not done this before, I would highly suggest starting from homer step 1 and then directly to the edgeR user guide vignette. Good luck!! http://homer.ucsd.edu/homer/basicTutorial/index.html
  • asked a question related to Gene Expression
Question
1 answer
I am would like to know if there is any optimized expression cassette for penecillin or beta lactam antibiotic genes or vancomycin.
i have gone through many research paper but most of the antibiotics have the cluster gene expression system and but there is no such expression cassette for it to introduce inot plasmid for expression in bacteria or Archaea.
you suggestion would be appriciated in this matter.
Relevant answer
Answer
Most standard cloning vectors have an ampicillin resistant gene (which is beta-lactamase). So those are nearly universal.
Unless you are performing protein purification for biochemical analysis, you don't require an optimized cassette. Most the naturally found genes for resistance will express adequately to confer resistance. So you just PCR out the genes from any plasmid carrying that resistance marker.
  • asked a question related to Gene Expression
Question
6 answers
I want to open a dataset of gene-trait association using:
data = read.table(file.choose(), header = T)
attach(data)
View(data)
But instead of column name (510 traits), these headers are shown:
na, na.1, na.2, ..., na.509
Please find the attached data (ethical point: this data is free access from https://maayanlab.cloud/Harmonizome/dataset/dbGAP+Gene-Trait+Associations)
Thanks
Relevant answer
Answer
Source: Artificial Intelligence Tools
There are a few reasons why column names might be converted to "na.xx" when reading headers in R.
One reason is that the data source does not actually contain headers in the first row. If this is the case, you can use the read.csv() function with the header argument set to FALSE.
Another reason is that the headers are not formatted correctly. R expects the headers to be on a single line, separated by commas or whitespace. If the headers are not formatted correctly, R may assign default names like "na.xx" to the columns.
Finally, it is possible that there are special characters or problematic characters in the header names. R may struggle to read these characters, which can cause the header names to be converted to "na.xx".
Here are some tips for resolving the issue of column names being converted to "na.xx" when reading headers in R:
  • Check the data source to ensure that it actually contains headers in the first row.
  • Use the read.csv() function with the header argument set to TRUE if the data source does contain headers in the first row.
  • Check the formatting of the headers to ensure that they are on a single line, separated by commas or whitespace.
  • Remove any special characters or problematic characters from the header names.
If you are still having trouble reading the headers in your data, you can try using the readLines() function to read the first row of the data file and then use the names() function to assign the headers to the columns.
Here is an example of how to do this
# Read the first row of the data file headers <- readLines("my_data.csv", n = 1) # Assign the headers to the columns names(data) <- headers # Read the rest of the data file data <- read.csv("my_data.csv", header = FALSE)
This should ensure that the column names are read correctly, even if they contain special characters or problematic characters.
  • asked a question related to Gene Expression
Question
5 answers
what is the reason for different CT values between technical duplicates in house keeping gene "human beta actin" in real time qPCR?
I do gene expression analysis. I have 4 genes and 1 house keeping gene which is beta actin. for all the genes, except beta actin, there is no difference between technical duplicates higher than 1. all the conditions are the same. but always beta actin duplicates show 3 or higher differences among duplicate CT values. all the samples are blood from different sources, including glioma blood samples or normal or...
our device is qrotor of qiagene.
and we use without ROX sybr green master mix.
If this problem was due to pipetting error, it must happen for all the genes, because the same operator in the same condition did all the genes. but now the problem is just for beta actin!!!
can anyone help me find the reason for this problem?
Relevant answer
Answer
I would suggest triplicates but I can't make suggestions about the variation in CT for just one set of primers.
  • asked a question related to Gene Expression
Question
1 answer
Doing research on MSS-CRC and have shortlisted a few targets. Was wondering if there are any public datasets that looks at gene expression of genes in MSS-CRC compared to other types of CRC and normal samples? Or how else can I compare the gene expression of my shortlisted genes in MSS-CRC vs normal patients/samples?
Relevant answer
Answer
Hi Daryl
you can test GEO profile (https://www.ncbi.nlm.nih.gov/geoprofiles/) to search for the kind of datasets you want to compare.
fred
  • asked a question related to Gene Expression
Question
4 answers
Which is more important protein or gene expression?
Relevant answer
Answer
  • asked a question related to Gene Expression
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
Finally figured out what's causing low Cts. I'll share it here.
It was the Extension temp. I was using a 3-step protocol (separate extension at 72C from annealing) according to the manufacturer's suggestion for primer Tm < 60C. The 18S ref gene did not have any problem with the 3-step, only the GOIs. After tweaking cDNA input, primer conc., and annealing temp without success, I decided to revert to the standard 2-step qPCR protocol (annealing/extension at 60C), and then the Cts all went down to 20's. This was a surprise though since I've read that the 3-step protocol is supposed to increase amplification efficiency, with Taq operating at its optimal temp. How come a single annealing/extension temp was better?
Thanks to all those who suggested answers.
  • asked a question related to Gene Expression
Question
3 answers
I am not a bioinformatician and don't know much about RNA-seq data analysis. I know that Bulk RNA-seq data must be normalized before differential analysis because differences in starting materials for RNA-seq need to be corrected. The starting materials of scRNA-seq are single cells, and the current normalization method is to assume that the total count of all genes in each cell is equal. But in fact, the overall expression level of each cell is not necessarily equal. Therefore, when comparing the expression level of a gene in the same type of cells that have undergone different treatments, wouldn't it be more reflective of the actual level of the gene expression to directly compare the denoised raw data without normalization? For example, by analyzing the sequencing raw data of C. elegans muscle cell at different ages, we can see that the gene Cox-4 is significantly downregulated with age (see the attached picture), but after normalization, it may no longer show such an obvious downregulation. I'm not sure if I'm correct. I'd appreciate it if anyone could answer my question.
Relevant answer
Answer
In single-cell RNA sequencing (scRNA-seq) data analysis, the preprocessing steps, including denoising and normalization, are essential to ensure that the data is suitable for downstream differential expression analysis. While it may seem intuitive to directly use denoised raw data without normalization for differential analysis, this approach is generally not recommended for several reasons:
  1. Library Size Variation: In scRNA-seq data, cells can have different total read counts due to technical variations, cell size differences, or other factors. This variation in library size can lead to biases in differential expression analysis. Normalization is essential to adjust for these differences.
  2. Scaling Factors: Normalization methods, such as library size normalization (e.g., TPM or FPKM) or more specialized methods for scRNA-seq (e.g., scran or scater), take into account the differences in total read counts and apply scaling factors to make the data comparable between cells. This ensures that you are comparing expression levels relative to the total expression in each cell.
  3. Statistical Robustness: Normalization helps to reduce technical variability while preserving biological variability. It ensures that the statistical tests used for differential expression analysis are more robust and reliable. Directly comparing raw counts can lead to spurious results due to technical noise.
  4. Batch Effects: Even if you denoise the data, you may still have batch effects or other sources of systematic variation that need to be corrected through normalization to identify true biological differences.
  5. Biological Interpretation: Normalized data provides more meaningful results for biological interpretation. The goal of differential expression analysis is to identify genes that are differentially expressed in biologically meaningful ways, not just those that have different raw counts.
In your example with C. elegans muscle cells at different ages, it's important to normalize the data to account for differences in library size between cells. This ensures that any observed changes in gene expression are more likely to reflect true biological differences rather than technical artifacts.
That said, you should still perform differential expression analysis on the normalized data. If a gene like Cox-4 is significantly downregulated with age, the normalization should help highlight this effect by reducing the influence of technical factors.
In summary, while it may seem reasonable to directly use denoised raw data for differential analysis, it's generally recommended to perform proper normalization to account for technical variations and ensure the robustness and biological relevance of your results in scRNA-seq data analysis
  • asked a question related to Gene Expression
Question
1 answer
I'm planning an experimental design to investigate the gene expression of mussels under thermal stress conditions, to compare their responses.
The experiment includes a control tank and a treatment tank, with enough mussels to sample 4 time points (60 mussels per time point per treatment).
