Science topic

DNA - Science topic

A deoxyribonucleotide polymer that is the primary genetic material of all cells. Eukaryotic and prokaryotic organisms normally contain DNA in a double-stranded state, yet several important biological processes transiently involve single-stranded regions. DNA, which consists of a polysugar-phosphate backbone possessing projections of purines (adenine and guanine) and pyrimidines (thymine and cytosine), forms a double helix that is held together by hydrogen bonds between these purines and pyrimidines (adenine to thymine and guanine to cytosine).
Questions related to DNA
  • asked a question related to DNA
Question
6 answers
Who (first) proposed/used/coined the term ‘translation’ in biology/genetics? What is the history behind the use of the word? Thank you!
Relevant answer
Answer
The oldest relevant reference in the OED is to a paper by Gamow and Yčas from 1955, namely Statistical Correlation of Protein and Ribonucleic Acid Composition.
  • asked a question related to DNA
Question
3 answers
Preprint Nuance
Relevant answer
Answer
I don't see a question, but there is a relevant observation to be made: Dealing only with genetics any black or white males are closer than any male and female. By the way, the color pigeonhole society employs are simplistic to the point of stupidity. 'Whites' are pink, not white unless they happen to be albinos. 'Blacks' are almost always some shade of brown,and Asians have a remarkable range of color tones.
  • asked a question related to DNA
Question
3 answers
I am running simulations with oxdna using advanced techniques of sampling as Umbrella Sampling.
Relevant answer
Answer
Yes and no. There are minor differences which can cause your results to be false. Reducing the grid box would definitely decrease the total volume so that the interactions will increase just like in a more concentrated situation. However, when your ligand and/or receptor (target molecule) goes outside the walls of grid box, it comes back from the other side (Leaving the grid from right, entering from left etc...). So in that sense, you create a new collision probability for the atoms to interact. Let's assume you have a tiny drug that you want to bind to your large protein to inhibit/suppress its activity. In terms of theoretical chemistry, it may not have any chance to attack and residue on the right side of that protein. As you limit the grid-box, some right parts of that protein will enter from the left side and your drug will be able to interact that side as well which is not supposed to happen. So if you want to increase the concentration go ahead and do it. Please do not decrease the grid-box volume to satisfy that need since it will yield wrong outcomes.
  • asked a question related to DNA
Question
5 answers
I did PCR of 6 amplicons and when I did agarose gel electrophoresis, they all appear consistently bigger than they should be (e.g. the amplicon should be 4000 bp, but is around band of 5000 bp marker).
Sizes of my amplicons, from left to right are:
top row - 2610, 4052, 4000
bottom row - 4860, 4565, 3235
The pictures are the same, just different exposition. There are 5 identical samples differing at annealing temperature, split by marker.
The marker looks quite bad. I don't know why, I used it previously. The conditions of electrophoresis are: 1% agarose with TAE; 50 V; 90 mins
Relevant answer
Answer
That sort of smear-y shape can be due to a few things:
loaded too much sample
dirty gel combs
removing combs too early
So, I bet if you washed & rinsed the gel casting equipment & gel rig; used fresh electrophoresis buffer; and made sure you let the agarose completely harden before removing the combs your bands will look much better.
I don't think the sizes are necessarily different from what you have predicted, pretty sure it's just the smear/wobbly bands that's making it hard to estimate the size.
Good luck!
  • asked a question related to DNA
Question
11 answers
I am a student currently working on a project that requires the sequencing of DNA from two species of California sunflowers. For the record I am using the ddRADseq method. Although I am familiar with size selection methods, I am not familiar with how to choose the size. The general range my mentor gave me is around 300-600 base pairs. What are the advantages and disadvantages for smaller sizes and bigger sizes. Thank you!
Relevant answer
Answer
The choice of DNA size for sequencing depends on the specific goals of the sequencing experiment and the sequencing technology being used. Here are some factors to consider when selecting the DNA size:
1. Type of Sequencing Technology: Different sequencing platforms have specific requirements and capabilities regarding DNA fragment size. For example, short-read sequencing technologies like Illumina typically require smaller DNA fragments (e.g., 150-500 base pairs), while long-read sequencing technologies such as Pacific Biosciences (PacBio) or Oxford Nanopore Technologies (ONT) can handle larger DNA fragments (e.g., several kilobases to tens of kilobases).
2. Genomic or Targeted Sequencing: If you're performing whole-genome sequencing, the DNA size should be representative of the entire genome, so fragmentation methods may be used to generate smaller, randomly sized DNA fragments. For targeted sequencing, such as amplicon sequencing or targeted capture, the DNA size is determined by the specific regions of interest and the primer or capture probe design.
3. Desired Coverage and Resolution: The size of DNA fragments can affect sequencing coverage and resolution. Smaller fragments may provide higher coverage across the genome but may result in lower resolution for structural variations and long-range genomic interactions. Larger fragments can capture more long-range genomic information but may require specialized sequencing technologies.
4. Sample Quality and Integrity: High-quality DNA with minimal degradation is essential for successful sequencing. Fragment sizes should be selected to ensure that the DNA is intact and free from degradation, especially for long-read sequencing technologies that require longer DNA fragments.
5. Downstream Analysis Applications: Consider the downstream applications and analyses that will be performed with the sequencing data. For example, if you're interested in identifying structural variations or long-range genomic interactions, longer DNA fragments may be preferred. Conversely, if you're focusing on variant calling or SNP analysis, shorter DNA fragments may be sufficient.