In our facilities, I have two separate recirculation systems (with heaters and coolers) with two tanks each.
Since the two tanks of one recirculating system are connected they cannot be considered as isolated tanks and therefore they are not replicates. I'm planning to distribute the control samples between the two tanks of system 1 and the treatment samples between the two tanks of system 2, and at each time point sample from both tanks of one system and pooling them together (e.g. 30 samples from tank 1 and 30 samples from tank 2 of the control system)
My question is: do I need to have tank replicates for this experiment? Or can I sample three groups of 60 mussels per timepoint and per treatment between the two tanks of one system? i.e. have sample replicates within the control temperature and the treatment temperature tanks at each timepoint?
Thanks!
Relevant answer
Answer
I assume the two tanks in system 1 cannot be independently temperature controlled and the same for system 2.
If the two systems have identical tanks and control/ciiculation mechanisms, then you could assume no tank effect and hence no need for tank replicates. If there are differences you could do a repeat of the whole experiment swapping the control and treatment tank system.
  • asked a question related to Gene Expression
Question
2 answers
To check the expression of JAK2, STAT3, SRC and indoleamine 2 and 3 dioxygenase genes in the blood samples of people who do not have any immunological or hematological diseases, is the best way to isolate PBMC and check gene expression in PBMC or Can this be done in whole blood?
Relevant answer
Answer
Dear Morvarid,
There are a number of methods to extract RNA. If you are intending to use an RNA extraction kit, any reliable company offers high quality kits. However, more conventional methods also result in high quality RNA. Depending on your aim (whether you test blood cells in an experiment or not) RNA can be extracted from blood and from isolated PBMC. In order to have high quality RNA, you must have highly viable cell pellet. Either extract RNA right after spinning your cells, OR snap-freeze using liquid nitrogen and store in a reliable -80 freezer until RNA extraction.
  • asked a question related to Gene Expression
Question
4 answers
I will perform gene expression for mir27a and mir155
so I need to check my primers by any suitable software
thank you
Relevant answer
Answer
Hi,if You have to check micro RNA then you check NCBI by- Medline - publication index - Association extractor - Term extractor - miRBase parser - miRBase.
The majority of miRNA publication association in mirpub database have discovered by applying text mining techniques on titles, abstract and full text of all available MEDLINE publication.
  • asked a question related to Gene Expression
Question
2 answers
hello i am a new master's student in an immunology lab and I have been assigned a topic to establish a stable cell line for swine interleukin 2 gene expression, but I do not know how to start my experiment, if someone can help me in this I will be highly obliged. Thanks
Relevant answer
Answer
It's a very fundamental question. First, a appropriate cell line is essential for constructing a stable transgenic cell line. According to the cell line, an efficient method to transfer gene to the cell line is need, such as expression plasmid, lentivirus. Finally, the cell lines that stably expressed swine interleukin 2 can be obtained after a series of screening.
An article may be helpful as follow.
  • asked a question related to Gene Expression
Question
2 answers
I want to use yeast as an expression system. So, I wanted to ask if Debaryomyces hansenii could be used for the same.
Although, Saccharomyces is a model system for gene expression studies. Can we also use Debaryomyces hansenii? Is this system suitable for plant gene expression studies?
Relevant answer
Answer
Thank You Anubhav Singh Nahar Sir for your response.
  • asked a question related to Gene Expression
Question
2 answers
Some genes that resulted from my differential gene expression analysis don't have corresponding ENTREZ ID and I want to perform KEGG pathway analysis but most tutorials analyze data with no missing ENTREZ ID
Relevant answer
Answer
Were the annotations to the gene IDs result in hypothetical protein? Did you use KEGG mapper? If not you can find relevant EC numbers or use uniport id retrieval (https://www.uniprot.org/id-mapping). If nothing works, you can search for homologus genes and their IDs try to map your pathway.
  • asked a question related to Gene Expression
Question
3 answers
How to get the RNA-seq data for gene expression analysis from SRA data?
Relevant answer
Answer
If you don't want to use sra-tools, you can search for the SRA id at https://www.ebi.ac.uk/ena/browser/ and download the fastq files directly.
  • asked a question related to Gene Expression
Question
2 answers
Hi,
I have Illumina RNA sequencing data for viral infection only or viral treated. I would quantify the viral gene expression, but the total reads counts is various between library. Please see fig. I am wondering do I need to downsample the total reads to 5 M or is it ok to quantify using the total viral mapped reads ?? Thanks in advance
Relevant answer
Answer
Great, thanks for your answer. But the viral genes have different lengths. I am wondering if I use the EDgR package. It will not take into consideration these differences. Thanks in advance
  • asked a question related to Gene Expression
Question
13 answers
Is it necessary to measure the quantity of single-strand cDNA after RT-PCR and before qPCR? TaqMan Universal PCR Master Mix manual (TaqMan Gene Expression Assay) (at picture) describes the desired quantity of template (1-100 ng) per reaction. So, I should measure the quantity of cDNA after RT-PCR (for instance, on Nanodrop spectrophotometer) and bring this amount to the desired range if needed?
Relevant answer
Answer
Use the same _amount_ of RNA in each reaction, wherever possible. Don't dilute your RNA prior to the reaction, there's no point. Dilute it as part of the reaction (i.e. add X ul RNA + Y ul H2O).
A lot depends on the max vol your kit can take, but if I know that I can use say...8ul of RNA total, and most of my RNAs are 200ng.ul-1 or above, I'll use '1600ng' as my target. If any of my RNA is less than 200ng.ul-1 (so I can't physically add 1600ng without exceeding the 8ul cap), I'll just add 8ul.
Most kits can take anything up to about 2ug, so that's your upper limit. 1ug would be fine, for example.
Once you've made your cDNA, dilute all of it to the same extent (1/10 dilution for everything). Definitely dilute it, though.
You're not just diluting out the target molecule population, you're also diluting out the cDNA synthesis buffer components so they don't interfere with your PCR reaction.
  • asked a question related to Gene Expression
Question
4 answers
Hi all , I am confirming gene expression using RT-PCR. If a gene is down-regulated does that mean it is not expressed in the strain?
Relevant answer
Answer
Yes, that’s correct. When a gene is down-regulated, it means that the expression of the gene is decreased. However, it does not mean that the gene is not being expressed at all. The product it encodes for is also decreased as a result of the decreased expression of the gene
  • asked a question related to Gene Expression
Question
7 answers
Hi All
Due to the high cost of RNA-seq per sample. Do you think that it will be correct if I bulk three-four biological replicates and send this bulk for RNA-seq?
Relevant answer
Answer
Dear all,
thanks for the insightful commentaries of my colleagues. However, I beg to differ in some detail as this problem goes a bit deeper. (Just my 0.02$)
1. Throwing three samples into one is, of course, a bad idea because you waste ressources. Why not make three differently barcoded libraries and then send them for sequencing on one lane. You do not loose information and you can always ask for more reads if your sequencing depth is not sufficient. Thus you save in sequencing costs but keep all the options.
2. Do not make technical replicates. If you master the technique they will be +/- identical. If you have technical problems no replicate will help you anyway.
3. If you run biological replicates I wouldn't use the classical R-programs. Most assume that there is a "true" value that you can't measure because of random variation in your method/sample. However that is not exactly what happens in nature. Imagine you derive three transgenic cell lines with an inducible transcription factor to find target genes. Now you compare 3 times TFon/TFoff. You get following values:
TFon TFoff
geneA
sampleA: 1000 100
sampleB: 100 10
sampleC: 10 1
It is clear that geneA is very interesting. However, if you define sampleA,B,C as triplicate most analysis programs will throw this gene out because base-line expression has a higher variation as the overall difference between on/off.
Alas, geneA may be a perfect and important target gene as you do not control the overall concentration of the transcripiton factor in the transgenic cells. So it may be perfectly ok that you see this large variation in base expression. Fold-change is what counts here. In biology a value very frequently depends on more than one factor and not all can be controlled. Classical statistics fails in this cases.