6. Library Preparation Methods: The choice of DNA size may also depend on the library preparation method used for sequencing. Different library preparation kits and protocols have specific requirements for input DNA size and fragmentation methods.
  • asked a question related to DNA
Question
10 answers
Hi molecular biologists, I'm wondering if any of you might be able to help me with a question I have.
I am attempting to insert the DNA sequence coding for a protein domain into a plasmid (the plasmid is popinF). The insert DNA (E. coli optimised) was synthesised by Thermo (and it has passed their QA/QC), and I've successfully inserted it into popinF and transformed E. coli stellar cells, before collecting 3 different colonies from a plate to perform minipreps and acquire the plasmid with inserts. The sequencing results came back for all of them, and confirmed that the full (and correct!) DNA sequence had been inserted into one of the 3 plasmids.
However, I found it very peculiar that one of my plasmids appeared to have my DNA insert, but in a degenerated form with regards to the sequence. In the alignment shown attached, I can clearly see that there is very very strong matching of the sequenced result to the DNA from ~230 base onwards, showing that the synthetic DNA has inserted. But the sequence prior to this region does not show a high correlation to my DNA insert, and I'm wondering how this could be, and what could have caused this? I know that the synthesised DNA must be correct because I've successfully put the full length sequence into another identical plasmid - could it be that this particular plasmid showing a degenerate sequence could have undergone mutations within the E. coli or have degenerated in other ways, and if so could anybody please expand on the mechanisms and nature of these mutations? If anybody has any insight into mutation events of DNA inserts in plasmids within bacteria or knows of any good literature that reviews it and how to avoid them during recombination/transformation, I would be very appreciative for the help!
Thanks very much all,
Rob
Relevant answer
Answer
Plasmid insert mutations can occur due to various reasons, including errors during DNA replication, exposure to mutagenic agents, or incorrect handling during molecular biology techniques. Here are some common causes of plasmid insert mutations and ways to avoid them:
  1. Replication errors: During DNA replication, polymerase enzymes may introduce errors leading to mutations. This can happen due to misincorporation of nucleotides or slippage during replication. To minimize replication errors, use high-fidelity polymerases for PCR amplification and ensure proper primer design to reduce the likelihood of misincorporation.
  2. Exposure to mutagenic agents: Plasmid DNA can be exposed to mutagenic agents such as UV radiation, certain chemicals, or reactive oxygen species. These agents can induce DNA damage and mutations. To avoid exposure to mutagenic agents, handle plasmid DNA with care, use protective measures such as UV shields, and store DNA samples properly to prevent degradation.
  3. Errors during cloning: Mistakes made during cloning procedures, such as incorrect primer design, improper ligation, or inefficient transformation, can lead to mutations in the plasmid insert. To avoid these errors, carefully design primers, optimize cloning conditions, and use appropriate positive and negative controls during cloning experiments.
  4. Insert instability: Some plasmid inserts may contain repetitive sequences or regions prone to instability, leading to mutations such as insertions, deletions, or rearrangements. To mitigate insert instability, sequence the insert region to identify any repetitive sequences or unstable regions and avoid using them if possible. Additionally, consider using alternative cloning methods or vectors that are more suitable for stable insert maintenance.
  5. Contamination: Contamination with nucleases or other enzymes can lead to degradation of the plasmid insert, resulting in mutations. To prevent contamination, maintain sterile conditions during molecular biology procedures, use certified DNAse-free reagents, and regularly check equipment for cleanliness.
  6. Storage conditions: Improper storage conditions, such as exposure to extreme temperatures or repeated freeze-thaw cycles, can damage plasmid DNA and introduce mutations. To ensure stability, store plasmid DNA at appropriate temperatures (-20°C or -80°C), avoid frequent freeze-thaw cycles, and aliquot DNA samples to minimize exposure to light and air.
  • asked a question related to DNA
Question
8 answers
Any method for DNA extraction from cotton fibre and fabrics.
Relevant answer
Answer
we are working on colored cotton fabric dna extraction kit
  • asked a question related to DNA
Question
4 answers
I'd like to detect the presence of different species in a DNA extract using qPCR. Are there specific targets already listed for each species (animals, yeasts)?
Thanks,
Relevant answer
Answer
Hi, Angrand, I use qPCR for detecting specific genera using genera specific primers. But detecting species would be a bit difficult. you can go with metagenome targeting whole 16S for species identification.
Thanks
  • asked a question related to DNA
Question
9 answers
I am getting zero DNA yield after using qiagen purification columns. I finally traced the problem to NEBuffer 3.1, but pH doesn't seem to the cause.
Essentially, I observe:
3 ug of DNA in 50 uL of water ->
qiagen purification column ->
1.5-2.3 ug of DNA
In comparison:
3 ug of DNA, 5 uL of 10X NEBuffer 3.1, bring to 50 uL with water ->
qiagen purification column ->
zero DNA
I thought it was a pH problem -- high pH can cause low efficiency. But I don't think pH is the problem. Because pH strips and qiagen's pH indicator say my pH is okay (pH<7). And I added 20 uL of 3 M sodium acetate (pH 5) and it doesn't fix the low yield at all. I observe:
3 ug of DNA, 5 uL of 10X NEBuffer 3.1, bring to 50 uL with water ->
Add 20 uL of 3 M sodium acetate (pH5) ->
qiagen purification column ->
zero DNA
Why does adding NEBuffer 3.1 cause low yield if not pH problems?
Relevant answer
Answer
I added 3 M NaAc pH5 and the purification still didn't work. Still 0% recovery.
I used pH strips too. The pH is low but still 0% recovery.
  • asked a question related to DNA
Question
6 answers
We would like to purchase around 10 thousand DNA oligos in a 96 well format (25 nmol). The cost per base is coming to around Rs 14-15. We wonder if there is any economical option available in the market.
Thank you
Relevant answer
Answer
Dear Colleague,
I trust you are doing well. In response to your request for suggestions on reasonably priced oligonucleotide synthesis services, both within India and internationally, I am pleased to provide a comprehensive overview aimed at facilitating your decision-making process.
Oligonucleotide Synthesis Services in India:
  1. Eurofins Genomics India Pvt Ltd: Eurofins is renowned for its high-quality sequencing and synthesis services. They offer competitive pricing for custom oligonucleotides, catering to various research needs, including standard, modified, and high-throughput oligo synthesis.
  2. Xcelris Labs Ltd: Xcelris is another prominent player in the field, offering a range of genomic services including oligonucleotide synthesis. Their services are known for being cost-effective and reliable, making them a popular choice among researchers in India.
International Oligonucleotide Synthesis Services:
  1. Integrated DNA Technologies (IDT): IDT is a global leader in the area of custom oligo synthesis, renowned for its high-quality products and services. They offer competitive pricing and have facilities in the United States, Europe, and Asia, ensuring timely delivery worldwide.
  2. Sigma-Aldrich (now Merck): Sigma-Aldrich provides a wide range of oligonucleotides through its custom DNA synthesis service. They are known for their reliable quality and extensive options for modifications, catering to diverse research requirements.
  3. GenScript: Offering both standard and customized oligonucleotide synthesis services, GenScript has a strong presence worldwide. Their services are competitively priced and are backed by excellent customer support and fast turnaround times.
Selection Criteria:
When selecting an oligonucleotide synthesis service, consider the following criteria to ensure you receive the best value and quality for your research needs:
  • Quality and Accuracy: High-quality oligos are crucial for the success of your experiments. Look for services with positive reviews regarding the accuracy and purity of their products.
  • Pricing: Compare prices among different providers, but also consider the cost-effectiveness in terms of quality and additional services provided.
  • Turnaround Time: Ensure the provider can meet your timeline requirements, especially if you are working on time-sensitive projects.
  • Customer Support: Efficient and responsive customer service can significantly enhance your experience, especially when customizations or modifications are involved.
  • Shipping and Handling: For international orders, consider the logistics of shipping and handling, including costs and the potential for delays or customs issues.
Recommendation:
Before finalizing your decision, it may be beneficial to request quotes from multiple providers and evaluate any bulk order discounts or promotional offers that could further optimize your investment. Additionally, reaching out to your professional network for firsthand reviews and experiences can provide valuable insights into the reliability and quality of the services you are considering.
Should you have any further inquiries or require assistance in contacting these services, please feel free to reach out.
Best regards,
With this protocol list, we might find more ways to solve this problem.
  • asked a question related to DNA
Question
7 answers
Hi, I want to insert 130 bp DNA into 3000 bp vector, however I could not obtain colonies. I used 50 ng insert DNA and also tried to increase insert amount but still I could not observe any colonies. Is there anyone who tried to insert small DNA (nearly 100 bp) and experienced this problem? If so, can you explain your solution?
Relevant answer
Answer
Yes, infusion cloning, also known as In-Fusion cloning, is a popular method used by many researchers to insert short DNA fragments into vectors with high efficiency. In-Fusion cloning is a versatile and powerful technique that enables the direct joining of DNA fragments—such as inserts and vectors—without the need for restriction enzymes or ligases. This method relies on the homologous recombination of linearized vectors and insert DNA sequences that share complementary ends.
How It Works:
In-Fusion cloning exploits the natural homologous recombination mechanism to seamlessly join DNA fragments. To prepare for In-Fusion cloning, both the vector and the insert DNA must have complementary ends, which can be created through PCR amplification using primers that contain the necessary overlaps. The method allows for precise insertion of the DNA fragment into the vector at a specific location, making it highly useful for cloning short inserts, mutating specific sites within a plasmid, or adding tags to proteins.
Advantages:
  • Efficiency: It has a high success rate, often yielding a high proportion of positive clones.
  • Simplicity: The procedure is straightforward, with fewer steps compared to traditional cloning methods, which reduces the potential for errors.
  • Versatility: It can be used to clone a wide range of DNA fragment sizes into any vector at any site chosen by the researcher, provided that the overlapping sequences are correctly designed.
  • No Need for Restriction Enzymes: Since In-Fusion cloning does not rely on restriction enzyme sites, it avoids problems related to the availability of unique restriction sites within the vector or insert sequences.
  • Seamless Cloning: The method does not leave any extra bases or scars between the insert and vector, which is particularly important for functional studies where such additions could affect gene expression or protein function.
Applications:
  • Inserting Short DNA Sequences: It's ideal for inserting short sequences such as tags, promoters, or small genes into plasmid vectors.
  • Site-Directed Mutagenesis: For introducing mutations at specific sites within a DNA sequence.
  • Gene Fusion: Creating fusion genes without any extra amino acids at the junction.
  • Multiple Fragment Cloning: It can be used to simultaneously insert multiple fragments into a vector, which is useful for constructing complex genetic constructs.
Considerations:
When using In-Fusion cloning for inserting short DNA into a vector, it's important to:
  • Design primers carefully to include 15-20 base pairs of homology to the vector ends.
  • Ensure the vector is linearized and purified away from any competing DNA species.
  • Perform a control reaction to gauge the background level of vector self-ligation or re-circularization, if any.