Therefore I'd recommend to run barcoded libraries - evaluate each one individually and look for the intersection of genes that come up as interesting in all three instances. Then follow up on these.
In the end no statistics can replace the good old biological confirmatory experiment anyway (although "wet" biology seems to be out of fashion nowadays).
Good luck with your experiment.
Best
Robert
  • asked a question related to Gene Expression
Question
1 answer
Does anyone know of any vector backbones that can co-express two gene of interest from divergent promoters (or a single bidirectional promoter that has comparable activity in both directions)?
Relevant answer
Answer
search for bicistronic plasmids in addgene
  • asked a question related to Gene Expression
Question
3 answers
when Real-time PCR consider a high throughput technique? Is gene expression analysis by qPCR give a low throughput data?
Relevant answer
Answer
thank you
  • asked a question related to Gene Expression
Question
5 answers
I have multiple sets of RNA-seq data and I want to compare gene expression between control and treated groups. My interest extends beyond differentially expressed genes; I also want to identify non-differentially expressed genes. I understand that Log2FoldChange and p-adj are commonly used to define differentially expressed genes. Alternatively, genes that fail to meet the criteria for differential expression are considered non-differentially expressed.
However, classifying a gene as non-differentially expressed does not definitively indicate that the RNA-seq data confidently establishes the absence of changes in gene expression. For instance, this could be attributed to substantial within-group variation or low gene counts that hinder unambiguous determination of expression levels. So, how can I effectively distinguish truly non-differentially expressed genes from those exhibiting significant within-group variation or yielding very low counts? Are there any software packages available for this purpose? Alternatively, are there established statistical methods or standards that can guide me in this regard?
Relevant answer
Answer
Chat-GPT really is a jabber-box. Context must be critically reworked by a person knowing the topic. Answers like the one posted by Rana above are not only not useful but also potentially misleading, not to say wrong. But most relevantly, it has nothing to do with a scientific interaction of researchers.
  • asked a question related to Gene Expression
Question
2 answers
I have 3 genes (eg. Gene A, B, C) that I am looking at expression levels for in the same dataset (same 100 genomics files, so samples are labeled 1-100). I have classified each sample as high or low based on expression level (eg, sample 1 is high expression for gene A, low for gene B, and low for gene C), and I want to statistically compare if the gene expression categorization is statistically different for each gene or not. (eg., is there a statistically significant overlap between which samples are high expression and which samples are low expression between Genes A, B, and C, or not?)
Thanks!
Relevant answer
Answer
This would be Chi-square or Fisher's exact test on the contingency table like below:
High Low
gene_A 500 500
gene_B 450 550
gene_C 300 700
  • asked a question related to Gene Expression
Question
4 answers
Greetings!
I have an issue that drives me crazy this evening...
I have a list of gene vectors, downregulated in different transgenic plants and I want to make a Venn diagram to visualize it and to show the intersections between plants.
But! The results from any package I used (in R) gaves me something like this (the uploaded picture 1)...
What's bothering me:
1. The numbers on "clear" (not intersected) parts of a diagram are lower, than the gensets I have. And I tried to use factor instead of character vectors, to remove possible duplications, to remove symbols (like space) that could cause software misunderstanding - all gaves me nothing... same result.
2. The intersection of vectors is not true - on the picture you can see that the intersection of 2 datasets (of 365 and 154 genes) - is 1133 genes!! How could that be?
The manual usage of intersect function on the same dataset gaves pretty correct results.
Maybe I am misunderstanding about Venn diagrams? Because in a web I found many examples of such strange mistakes - on the second picture from Datanovia you can see that the intersection of the red elliplse (of 58) and yellow (of 144) is 66!
It seemes logical to me that the intersection of 2 vectors cannot be greater than the length of a smaller vector. What am I doing wrong or misunderstanding?
Relevant answer
Answer
I believe Rob is correct.
Since you are using the intersect function, the numbers in your figure (e.g. 365 and 154) are the number of genes without any intersection.
The total genes of each set (e.g. OE21) will be the sum of all the numbers in each intersection + genes with no intersection. I couldn't do full sum for you as the core intersection number is missing.
  • asked a question related to Gene Expression
Question
2 answers
I have a results from qpcr experiment for siRNA after transfection, how to know the effect of siRNA in Knock down from qpcr results
Relevant answer
Answer
  • asked a question related to Gene Expression
Question
1 answer
We checked qRT-PCR-based gene expression in our homozygous knockout cell line and mostly observed the downregulation of targets compared to wild-type control cells. We recently performed a transient knockdown using siRNAs against the same RBP in the wild-type control cell line, but the target genes were upregulated. What could be the reason behind these opposite effects in homozygous Knockout cell line vs transient knockdown in wild-type cells?
Relevant answer
Answer
Kishor Gawade It is not uncommon to get such results in case of siRNA.
Pls refer to the following comparative study which presents a protocol to avoid false positive results.
Hahn P, Schmidt C, Weber M, Kang J, Bielke W. RNA interference: PCR strategies for the quantification of stable degradation-fragments derived from siRNA-targeted mRNAs. Biomol Eng. 2004 Nov;21(3-5):113-7. doi: 10.1016/j.bioeng.2004.09.001. PMID: 15567105.
  • asked a question related to Gene Expression
Question
2 answers
Which tools or methods are used to compare the relative gene expression of real-time PCR value other than the Livak method??
I have values of CQ of both gene and internal reference. I want to know which methods compare the gene expression among the samples. The relative expression between samples.
I have used the Livak method. But I want to know other methods or tools.
Please guide me.
Thank you
Relevant answer
Answer
Hello! I like to use the "qpcR" R software to fit curves, but it isn't necessary. you can use the "delta delta Ct" method. Here is the gold standard instructions for qPCR experiments. Good Luck! https://www.gene-quantification.de/national-measurement-system-qpcr-guide.pdf
  • asked a question related to Gene Expression
Question
3 answers
For heatmap generation, how to retrieve required data from SRA dataset?
Relevant answer
Answer
If you only care about normalized data to make a heatmap, you can use recount3. NCBI itself is also starting to offer raw and normalized RNA-Seq counts (beta version). You can also take a look at GEOexplorer
  • asked a question related to Gene Expression
Question
6 answers
Hello everyone
I have an exon sequence of a gene and I would like to get its cDNA for primer design to test the expression of that gene
could you please help to provide any tool that can convert Exon sequence to cDNA?
Relevant answer
Answer
OK, given the context of the research, you are going to want to start by using regular PCR to see if the region of the gDNA can be amplified at all.
In-silico predictions often have errors. You'll save yourself a LOT of stress if you can show that the genomic region exists in the DNA.
Besides, you'll need to clone that region of the genome to make a standard curve for your qPCR. It's a "win win" to check for the gDNA first.
  • asked a question related to Gene Expression
Question
2 answers
How does the environment affect genetic factors and gene expression and environmental factors that can influence gene expression plants?
Relevant answer
Answer
Environmental factors such as soil nutrients, temperature, water availability and light intensity influence the genetic and chemical diversity of plant populations. These environmental conditions can exert strong selection pressures; they could even determine the evolutionary course of plant populations. Gene expression can be altered by environmental factors such as food, drugs or exposure to toxins. These changes can range from small to so significant that certain genes in our system can be turned off or on when they are supposed to be the opposite way. Environmental factors such as soil nutrients, temperature, water availability and light intensity influence the genetic and chemical diversity of plant populations. Genetic factors as well as local conditions affect the growth of an adult plant. The growth of an animal is controlled by genetic factors, food intake, and interactions with other organisms and each species have a typical adult size range. Primary abiotic factors are light, temperature, water, atmospheric gases, and ionizing radiation, influencing the form and function of the individual. For each environmental factor, an organism has a tolerance range in which it is able to survive.
  • asked a question related to Gene Expression
Question
2 answers
Thanks.
Relevant answer
Answer
You need whole protein to analyze gene expression. How is just one unit help in studying the expression, when you never know how many regulatory sites might be present in the other half.