In-Fusion cloning has been widely adopted in molecular biology labs due to its ease of use and high efficiency, making it a go-to method for cloning and genetic manipulation projects.
l This protocol list might provide further insights to address this issue.
  • asked a question related to DNA
Question
8 answers
Hi,
I'm new to qPCR, so please forgive me if my question may seem stupid. I am using a PikoReal machine to quantify viral load (DNA) with the help of a probe. So I'm testing viral DNA in bird DNA samples and am always adding a positive, as well as a no template control to each plate.
My problem is that a lot of my samples are weakly positive, so with Cq>35, and the automatic baseline calculated by the PikoReal software is therefore often too low (please see attached PDF for examples). This results in dodgy duplicate values (e.g. 24 in one well, NaN in the other well for the same sample), because the software is actually measuring background noise in some wells because the baseline is set too low.
I am not sure how to adjust the baseline in a way that
a) the results are not "fake" (e.g. by cutting out weak positives by lifting the baseline too much), but as close to the truth as possible
b) the results are comparable across many plates, as I have 100s of samples.
Any help would be much appreciated :)
Relevant answer
Answer
Dear Esteemed Colleague,
Thank you for your inquiry regarding the handling of baseline and threshold differences in quantitative Polymerase Chain Reaction (qPCR) analyses. Addressing these differences is crucial for ensuring the accuracy and reproducibility of qPCR data. Below, I provide a comprehensive and structured approach to manage these variations effectively.
1. Understanding Baseline and Threshold in qPCR:The baseline in qPCR refers to the initial cycles of the amplification process where the signal is too low to be distinguished from background noise. The threshold is set at a level where the signal begins to be significantly detected above the baseline, marking the point where exponential growth of the PCR product is first observed. Correctly setting the baseline and threshold is essential for accurate quantification of the initial amount of template DNA.
2. Baseline Adjustment:To deal with baseline differences, it is imperative to standardize the baseline setting across all runs. This can be achieved by:
  • Selecting a Uniform Baseline Range: Identify a range of cycles (usually within the early cycles) that precedes the exponential phase of amplification across all samples. The software used for qPCR analysis often allows for manual adjustment of the baseline settings. Ensure that this range is consistent across all your qPCR runs to minimize variation.
  • Automated Baseline Setting: Utilize the qPCR instrument's software for automatic baseline adjustment if available. This feature analyzes the fluorescence data to determine an optimal baseline setting for each run, reducing user-induced variability.
3. Threshold Setting:Threshold differences can be addressed by:
  • Fixed Threshold Setting: Set a fixed threshold value that is applicable to all your qPCR experiments. This value should be within the exponential phase of amplification but low enough to be in the early stages of this phase. Consistency in threshold setting allows for more reliable comparison between runs.
  • Automatic Threshold Determination: Many qPCR instruments come with software capable of automatically determining an optimal threshold for each run based on the amplification data. While this method can reduce manual errors, it is crucial to review the automatically set threshold for consistency across different runs.
4. Normalization and Controls:Normalize your qPCR data using reference genes that are stable across your samples. Including multiple reference genes for normalization can further mitigate the impact of baseline and threshold variability. Additionally, use positive and negative controls in each run to monitor the performance of the qPCR assay and to ensure consistency across different runs.
5. Replicates and Statistical Analysis:Perform technical replicates for each sample to assess the reproducibility of your results. Statistical analysis of the replicates can help in identifying outliers and in estimating the variability within your qPCR data, allowing for more accurate interpretation.
6. Software and Analysis Tools:Leverage advanced qPCR analysis software and tools that offer more sophisticated methods for baseline and threshold adjustment, including algorithms that can account for variability across different runs and samples.
In conclusion, dealing with baseline and threshold differences in qPCR requires a meticulous and standardized approach. By implementing the strategies outlined above, you can enhance the accuracy and reliability of your qPCR results, thereby facilitating more precise quantification and analysis of nucleic acid samples.
Should you require further assistance or wish to discuss more advanced strategies, please do not hesitate to contact me.
Warm regards.
Reviewing the protocols listed here may offer further guidance in addressing this issue.
  • asked a question related to DNA
Question
1 answer
Can BHQ quench the fluorescence of FAM in the case shown below?
Relevant answer
Answer
A distance of 20 bp in DNA (I assume) has a length of 68 Angstrom. This should be close enough for a significant extent of FRET quenching by BHQ-1.
The behavior of FAM in this system could be surprising. It is likely to be at least partially quenched already due to interactions with the DNA, regardless of the BHQ. If the oligo is denatured, its fluorescence intensity could increase. In other words, for certain uses, the BHQ may be unnecessary.
  • asked a question related to DNA
Question
4 answers
Hello everyone
I need your help with a problem I can't seem to solven :
I'm planning to do some sequencing of freshwater algae. So I referred to the primer pair made by Stoeck et al. 2010 and Balzano et al. 2015, which is supposed to be general, according to several articles I've read, and quite effective:
Forward primer: V4F (5'-CCA GCA SCY GCG GTA ATT CC-3')
Reverse primer: V4RB (5'-ACT TTC GTT CTT GAT YRR-3')
However, after testing several different PCR cycles and checking on an agarose gel, I very rarely obtain a single band of ~400bp (the desired size).
Most of the time, I end up with either no migration band or several other non-specific bands, including one that is 300bp larger than the desired band.
You can check that on the picture.
I have used the cycles recommended by several articles using these primers (Salmaso et al 2020, Latz et al 2022, Balzano et al 2015...), but I don't get any satisfactory results.
I also carried out several tests with different hybridisation temperatures, reduced the proportion of DNA in the PCR mix, added DMSO and reduced the number of cycles, but these did not give satisfactory results.
But unlike most of the articles that use KAPA HiFi HotStart, the basic polymerase in the Swedish studies, I use pHusion HF HotStart Polymerase.
  • Do you think these non-specific amplifications could be linked to the difference in polymerase?
  • Have you ever had this kind of problem with primers?
  • What do you recommend?
Thank you very much for any help you can give me.
Good luck with your research !
Thomas Charpentier
Relevant answer
Answer
Check the temperature The average Tm of both primers should be used as the annealing temperature in PCR. Increasing the annealing temperature can reduce non-specific reactions and The DNA sample may not be pure enough, so it may not amplify in certain strains. You can try washing and re-precipitating the sample DNA to remove contamination. Identifying microalgae, you can use 18S rDNA for eukaryotic microalgae and 16S rDNA for prokaryotic microalgae.
  • asked a question related to DNA
Question
6 answers
Hello everybody.....
the concentration of my plasmid DNA is 126ng. i want to do real-time PCR taqman probe detection......i want to do serial dilution that should start from 10e8 copy number....so can anybody tell me how to dilute it (how much plasmid DNA and ddwater should be added to make it 10e8). i have attached the calculated values in the attachments. thank you
Relevant answer
Answer
Hello,
Titration of mixed solutions containing nitric acid (HNO3) and sulfuric acid (H2SO4) can be challenging due to the complex chemistry involved. However, there are methods available to accurately determine the concentrations of these acids in a mixture. One commonly used technique is the "back titration" method, which involves a series of chemical reactions to determine the acid concentrations step by step.
Here is a detailed and orderly explanation of the back titration method for titrating HNO3-H2SO4 mixtures:
  1. Dilution: To begin, you should dilute the mixture of HNO3 and H2SO4 with a known amount of deionized water. This dilution step is crucial to ensure that the concentrations are within the range that can be accurately measured using standard titration techniques.
  2. Titration of Excess Base: Add a solution of a strong base, typically sodium hydroxide (NaOH), of known concentration to the diluted acid mixture. The base will react with the excess sulfuric acid to form sodium sulfate (Na2SO4) and water (H2O). The reaction can be represented as follows:H2SO4 + 2NaOH → Na2SO4 + 2H2OThe reaction with nitric acid (HNO3) does not occur at this stage, as it is a weaker acid.
  3. Determination of Excess Base: Continue adding the NaOH solution until the excess base is completely neutralized, resulting in a pH jump. You can monitor the pH using a pH meter or pH indicator. At this point, all the H2SO4 has been converted to Na2SO4.
  4. Titration of Residual Nitric Acid: The remaining nitric acid (HNO3) in the solution is now titrated with a standard sodium hydroxide (NaOH) solution. The reaction between HNO3 and NaOH can be represented as:HNO3 + NaOH → NaNO3 + H2OThe endpoint of this titration is determined using a pH indicator that changes color at the pH at which the reaction is complete. Phenolphthalein is commonly used as an indicator in this case.
  5. Calculation: Calculate the concentrations of H2SO4 and HNO3 based on the volumes and concentrations of the NaOH solutions used in both titration steps. It is important to consider the stoichiometry of the reactions to determine the molarities of the acids accurately.
This back titration method allows you to determine the concentrations of both HNO3 and H2SO4 in the mixture. It is important to perform accurate measurements and titrations to obtain reliable results. Additionally, using standardized solutions and appropriate laboratory techniques is essential for the success of this titration method.
I hope this explanation helps you understand the procedure for titrating HNO3-H2SO4 mixtures. If you have any further questions or need clarification on any aspect of this method, please feel free to ask.
With this protocol list, we might find more ways to solve this problem.
  • asked a question related to DNA
Question
13 answers
I am trying to adapt a DNA extraction protocol to allow me to extract both RNA and DNA from the same sample. I plan to put the sample through Qiagen Allprep DNA/RNA mini kit, but I am not sure at what step I lose the RNA.
SDS/CTAB cleanup
a. Add 10 μl SDS (10%) + 1 μl proteinase K (10 mg/ml stock) to each tube and incubate at 56 °C for 20 min. At this point, pre-incubate CTAB/NaCl solution at 65 °C.
b. Add 35 μl NaCl (5 M) + 28.1 μl CTAB/NaCl (2.5%). Pulse vortex. Incubate at 65 °C for 10 min then perform a quick spin.
c. Add 200 μl phenol:chloroform:isoamyl alcohol (25:24:1) pH 8.0. Pulse vortex. Centrifuge 8,000 × g for 5 min at RT.
d. Collect the aqueous fraction. Add 200 μl chloroform. Pulse vortex for 3–5 sec. Centrifuge 8,000 × g for 5 min at RT.
e. Collect the aqueous fraction. This is final Virus Nucleic Acid.
The final viral nucleic acid goes through the Qiagen DNeasy Blood and Tissue kit.
I am not sure if the final nucleic acid contains RNA. If it does not contain RNA, I would like to know at which step I should put my sample through the kit.
I read protocols using CTAB to extract RNA as well as DNA, but I am not so sure about phenol:chloroform:isoamyl alcohol or plain chloroform. I read a similar protocol for extracting RNA that used CTAB and phenol:chloroform:isoamyl alcohol, but they replaced the chloroform with isopropanol and centrifuged, collecting the pellet as final RNA.
If someone could help me sort this out, it would be great!
FYI, this is part of a protocol to enrich for viral particles and extract the nucleic acid from stool with the least amount of human or bacterial contamination.