  • asked a question related to Gene Expression
Question
1 answer
The F1 hybrids of these lines tend to show either a bias in expression level or an allelic level bias compared to the transcriptome of the selfed material. The former means that the F1 is similar to one parent in the expression level of a gene, while the latter means that the F1 expresses more gene sequences from one parent due to genomic differences between the parents. However, in my transcriptome analysis of F1 hybrids from one crop, I found that about 98% of the genes were expressed simultaneously and in almost equal amounts from the parents' chromosomes. However, in terms of the total level of gene expression, F1 showed a large amount of biased parental expression.
The following is the general course of my analysis:
(1) Transcriptome data from parents and hybrids were cleaned and compared to the reference genome.
(2) Extract SNP information using bcftools and filter for heterozygous mutations in the parents and pure mutations in the hybrids.
(3) Ensure that the parental mutations are biallelic and that the parents do not have the same base at the same locus.
(4) Ensure that all biological replicates are identical in type of mutation.
(5) Match SNPs to gene regions and add up the counts of all SNPs within a gene.
(6) Perform differential expression analysis using DESeq2 to determine if there is allelic preference for gene expression in the hybrid from the parents.
My question is whether the relationship between expression levels and allelic levels is clear, or whether my analysis process is so flawed that it does not support the current results.
Thank you all!
Relevant answer
Answer
The results of the transcriptome analysis of F1 hybrids suggest that the relationship between expression levels and allelic levels can be complex. However, the researchers were able to identify some of the factors that contribute to this complexity. This information could be used to develop new methods for predicting the expression of genes in F1 hybrids.
There are a number of factors that can contribute to the complexity of the relationship between expression levels and allelic levels. One factor is that the expression of a gene can be affected by a number of different factors, including the environment, the developmental stage of the organism, and the presence of other genes. Another factor is that the expression of a gene can be influenced by the interaction of different alleles. One allele may be dominant over another, meaning that it is expressed more strongly.
  • asked a question related to Gene Expression
Question
6 answers
Hello everyone,
I am running a qPCR assay. I chose gradient temperature option for each of my primer to get the best conditions the amplification happens (without heterodimers- NA in negative controls). However, I have seen that my housekeeping gene and one of my target gene have different annealing temperature. Can I run another qPCR set-up just for this gene by choosing gradient temperature option ? For instance; my gene in question in a row with 54C and housekeeping gene in a row with 60C. I think as far as the machine reads the signals at the same time, it won't pose a problem but I just want to make sure.
Many thanks,
Tuba
Relevant answer
Answer
Bertrand Cornu Can Kiessling Audrys G. Pauža Mohamed Khashan Dino Santos Matias Thank u all. After many trials I have decided to use single temperature. I have to admit that still qPCR experiment is not so objective to me (changes according to conditions very easily). However, since everyone use the same method, and what matters is to compare the mrna level for the same protein, I guess it is fine.
  • asked a question related to Gene Expression
Question
3 answers
Hi Dears,
could you please have anyone share the tutorial or model file format for heatmap analysis for gene expression data set. Actually i would like to add additional row information in that.
Thank you very much in advance
Relevant answer
Answer
Heatmap is not an analysis, it is just a visualization method of the results from analysis. There are many tools which can be used to make heatmap in R, python etc. If you have dataframe ready, any visualization tool can be used.
In R pheatmap or heatmap3 are examples.
  • asked a question related to Gene Expression
Question
2 answers
I want to know that, when a heterologous gene is expressed under CMV promoter in mammalian cells, what is the percentage of this heterologous gene's mRNA in comparison to total cellular RNA and total cellular mRNA? Is there any mention of this in the literature?
Relevant answer
Answer
Any transient transfection can be titrated to achieve a desired expression level. The CMV promoter is very strong and will work in most cell types. The mRNA and protein levels for your gene will vary depending on RNA stability, size, translational efficiency, etc. So, by transfecting varying amounts of your expression vector mixed with something inert, like Salmon sperm DNA, you can empirically determine how much you wish to express. For 100mm plate we use 6ug DNA with lipofectamine or fugene. But that can be 100 ng of vector with 5.9 ug inert DNA. Vector can be anywhere from 100ng to 6 ug.
  • asked a question related to Gene Expression
Question
3 answers
i did not find any resonable defination for the DGE
can anyone define this tearm for me please ?
Relevant answer
Answer
When there is differences of more than 2 fold in a gene for a condition is known to be differentially expressed.
  • asked a question related to Gene Expression
Question
3 answers
I am trying to start a new project but I am not familiar with machine learning algorithms. I want to build a predictive supervised model that is able to classify samples into clusters. This clusters are defined by a gene signature. I basically have gene expression matrices.
I would like to know which type of machine learning is the best in performance and prediction for this type of data and query. I've been looking at Deep Learning but I still can't find which one would fit better.
Relevant answer
Answer
There are several machine learning algorithms that can be used for clustering and classification of gene expression data, including deep learning algorithms such as Convolutional Neural Networks (CNNs), as I mentioned in my previous answer.
Another popular deep learning algorithm for clustering and classification tasks is the Deep Belief Network (DBN). DBNs have been used for gene expression analysis and have shown good performance in identifying gene clusters associated with diseases and other phenotypes.
Other commonly used machine learning algorithms for clustering and classification of gene expression data include:
  1. Support Vector Machines (SVMs): SVMs are widely used in bioinformatics and have been shown to be effective in gene expression analysis.
  2. Random Forests: Random Forests are an ensemble learning algorithm that can be used for classification and feature selection tasks in gene expression analysis.
  3. K-means clustering: K-means clustering is a popular unsupervised clustering algorithm that can be used to identify gene clusters based on similarities in their expression profiles.
Ultimately, the choice of machine learning algorithm will depend on the specific characteristics of your dataset and the problem you are trying to solve. It's recommended to experiment with different algorithms and compare their performance to identify the best approach for your specific problem.
  • asked a question related to Gene Expression
Question
4 answers
I want to ask a technical question, I have treated and non-treated Male and Female RNA seq samples, and I want to do sex-biased gene expression. My concern is that should
I compare the male vs female samples or male vs normal and female vs normal when analyzed for sex-biased gene expression using Dseq2
Relevant answer
Answer
Honestly, DESeq2 takes almost no time to run: it's by far the easiest part of the entire pipeline.
So...do it all. All comparisons.
Male treated vs male untreated -> effects of treatment on males specifically
Female treated vs female untreated -> effects of treatment on females specifically
Male and female treated vs male and female untreated -> sex-agnostic effects of treatment
Male untreated vs female untreated -> sex-specific differences under normal conditions
Male treated vs female treated -> sex-specific differences under treated conditions
male (all) vs female (all) -> treatment-agnostic effect of sex
What you'd hope is that the same genes would crop up under the same sort of comparisons (i.e. the best hits for male treated vs male untreated would be the same as those for female treated vs female untreated), and any genes that bucked the trend by being highly altered in males but not females (or vice versa) would also pop out of the other datasets, allowing you to determine whether the differences reflect sex alone, or are a consequence of sex + treatment.
RNAseq data always gives you far, far more candidates than you can realistically handle, so you can be quite aggressive in your culling of "interesting but not that interesting" candidates, but making multiple comparisons in this manner allows you to interpret your results with more nuance. And it's really quick and easy to do.
  • asked a question related to Gene Expression
Question
3 answers
RNA-seq analysis
Relevant answer
Answer
  • asked a question related to Gene Expression
Question
2 answers
I wish to download TCGA gene expression from XENA browser using the following link:
Yet they don't clearly mention what are the units and the normalization performed. All that is mentioned in units is log2(norm_value+1) and its unclear what is this norm_value. is it RSEM, TPM or FPKM?
If anyone knows and can point me out to a resource that explains how their normalization was performed it would be very much appreciated
Relevant answer
Answer
The TCGA gene expression units in the PANCAN batch effects normalized gene expression data available on the Xena browser are in the form of log2-transformed normalized expression values, where the normalization method used is not explicitly stated.
The normalization method used for the EB++AdjustPANCAN_IlluminaHiSeq_RNASeqV2.geneExp.xena dataset is not specified on the Xena browser website. However, based on the description of the normalization units, it is possible that the normalization was performed using the log2(TPM+1) or log2(FPKM+1) method.