Relevant answer
Answer
Hello,
In the context of RNA extraction, both phenol:chloroform:isoamyl alcohol and plain chloroform have been traditionally used, each serving specific roles in the process of isolating high-quality RNA.
  1. Phenol:Chloroform:Isoamyl Alcohol: This mixture is commonly used in RNA extraction protocols for its effectiveness in separating nucleic acids from proteins. The phenol denatures proteins and facilitates their partitioning into the organic phase, while chloroform enhances the separation of the aqueous and organic phases. Isoamyl alcohol, typically added in a smaller proportion (e.g., 25:24:1 phenol:chloroform:isoamyl alcohol), helps in reducing foaming and also aids in the phase separation. When this mixture is added to an aqueous solution containing RNA, upon centrifugation, it forms two phases: an aqueous phase (containing RNA) and an organic phase (containing proteins and lipids). The RNA in the aqueous phase can then be further purified.
  2. Plain Chloroform: Plain chloroform can also be used in RNA extraction, primarily to remove phenol (if used in previous steps) or to further purify the RNA. When used after a phenol treatment, chloroform helps to eliminate residual phenol from the aqueous phase, which is crucial because phenol can interfere with downstream applications such as RT-PCR. Chloroform alone is less effective than the phenol:chloroform mixture in separating RNA from proteins and DNA, but it's a valuable step in ensuring the removal of potential contaminants.
  3. Protocol Considerations: It’s important to follow the protocol's specific guidelines for the use of these chemicals, including the ratios and volumes. The choice between using phenol:chloroform:isoamyl alcohol or plain chloroform will depend on the nature of the sample, the presence of contaminants, and the specific requirements of the downstream applications.
  4. Safety and Handling: Both phenol and chloroform are toxic and require careful handling under a fume hood, with appropriate personal protective equipment. Their disposal must also adhere to safety and environmental regulations.
  5. RNA Quality and Yield: The quality and yield of RNA obtained can be affected by factors such as the pH of the phenol used (acidic phenol is often used for DNA extraction, while neutral or slightly alkaline phenol is preferred for RNA), the integrity of the sample, and the thoroughness of the phase separation.
In summary, both phenol:chloroform:isoamyl alcohol and plain chloroform have roles in RNA extraction, with the choice and use depending on the specific requirements of the RNA extraction protocol and the nature of the sample. Proper handling and adherence to protocols are essential for obtaining high-quality RNA suitable for downstream applications.
Check out this protocol list; it might provide additional insights for resolving the issue.
  • asked a question related to DNA
Question
2 answers
Who agrees both my book and my short film might help morphology? How? Why?
My Book:
Relevant answer
Answer
"Help morphology", what does this mean? Belief is for religion, it is not an intellectual position nor method of science.
  • asked a question related to DNA
Question
8 answers
I am trying to carryout diagnostic digest with AatII and DraIII (Adei) but the two did not cut as expected. Could it be because of CpG sensitivity?
The tests are the paired lanes separated by blank from left to right. The first lane on the left is treated with enzyme (Bam HI, AatII, Xbal, Dra III). The second lane contains the uncut plasmid in each case.
These are not PcR products, they are plasmids from miniprep.
Relevant answer
Answer
If a restriction enzyme is CpG methylation sensitive, you need to consider the following:
  1. Methylation Status: Check the methylation status of the DNA you want to digest. If it's highly methylated at the enzyme's recognition site, it may not work effectively.
  2. Digestion Conditions: Optimize digestion conditions, including enzyme concentration, buffer composition, and reaction temperature, as CpG methylation sensitivity can vary between enzymes.
  3. Use Alternative Enzymes: Consider using alternative restriction enzymes that are not CpG methylation sensitive or using a combination of enzymes to achieve the desired DNA cleavage.
  4. Enzyme Inhibition: Some enzymes can be inhibited by CpG methylation. In such cases, you may need to perform a demethylation step before digestion.
  5. Bisulfite Treatment: If your goal is to study DNA methylation, consider bisulfite treatment to convert methylated cytosines to uracils, allowing subsequent analysis without restriction enzyme issues.
  6. Consult the Literature: Consult enzyme-specific literature and databases for information on CpG methylation sensitivity and recommended conditions.
Overall, optimizing conditions and exploring alternative enzymes are key to making CpG methylation-sensitive restriction enzymes work effectively for your experiments.
l Take a look at this protocol list; it could assist in understanding and solving the problem.
  • asked a question related to DNA
Question
5 answers
We are currently working on a project involving the extraction of DNA and RNA from various types of animal samples, such as whole blood, serum, faeces, etc. The objective is to detect various pathogens through Next-Generation Sequencing (NGS). Our approach for each animal is to combine all the extracted DNA and RNA (converted to cDNA) together (e.g., its DNA from faeces + RNA from faeces + RNA from serum + ...), thus reducing the number of samples to be processed during library preparation.
We have an extraction kit that allows us to either extract DNA and RNA together in a single tube, or extract DNA in one tube and RNA in a different tube. Since our intention is to mix them anyway, we are considering the former option. Nevertheless, we are uncertain whether this will impact the RNA-to-cDNA conversion. Will the presence of DNA affect the conversion process? Additionally, are there any potential effects on the integrity of the DNA? While extracting DNA and RNA together would offer significant benefits in terms of saving time, consumables, and reagents, we will not proceed with this option if it might adversely affect the quality of our extracted DNA or RNA.
Thank you very much for your time and assistance.