The normalization units, log2(norm_value+1), suggest that the data may have been normalized using a method that produces values similar to TPM or FPKM. The "+1" is added to the value to avoid taking the log of zero, which can cause errors in downstream analysis.
To get more information on the normalization method used, you can try contacting the TCGA or Xena browser support team for more details. Alternatively, you can check the original publications or data sources for the dataset to see if the normalization method is explicitly stated there.
  • asked a question related to Gene Expression
Question
5 answers
want a brief explain for this
Relevant answer
Answer
Just to add a bit more complexity, unless you have very strong molecular evidence to back up the gene model, any PREDICTED introns/exons/promotors/etc are just predictions. Bioinformatics tools use "generally true, most of the time" rules. But if you care about a particular gene, you'll have to spend time either in the literature or at the bench to see the evidence that supports/refutes that model for that specific gene.
The "intron" might not even actually be an intron.
  • asked a question related to Gene Expression
Question
1 answer
I am planning my experiment to target the enhancer region of my gene of interest by sgRNA to reduce gene expression. I need to pick up sgRNA that can go the job.
Prior, I would like to construct a small sgRNA library for CRISPR screening. Could you please advise which company does such requests?
Relevant answer
Answer
  • asked a question related to Gene Expression
Question
20 answers
Hi! I have been calculating fold changes in the gene expression using delta of delta method, and the average of the control values does not equal 1. I asked a colleague who does similar tests regularly, and he says that has always been the case for him. Is that really how it should be, or have I made a mistake somewhere?
Thank you!
Relevant answer
Answer
The indicator (aka "dummy") variable is a variable that takes the values 0 or 1 to indicate the membership to a group. It takes the value 1 if the corresponding response value (the dCt) is from the "treatment group", and it is 0 otherwise.
In the linear predictor, this variable is multiplied by the coefficient coding the treatment effect as the expected difference in the response (the dCt values) between the indicated group (treatment) and the reference group (the group not being represented by an indicator variable; here: the control group). Nothe that the difference in dCt values is called ddCt.
It may be helpful to read some books about (general) linear models.
  • asked a question related to Gene Expression
Question
3 answers
Hi all,
I recently ordered and began culturing a new cell-line that are meant to be a canine line. I was interested in testing their expression of a particular gene, and of course compared this with a common house-keeping gene. I ran qPCR this morning, and gene expression of both my housekeeping gene and my gene of interest are undetectable, despite using canine primer probes.
My colleague believes these cells I have been growing may not be what they are advertised to be - they grow extremely fast (faster than HEK293 in my experience). I will be testing the cDNA I made with both human and mouse primer probes tomorrow to see if these primers work. However, I'm concerned I have done something wrong - I am wondering if maybe the cDNA reaction was set-up wrong? I'm not so sure if this is the case, I check everything off as I go... unless there is something wrong with the heating block... How can I test my cDNA before setting up my qPCR reaction tomorrow? Should I make more cDNA from my RNA? Or, should I go ahead and set up qPCR?
Is there any explanation other than these cells being the wrong species? Perhaps I used the wrong program when doing qPCR? Any advice is helpful - thanks!
Relevant answer
Answer
Oooh this is an interesting one. There's quite a bit here.
Firstly - I don't think you can assume that if mouse or human primers DO work, that means the cells are human/mouse and not canine. You could check the primer sequences against the dog genome with BLAST and see how similar they are.
Second - Sanity check your reaction setup. How much RNA did you put into each cDNA synthesis, and also what kind of primers and how much? How much cDNA template did you put into your qPCR?
Third - yes you can check if your cDNA synthesis worked. Check the concentration using Nanodrop or Qubit. If it's less than about 20ng/uL, your reaction failed. If it's more than that, run it on a 1.5% agarose gel and see what the size distribution looks like. You should see a broad smear from ~50nt up to ~1000nt - if it's very skewed towards low molecular weight fragments, the problem is that your RNA was degraded.
I would avoid wasting further RT or qPCR reagents until you know if your cDNA synthesis worked and what the size distribution is like.
Also - unorthodox suggestion but it just might work - would any of your canine-specific primer sets detect canine gDNA? If so - does anyone you work with have a pet dog? You would be able to extract a modest amount of canine gDNA from a cotton swab vigorously rubbed on the gums of any dog. This would at least let you check whether your canine-specific primers do recognise the dog genome. (If they are mRNA specific primers, you're out of luck here.)
  • asked a question related to Gene Expression
Question
2 answers
How Antisense RNA is used to upregulate gene expression? Please share some specific examples.
Thanks in advance!
Relevant answer
Answer
Thank you Juan Pablo Tosar
  • asked a question related to Gene Expression
Question
4 answers
if gene expression is low in specific diseases based on previous research,
But when I measured the protein expressed by the same gene in blood of same disease by ELISA I found high serum level?
what is the explanation for this difference between gene expression and its protein concentration in blood?
Relevant answer
Answer
Not necessarily. The level of gene expression does not always directly correlate with the amount of protein produced. There are many factors that can affect protein expression, including post-transcriptional modifications, translation efficiency, protein stability, and degradation.
  • asked a question related to Gene Expression
Question
4 answers
I'd like to know that what are the different ways to know/identify whether a particular Gene is expressed or not ?
Few points from my side are :
1) identifying it's corresponding m-RNA transcripts level.
2) identifying the protein that was produced by the expression of that particular Gene.
Any other points ?
Relevant answer
Answer
Hi,
You can do qPCR to check the expression of the target genes.
  • asked a question related to Gene Expression
Question
4 answers
I am trying to extract RNA from frozen liver tissue samples for gene expression analysis. The ratio look good on Nanodrop but when i proceed it on gel it shows degradation (smears).
what does it mean ?
Can someone help ? It will be a great favor.
Relevant answer
Answer
As Katie said, degraded "smear" RNA looks the same as intact RNA in Nanodrop. If you want to prevent RNA degradation from frozen livers, I recommend using RNAlater. If you are using an extraction method such as TRIzol, it is better to use a column method such as RNeasy. And in some cases, using 50% ethanol (instead of 70% ethanol) may increase RNA yields from liver samples.
  • asked a question related to Gene Expression
  • asked a question related to Gene Expression
Question
3 answers
I want to do RTPCR for some biomarkers from the serum. But unfortunately, the RBCs get lysed due to this it turns slightly RED. Does it affect the gene expression or RTPCR RESULTS?
Relevant answer
Answer
@Don Ashley,
Please provide the procedure of purification by Ficoll-Hypaque method.
  • asked a question related to Gene Expression
Question
4 answers
Kindly suggest any research that is based only on in-silico approaches for plant (especially tomato) transcriptome analysis under different stresses. There is a lot of literature that includes gene expression analysis using wet lab experimentations for transcriptome analysis.
Relevant answer
Answer
Hello everyone. I have received results for RNA quantification / RNA seq analysis from a company.
There are large files. Can anyone guide me how to access the required results. Thank you
  • asked a question related to Gene Expression
Question
3 answers
I have read that dd-PCR does not require a housekeeping gene - could anyone explain why? We are trying to design a dd-pcr triplex for avian immune gene expression.
Thank you
Ellie
Relevant answer
Answer
ddPCR (Digital Droplet PCR) is a PCR-based method that partitions the reaction into thousands of tiny droplets, each containing a single copy of the template DNA. The droplets are then analyzed using flow cytometry to determine the number of positive and negative droplets, which in turn reflects the number of copies of the target DNA.
The reason why ddPCR does not require a housekeeping gene is that it is inherently a very sensitive and specific method. The droplet partitioning allows for a high degree of multiplexing, meaning that you can test for multiple genes at once, and the digital nature of the method enables very precise quantification of the target DNA. The high sensitivity of ddPCR means that even low levels of target gene expression can be detected and quantified, which reduces the need for normalization to a housekeeping gene.