Relevant answer
Answer
The presence of genomic DNA in the sample can be considered as a contaminant because of that during running Real time PCR, it affects quantity of cDNA (which is expected to come from expressed genes/transcripts). Since some DNA probes can bind to any double stranded DNA molecules, the presence of genomic DNA can affect quantification of cDNA.
  • asked a question related to DNA
Question
8 answers
Is there any program to convert ssDNA sequences to possible three-dimensional conformation for MD simulations?
Edit: It is an aptamer generated against specific target
Relevant answer
Answer
Ajay Yadav i tried to modify RNA to DNA by using discovery studio by selecting deoxyribose. However when i select Show sequence, I can still see Uracil instead of thymine. Nikhil Maroli Ajay Yadav Do you have any suggestions how to convert structure obtained by RNA composer to DNA aptamer.
  • asked a question related to DNA
Question
3 answers
I want to find the UTR sequence of mRNA sequence of bacteria protein. Can anyone suggest a insilico process for that
Relevant answer
Answer
Hi Harshita
lot of possibilities, but the main ones are to go to the NCBI or UCSC database (for instance, just type NCBI XXXX YYYY UTR region, where XXXX is your bacteria and YYYY your gene) in google.
or just give the species and target in research gate...maybe someone could answer ;)
all the best
fred
  • asked a question related to DNA
Question
1 answer
I want to extract genomic host DNA for targeted SNP sequencing of stool samples. Can someone help me with some details of the procedure.
In particular, I am interested how to clean the extracted DNA mix from microbial DNA, plant/food residue DNA etc?
Also, when the extraction does not happen immediately after sample collection, is it critical reading quality and quantity of the DNA to freeze the stool until further use?
Many thanks for your help!
Relevant answer
Answer
Have you found a way to efficiently enrich host DNA? I also encountered this problem.
  • asked a question related to DNA
Question
3 answers
Hello,
I am transfecting linear DNA along with an Adenovirus transduction to HEK cells and need to isolate the DNA from both the linearised plasmid and viral genome at different passages for restriction digest analysis. I am not interested in the nuclear DNA
Total DNA extraction will include all genomic DNA which I fear will interfere with the restriction digest and produce a highly visible smear on the agarose gel. Ideally, I would want to just isolate cytoplasmic DNA.
Will it be okay to use the total DNA? Could I isolate extrachromosomal DNA using a standard miniprep kit, although they are meant for bacteria? Or would it be better to perform cytosolic isolation followed by DNA analysis?
Relevant answer
Answer
Given the low quantities of linearized DNA that will be present you definitely need some form of enrichment protocol. A miniprep kit might work.
  • asked a question related to DNA
Question
7 answers
I've read that template-switching can create duplications, but those duplications are inverted (aka TIDs - tandem inverted duplications).
Can template-switching result in a non-inverted duplication as well? If so please pass reference to me. Thanks!
Relevant answer
Answer
In certain DNA replication errors, particularly during template-switching events like strand slippage or misalignment, the resulting duplication can indeed be either in the same orientation (tandem duplication) or inverted (inverted duplication) relative to the original sequence. This can occur under specific circumstances and mechanisms during DNA replication.
In template-switching, which is often associated with replication errors like slipped-strand mispairing, the DNA polymerase temporarily dissociates from the template strand and switches to a nearby template strand before continuing replication. The orientation of the newly synthesised DNA strand and the template strand to which it switches can determine whether the resulting duplication is in the same or inverted orientation. Here's a basic explanation:
  1. Tandem Duplication: If the polymerase switches to the same orientation of the template strand, it will synthesise a new DNA strand in the same direction as the original sequence, resulting in a tandem duplication.
  2. Inverted Duplication: If the polymerase switches to a template strand in the opposite orientation, it will synthesise a new DNA strand in the reverse direction relative to the original sequence, resulting in an inverted duplication.
The specific circumstances and outcomes of template-switching events can vary, and they are influenced by factors such as the length and sequence of the repetitive elements in the DNA, the timing of the switch, and the precise mechanisms involved in DNA replication. These events can lead to genetic rearrangements and contribute to the diversity of genomic structures, which can have implications in terms of genetic variation and evolution.
  • asked a question related to DNA
Question
1 answer
And is there any insight if DNA methylation would be impacted?
Relevant answer
Answer
The second question is easier, and the answer seems to be no
As for simultaneous extraction of RNA and DNA from blood, I am not aware of any convincing kits/protocols
(eg
and especially there is no comparison/standardization of any sort. If you can, isolate RNA/DNA separately; if not, can look at the Qiagen Allprep kits and experiment with blood or isolate total nucleic acid and treat w RNAse/DNAse.
  • asked a question related to DNA
Question
3 answers
Hello,
I have performed nucleic acid extraction using the CTAB protocol, and in the final step, I eluted the material with 50 ul of RNase-free ddH2O. A portion of this material was subjected to DNAse treatment. Currently, I have 30 ul of the extracted material remaining and would like to inquire about the possibility of using RNAse A from the "NucleoSpin Plant II, Mini kit for DNA from plants" by MACHEREY‑NAGEL to treat DNA that has already been extracted with the CTAB protocol.
The RNAse A from this kit is provided as 30 mg of lyophilized material, which should be resuspended in 2.5 ml of ddH2O, resulting in a concentration of 12 mg/ml.
I would greatly appreciate guidance on whether it is suitable to use this specific RNAse A for treating DNA already extracted using the CTAB protocol. If it is feasible, I kindly request information on the quantity to be added for effective treatment.
Thank you for your expertise and assistance.