That being said, it is always good practice to check for the suitability of the gene of interest as a reference gene and the specificity of the primers, in order to avoid unspecific amplification or cross-reactivity. Additionally, if you are interested in comparing the expression of your target genes across different samples or conditions, it may be useful to include a housekeeping gene as a reference to control for variations in sample quality or RNA integrity.
  • asked a question related to Gene Expression
Question
3 answers
Has anyone compared exogenous gene expression in Thp-1 cells using different promoters? Would EF-1 or CMV be better?
Thanks!
Relevant answer
Answer
It is known that CMV promoter has a tendency to lose its transcriptional activity and this is theorized to be due to gradual methylation at its CpG sites which may lead to low-level expression. In that case EF-1a would be better.
I have attached few papers for your reference.
Best.
  • asked a question related to Gene Expression
Question
3 answers
Hi,
Except humic acid existence in soil samples, are there other reasons to dilute DNA or cDNA concentrations of soil samples prior to performing qPCR assay? And after dilution and qPCR assay, how we can calculate a gene/transcript expression? To clarify what I meant, for example, if we dilute DNA concentration by 5:1, after getting the raw number from qPCR machine, should we multiply the received number by 5 to reach exact gene expression copy number?
Appreciate
Mehrdad
Relevant answer
Answer
You generally dilute samples to 1) preserve some DNA/cDNA for later use and 2) Lessen the concentration of inhibitors. Regarding "if we dilute DNA concentration by 5:1, after getting the raw number from qPCR machine, should we multiply the received number by 5 to reach exact gene expression copy number?", yes you would have to multiply by the dilution factor to get the quantity in the starting tube (this is assuming absolute quantification method via real time PCR). Alternatively, you can also use digital PCR if you have access to get absolute quantification without the use of standards. Some materials for you to look at:
  • asked a question related to Gene Expression
Question
5 answers
Hello, everyone!
As in the title, I understand that plasmids must be transported into the nucleus in order to be expressed. I am wondering if that means the whole plasmid is required to be in the nucleus. Or, actually, the plasmids are degraded in the cytoplasm into small pieces, and some of the pieces contain a signal and gene that are then translocated into the nucleus for expression.
Could you enlighten me, please? It would be nice if you give me with any evidence or paper to support the statement.
Thank you in advance for your explanation!
Nina
Relevant answer
Answer
Generally the entire plasmid is imported into the nucleus, you can often find multiple copies of the plasmid in the cell. Transient expression is often achieved without any sort of integration into the nuclear genome, but it does go away with time. For stable expression you need to select for integrants.
  • asked a question related to Gene Expression
Question
3 answers
Hello scientists, I was hesitant to ask such a question because it is somewhat simple, but in the end there is no shame in the learning process, so, What consumes more energy, the polymerization of coding region or its translation?
and how to calculate the energy required for transcription?
Thanks in advance.
Relevant answer
Answer
If by "polymerisation of coding region" you mean "mRNA synthesis", then honestly, there is no fixed answer to this question (for eukaryotes, certainly).
Many genes have enormous lengths of intronic sequence, all of which must be transcribed, only to then be spliced down to a comparatively small mRNA for translation. Dystrophin, for example, is 2.3 million bases long, but is spliced down to ~14,000 (i.e. 99.5% of the transcribed nucleotides are spliced out and recycled).
Here transcription clearly requires a greater energy input on a 1:1 mRNA:protein basis, whereas other shorter (or intronless) genes might be closer to parity.
However you also need to factor in energy requirements for recruitment of initiation factors, unwinding of the transcriptional start site, scanning, abortive initiation, etc, which will also vary from gene to gene. Some genes are permissive, other are less so.
You also need to factor similar variables to translation: targeting of mRNA, recruitment of translation initiation factors, unwinding and scanning of 5' UTR, etc.
And finally, this all assumes you are comparing mRNA and protein on a 1:1 basis, which is entirely inaccurate. A single mRNA can be translated many, many times, so a large energy investment in one transcript might ultimately pale in comparison to the vast energy investment in making 10,000 proteins from that one transcript. Alternatively, a single mRNA might be degraded without ever being translated even once (some 'immediate early' genes are transcribed continuously, 'just in case', but then degraded if not needed).
If you want to know (more generally) 'what uses more energy in a cell, transcription or translation?', then the answer is almost always translation: translation (i.e. protein synthesis) consumes about 50% of the energy budget of a proliferating cell. Ribosomes are not very efficient, and cells contain LOTS of them (~80-85% of cellular RNA is just ribosomes). They are always in use.
I wouldn't worry about asking questions like this, by the way: it's an excellent question and exposes quite how many complexities there are to even such an ostensibly simple consideration.
  • asked a question related to Gene Expression
Question
4 answers
I have scRNA sequencing data and I want to perform differential gene expression in macrophages. However, I want to include bioinformatics that could complement my studies. Is there any tool that I can include?
  • asked a question related to Gene Expression
Question
4 answers
I have read some articles that used the double mutation to assay gene expression and function in bacteria. I do not know why we need to do this technique. Could someone help me to explain this problem please?
Relevant answer
I am genuinely thankful for your assistance.
  • asked a question related to Gene Expression
Question
1 answer
help
Relevant answer
Answer
hi
Green fluorescent protein (GFP) and amino 3 glycosyl phosphotransferase (neo) are two different genes that are often used for different purposes in molecular biology research.
GFP is a small protein that naturally fluoresces green when exposed to certain wavelengths of light. It has been widely used as a reporter gene to monitor gene expression in various cell types and organisms. GFP has several advantages as a reporter gene, including its high sensitivity, rapid expression, and stability. It is commonly used to monitor gene expression in real-time and to visualize the localization and dynamics of gene expression in cells and tissues.
Neo, also known as the neomycin resistance gene, encodes an enzyme that confers resistance to the antibiotic neomycin. It is often used as a selectable marker in genetic engineering to allow the selection and enrichment of cells that have been successfully transfected with a gene of interest. Neo is commonly used in conjunction with other selection markers, such as hygromycin or kanamycin, to allow for multiple rounds of selection and enrichment.
In general, GFP is a better choice for monitoring gene expression levels because it is sensitive, stable, and can be easily visualized using fluorescence microscopy. Neo is not typically used for monitoring gene expression levels because it does not directly affect gene expression, but rather confers antibiotic resistance to cells that have stably incorporated the gene into their genome.
It is worth noting that both GFP and neo have limitations and may not be suitable for all applications. For example, GFP may not be expressed at detectable levels in some cell types or under certain conditions, and its fluorescence may be affected by factors such as pH or the presence of other fluorophores. Neo may also have negative effects on gene expression and cell growth, and may not be effective in all cell types or organisms. Therefore, it is important to carefully consider the specific goals and requirements of your study before selecting a reporter gene or selectable marker.
  • asked a question related to Gene Expression
Question
2 answers
I'm trying to induce inflammation in THP-1 by priming with LPS (1ug/ml) for 4 hours and followe by nigericin (7.5uM) stimulation for 1 hour. I measure the gene expression of IL18 and IL1beta, IL1beta increased after LPS, but IL18 decreased. I've tried 3 times but the trend was the same. Has anyone met this problem before?
Relevant answer
Answer
Malcolm Nobre Thank you!
  • asked a question related to Gene Expression
Question
3 answers
Suppose I have a Chromogenic Western Blot result showing a series of bands. If I wanted to know the exact number of proteins in a particular band, would it be possible to execute a gene expression analysis? If yes, how can I do it?
Relevant answer
Answer
If you have a known steady-expression control you can do a relative protein abundance comparison. But I don't think that's what you are asking.
  • asked a question related to Gene Expression
Question
14 answers
Hi to all.
My question is how can I optimize my RTqPCR if the cDNA dilutions ended up in similar Cq?
I synthesized my cDNA from 350 ng total RNA, assuming 1:1 production I should have 350 ng cDNA in 20 ul right? Then I did a dilution of 1/2, 1/5, 1/10 and 1/20 (I know the first three are consider quite a lot to be used in the run) and used them in a 20 ul run. The gene is a ref. gene: GAPDH. Interestingly the Cq values aren't that different between the dilutions (~29, ~30, ~31 and ~30). Obviously these aren't good values but I don't know what can I do to optimize the run.
Relevant answer
Answer
Alright, I will try to share what tips/tricks I can.
Honestly, while RNA is vastly more labile than DNA, it isn't really some sort of mystic-grade vulnerability, and you don't need utterly RNAse free environments to isolate perfectly viable RNA. They will help, obviously, but just starting with RNAse-free stuff, using careful pipetting and not making obvious mistakes will usually be sufficient.
So: use filter tips. Here the filter is primarily protecting your sample from whatever gunk might be hiding up in your pipette barrel. Use filter tips for everything (1000ul, 200ul, 10ul).
Use RNAse-free microcentrifuge tubes (most prepacked tubes should be certified RNAse free): keep a dedicate bag for RNA work, keep the top sealed/folded over when not in use, and only fish out tubes with gloved hands. If you put an ungloved hand into the bag, then assume the bag is now no longer good for RNA work (or use at your own risk).
Use RNAse free water for everything: either buy it, or make your own using DEPC or DMPC: add DEPC to 0.1%, shake vigorously and leave at 37degrees overnight with the lid of the bottle slightly loose. Autoclave, then close the lid tight.
Take small aliquots for working (I tip out 50ml at a time into a falcon tube) so you're not constantly dipping in and out of your stock. If an aliquot gets contaminated, or you suspect it's contaminated, throw it away, make another.
Use a bench area you trust: this doesn't mean you need a dedicated area, but use common sense (if a genomic DNA extraction protocol involves 'add 100ul of RNAse H', for example, go do that protocol somewhere else).
Use common sense in general: just be aware that the primary source of RNAse activity is the investigator: we are covered in bacteria all the time, and all of those are robust RNAse sources.
Wear gloves. Wear them basically all the time. If you think the gloves are dirty, change the gloves.
Next up: practical tips/tricks and when to be most careful.
If you can, freeze tissue. Freeze everything until you need it not to be frozen. RNA inside a sample frozen at -80 will endure far better than RNA inside fresh tissue, and while its frozen, it cannot be broken down by RNAses (they're frozen too).
Try to keep tissue frozen RIGHT up until you lyse/denature everything.
Frozen tissue is safe.
Lysis: I use trizol (or trizol equivalent) methods for almost everything. Almost nothing survives the addition of large amounts of chaotropic salts dissolved in phenol: a frozen sample covered in RNAses can still be used for RNA extraction if you dump it straight into trizol, because the RNAses will unfold and denature right along with everything else.
I typically freeze tissue in liquid nitrogen, store it at -80, crush to to powder under liquid nitrogen (i.e. never let it defrost) and then add trizol directly to the frozen powder. The first time the tissue melts, it's melting in phenol.
RNA inside trizol suspension will endure, and can indeed be frozen at -80 for longer-term storage.
RNA in trizol is safe.
Once you add chloroform to initiate phase separation: THAT'S when you need to start being extra careful. The aqueous phase is RNA in solution, and it's essentially unprotected. Collect aqueous phases one at a time, tilting the tube to minimise stuff falling into it. Cap tubes as soon as you're done transferring.
I typically use isopropanol precipitation rather than columns, because I like to see the size of my pellets, but all downstream stuff from phase separation is extra-careful-time. Precipitated RNA itself is actually fairly safe, since RNAses can't really degrade a solid chunk of dry RNA (accordingly, you can also freeze pelleted RNA at -80 for some weeks).
If you're going down column-based preps, then all the on-column stuff is largely out of your hands. Keep the columns wrapped up and clean (most come individually wrapped, but if they're in a bag, treat that bag as for tubes, above: gloves for all the things, seal up when not in use).
Isolated RNA should be either frozen immediately, or kept on ice for spectrophotometry/bioanalyser, and THEN frozen.
Try to make it into cDNA as soon as possible, and try to minimise freeze thaw: better to make a lot of cDNA in one batch than to keep dipping into it for multiple one-step reactions.
  • asked a question related to Gene Expression
Question
4 answers
I've read that I need cDNA standard curves to test primer efficencies.
I'd like to know if I could use genomic DNA instead, since I've got too many different conditions from which different cDNAs have been synthesized.
In the end, depending on the efficencies, I've read that I may use either the Double Delta or the Pfaffl method to quantify the relative expression of genes.
Relevant answer
Answer
Typically not. Most folks studying gene expression are using primers that amplify a target region that is spanning an intron. The difference in product size from gDNA vs. cDNA would mean you can't do a fair comparison of amplification efficiency.
The only way this would be possible is if:
1. Your primers amplify the exact same product in gDNA vs cDNA (not ideal since it's much harder to rule out contamination/incomplete DNAase treatment).
2. Your DNA extraction is extremely high-quality
3. All of your genes of interest are from the same part of the genome (e.g. all nDNA, no mtDNA or cpDNA)
4. You are doing a relative expression-level study (not exact copy number).
Most folks clone their target region of interest as a cDNA into a plasmid.
Good luck!
  • asked a question related to Gene Expression
Question
4 answers
Hello scientific community. Please help me to find out the standard error values for a relative gene expression got via QPCR.
I have performed QPCR experiment and got relative gene expression values by delta delta method. Now I am stuck to find out standard error values.
If someone can help me by providing the formulated Excel sheet or can suggest me easy to use software if you suggest me the software to use then please also guide me how to use this software to get standard error values for relative gene expression.
I shall be high thankful.
Relevant answer
Answer
Reyhaneh Ravanbakhsh thanks alot. I Will contact you.
  • asked a question related to Gene Expression
Question
3 answers
I have values of two treatment groups with similar gene target and all 3 groups have the same beta actin value. Treated 1 = 21.00 cq and Treated 2 = 20.50 cq, while both treatments having the same Control = 19.00 cq. From this rough data, is it reliable to say Treated 2 have higher expression than Treated 1 against Control due to lower cq value by 0.5? Thank you in advance
Relevant answer
Answer
Try to add technical replicates and at least 3 biological replicates to get a statistically significant data. However practically with the difference of 0.5 cT you cannot actually comment too much about the expression of gene. A higher number of biological replicates would help in this case.
  • asked a question related to Gene Expression
Question
6 answers
I am using Optineurin gene and I want to establish stable cell line for specific gene expression. But when I have transfection using antibiotic resistance media, it almost died nearly 90%. SO after passage several time and change the media for remove the death cell. Most importantly, I used positive sample by Doxycycline. But it also showed not good efficacy in transfected cell. The gene was derived by using Flp-In system. When I used to transfection, the cell confluency was around 80% and I used following the method of Invitrogen Lipofectamine™ 3000 Transfection Reagent . Have any suggestion to get the good efficacy number of cell ?
Relevant answer
Answer
It depends on your next plan. If you would like to reuse the transfected cells for downstream procedures, then you can sort positive cells by FACS. If you are trying only to see the transfections efficiency you can use flowcytometry. However, in the mentioned procedures you have to tag your cells with GFP ... etc. Another possible way is to use RT-qPCR, so you can confirm a rise in the expression of your gene of interest.
Hope that was helpful.
Best ....
  • asked a question related to Gene Expression
Question
4 answers
Is it proposed to use replicates (and if yes, how many) when doing spatial omics, using the same type of tissues but from different animals within the same phylum?
Relevant answer
Answer
Sure, costs are relevant, but much more relevant is the clear definition of a statistical model that maps meaningful biological features. This is simply not really clear how to do that in a multidemsional space.
If you found some kind of pattern (however you define it), it is certainly good practice to repeat the entire experiment and analysis at least onece (better twice or trice) to see if the pattern you identified occurs (more or less) robustly. If the observed pattern indicates some biological interpretation, you can go back to biology ind infer the hypothesized effects by new experiments (knock-ins, knock-outs, blocking, competeing, histology, etc.).
  • asked a question related to Gene Expression
Question
1 answer
I've few queries regarding bacterial and mammalian plasmids for expression of Gene of Interest. What plasmid elements/components that are differ between bacterial and mammalian Plasmids to express a gene of Interest.
According to me :
The elements/components that are common between bacterial and mammalian Plasmids are :
  1. Bacterial ori of replication.
  2. Bacterial selection marker.
  3. Promotor + gene of Interest for Expression of Gene.
The elements/components that are differ between bacterial and mammalian Plasmids are:
  1. Mammalian Ori such as  EBV or SV40 if the Transfected cells expressing the Epstein–Barr virus (EBV) nuclear antigen 1 (EBNA1) or the SV40 large-T antigen for Episomal replication of Transfected plasmid.
  2. Mammalian selection marker (For positive selection of cells that take up plasmid).
  3. Promotor + gene of Interest for Expression of Gene + PolyA (example SV40 pA or CMV pA)
  4. Reporter Gene.
I'd like to know is there any other differences?
Thank You.
Relevant answer
Answer
Plasmids for mammalian expression use different organism specific promoters, a eukaryotic ribosomal binding site, an intron in the CDS of the gene of interest to avoid bacterial expression and to increase expression in the mammalian cells and a poly A tail after the stop codon to reduce mRNA degradation.
  • asked a question related to Gene Expression
Question
2 answers
I read most of publications that done on this point but I didn't found the housekeeping gene. I will be thankful for any advice. Thank you in advance for your time to answer my question.
Relevant answer
Answer
Hi Mohamed M Rashad , I recommend this research published this year in Plos One: An approach to quantitate maternal transcripts localized in sea urchin egg cortex using RT-qPCR with accurate normalization (https://doi.org/10.1371/journal.pone.0260831). The authors made a rigorous evaluation of the reference gene candidates with different approaches such as BestKeeper, CV, ΔCt, geNorm, and NormFinder. You can test the same genes or design new ones based on the reference literature cited in this article or with the sequences deposited in NCBI. Otherwise, you can try another method to normalize the gene expression, like cDNA quantification with Oligreen.
Best regards,
Clei.
  • asked a question related to Gene Expression
Question
1 answer
Hello everyone,
I am analysing the expression of several genes coming from BV2 cells that I cultured some months ago. I am having trouble finding expression of some genes, and now I am doubting that the orginal cells were healthy and that may have altered gene expression. From the PCRs I have ran so far I have obtained good expression of 18s gene and RPL13 (housekeeping genes) but found no expression of RPL37. I attach here picures at 20x and 40x of control wells from my experiment. Please, if you have any comment/information/suggestion on how to check if the cells were healthy or how to find the transcriptome of BV2 cells, it woulf be of much help.
Relevant answer
Answer
Check this PMID: 32922284
  • asked a question related to Gene Expression
Question
5 answers
I extracted RNA from pellets of mammalian cell culture and turned it into cDNA. To determine the level of gene expression, I'll use qPCR. Should I first run the cDNAs through agarose gel electrophoresis? What outcomes should I expect when I use it?
Have a nice day.
Relevant answer
Answer
Hi there,
If you want to check integrity of the RNA, you could run a denaturing agarose gel of the total RNA.
Once cDNA synthesis is done, there is no need for an agarose gel electrophoresis.
You will anyways use housekeeper genes in qPCR. That way you can see if DNA synthesis failed.
  • asked a question related to Gene Expression
Question
8 answers
Hi everyone,
I performed a qPCR to assess the collagen type 1 gene expression in the mesenchymal cell line.
prior to the test, I got a little amount of total RNA than what I regularly get (10 ng/ul) as the cell growth was low.
so during performing the qPCR I found out that my housekeeping gene has a Ct of about 30 but the target gene (collagen 1) has no Ct in 40 cycles. so I decided to extend the cycle number to 60 cycles. I observed the Ct for collagen type 1 on cycle 50.
if there were no primer dimer and nonspecific products in the result, is this result reliable for gene expression assessment?
thank you for your kind guidance in advance.
Relevant answer
Answer
As a rule of thumb, 35-36 cycles is enough to amplify just about anything (i.e. a single starting template molecule), and for qPCR you can usually assume a Cq of ~35 represents the absolute lower limit of detection.
Accordingly, Cq values of ~34 = 2 molecules, ~33 = 4, and so on.
A reference gene giving a Cq of 30 means there are countable numbers of reference transcripts in your wells (~30-40 or so), and this should never be the case for a reference (which should generally be fairly abundant). You have very little starting material, and the reference is telling you this.
Remember, you...cannot have less than one molecule in a well. PCR doesn't work on "half a template" or "0.001% of a template": there's a molecule there, or there isn't.
Getting a Cq at cycle 50 means it isn't there.
Alternatively, you could have a ruinously inefficient PCR, in which case you'll see what _should_ appear at cycle 25 actually appear at cycle 50 (or whatever), and if this is the case, your data is meaningless. This isn't so bad, though: you can always redesign primers to get a more efficient PCR reaction.
  • asked a question related to Gene Expression
Question
2 answers
Hello everyone,
I am looking for gene expression data for affected/metastatic and unaffected/non-metastatic lymph nodes in breast cancer patients to find whether our two genes under question are differentially expressed between them.
Does anybody have any idea where I can find such a database? I have already searched GEO and TCGA.
Best regards,
Mehdi Montazer
Relevant answer
Answer
Ellen A G Chernoff Dear Professor Chernoff, thank you so much for the advice. I did not know about these tools. I will check them.
  • asked a question related to Gene Expression
Question
4 answers
When searching for a proper R package to perform Stouffer's p-value combination on Sum Stats of gene expression values, we have read about quite some possible ways, but unfortunately so far the packages were not applicable for Sum Stats consisting of log2 Fold change and p-values per gene per study only. Does someone of you know a package that can fullfill our needs?
Since the genes included in the individual studies are regulated into different directions (up-/down-regulated), we are aiming for Stouffer's to not lose the direction of the gene expression.
Relevant answer
Answer
Well you might start with this p-value selection doesn't work at all so why would would you combine because meta-analysis is really a selection process. Start with some of these. Best wishes David Booth
  • asked a question related to Gene Expression
Question
6 answers
Hello, I'm going to make a mean log expression value plot for my microarray data,I found from an article that it should be done by VANESA ,but actually I couldn't install this software.
do you know any other software or platform which I can use?
Relevant answer
Answer
R Intros, general:
Adding text to scatterplots:
Specific:
You can read the data into R using read.delim(). It created a data.frame. Each column is a variable, each row is a set of observations. If the columns in the csv file has a header, the header names are the column names = variable names in the data frame (e.g. Gene, Control, Treated).
Simple plotting can best be done using plot(). However, adding labels automatically (so thatthey don't overlap) is difficult. The package ggplot offers a function geom_test_repel() that can do produce non-overlapping annotations. So You may use ggplot to create the plot.
PS: there is the package worldcloud that offers the function textlayout() to achieve a similar rusult without the need for ggplot, but I think here ggplot is the more convenient alternative.
  • asked a question related to Gene Expression
Question
5 answers
Hi all,
I am looking for assistance troubleshooting my RT-qPCRs (using SYBR Green) for delta delta cT analysis. I have one endogenous control (ELF1a) and a single target gene. I ran standard curves for each gene (control and target) using serial dilutions of cDNA from a control sample. The input cDNA concentrations from the serial dilutions for each standard curve were 100ng, 10ng, 1ng, 0.1ng, and 0.01ng. For my experimental samples, 1ng of cDNA was used. Samples were run in duplicates.
My standard curve analysis revealed a slope of -2.017 (R2 = 0.983) for the endogenous control and -1.845 (R2=0.985) for the target gene, which equates to >200% reaction efficiency. I am inexperienced doing qPCRs, but my interpretation of the melt peaks is that there aren't primer dimers nor non-specific banding in the reaction. Furthermore, the 260/280 readings for all of my RNA samples were 2.1.
I am at a loss as to how to approach troubleshooting this reaction. What do you recommend?
Relevant answer
Answer
Why don’t you design new primers for those that gave you a a double peak in melt curve analysis? How many HK genes do you need to have ideally? I guess 2 must be sufficient, but I would test at least 5 and pick 2 that work best. Good luck!