Best regards,
Relevant answer
Answer
Yes, you may use RNAse A for treating DNA which has already been extracted. You may add 50μg/ml of RNase A and incubate the mixture for 1 hour at 37 degree C. The treatment of DNA with RNase A should be done in Tris buffer at the end of the extraction protocol. Salting out step can be repeated as before according to the protocol which you have followed to obtain DNA.
The DNA pellet may be dissolved in Tris-EDTA for DNA protection from degradation by metal dependent nucleases during storage.
Best.
  • asked a question related to DNA
Question
5 answers
Hi
I have certain queries regarding plasmid-DNA transport inside the cell and would be glad if anyone could help me out. 1) As we know there are several proteins/components, which bind DNA specifically or non-specifically and are responsible for DNA compaction inside the cytoplasm. Among those, which are the ones that go inside the pore and make sure that DNA remains in compact form? 2) If proteins responsible for DNA compaction don’t go inside NPC pore except transportin/importins/Ran then how DNA is able to maintain its compact shape throughout the NPC translocation process? Regards -Manoj
Relevant answer
Answer
1) Histones are primarily known for their role in packaging and arranging DNA into nucleosomes. However, there is evidence that histones can also be found outside the nucleus, including in the cytoplasm. In the cytoplasm, histones have been shown to bind to lipid droplets to fight intracellular bacteria. Additionally, histones can shuttle between the nucleus and cytoplasm. Karyopherins, which are nuclear import receptors, can interact with histones and function as chaperones, protecting histones from non-specific interactions in the cytoplasm. Furthermore, studies have suggested that karyopherins may participate in the deposition of histones into nucleosomes. Therefore, while histones are primarily associated with nucleosomes, they can also exist as individual entities in the cytoplasm and participate in cellular processes outside of chromosomal gene regulation.
2) Histone binding to plasmids can occur in a non-specific and non-sequential manner. Nucleosome assembly on plasmids appears to be random rather than cooperative, with nucleosome formation equally likely to occur over all regions of the DNA molecule. This random assembly is supported by the observation that reconstituted plasmids lacking linker histones show no tendency towards precipitation or aggregation. Additionally, histones have been shown to bind bacterial plasmids with high affinity and form nucleosomes in vitro. The binding of histones to plasmids can limit the access of transcription factors to their recognition sites, affecting transgene expression. Therefore, histone binding to plasmids is not specific or sequential, and it can impact gene expression by modulating access to the DNA sequence.
  • asked a question related to DNA
Question
6 answers
total Dna extraction from poultry tissue, kidney, liver, heart using spin column.
Relevant answer
Answer
Upper gel:
In samples 3 & 4 there is obviously no or very little DNA.
In my opinion all other DNAs are not properly dissolved and the gel is overloaded; and you might have RNA contamination.
Ensure your DNAs are completely dissolved and as suggested by others determine the concentration and reload equal amounts.
And, as also suggested by others, you should always seek advice from your supervisor or a senior lab member.
  • asked a question related to DNA
Question
4 answers
I have run multiple agarose gels in which I have at least two combs.
I continually see the lower part of the gel visualising much fainter than the top portion.
The last band of my ladder (250 bp) often disappears. It is quite a problem as I often don't see faint bands in this area if I have loaded my PCR products in the lower portion of the gel.
See images attached - look especially at the ladders in the top vs bottom
In the one image you can see the gel itself after a run in the tank, where the loading dye is significantly lighter in the bottom portion than the top - so it's not a problem with the visualising equipment.
I have run the gel for different times (30-60 min) as well as at different voltages (100V vs 120V) and see the same.
The TAE buffer has been changed
I have observed the same with another gel tank.
The intercalating dye, SBYR Safe, has been replaced with a new aliquot.
Other individuals have also experienced the same issue
Relevant answer
Answer
I use ethidium bromure (BET) to visualize PCR products. This dye migrate in opposite direction of DNA. If my gel run for a too long time the dye risk to be over of small sizes. I supose it is your case.
What you can try: increase agarose concentration at 2% or 3%.
Reduce the time for the migation : 15 min should be enough. You can adjust the voltage.
You can increase your SBYR amount.
Another tip : don't use detergent to clean your gel tank
  • asked a question related to DNA
Question
3 answers
Hello,
I don't understand the difference in what you can see after running your DNA on an urea or an alkaline gel. I know how alkaline works: high pH, hydrogen bonds are prevented, as a result your DNA migrates as ssDNA. But as far as I understand, it is the same with urea: you denature secondary structures with urea, but in the end it is also used to purify or analyze ssDNA.
I already run multiple urea PAGEs and I could distinguish between different ssDNA molecules on the gel. Now my supervisor told me to run an alkaline gel. However, I do not understand what additional information it would give me, since both are denaturing gels? How do you decide to run an urea or an alkaline gel?
Relevant answer
Answer
I looked into the paper from 1988 describing the method. "Thus, it is clear that alkaline conditions will permit a more sensitive detection of DNA damage, including single- and double-stranded DNA breaks and alkali-labile regions, such as apurinic and apyrimidinic sites [9] and phosphotriesters [10]"
Well, not identifying only breaks but also...
I know that people in the neighboring DNA repair group were dealing with the repair of abasic sites.
  • asked a question related to DNA
Question
9 answers
Anyone know the problem? why the Ct values for my samples are fluctuating so much and even not reaching threshold line? is this related to my unspecified primer? because after I checked my primer again, It could bind several genes outside of my target gene. Then shouldn't the non-specific amplification show a false positive? or high fluorescence level, can non-specific primers also cause low fluorescence? I'm sure it's not the DNA sample / template that's the problem, because when I used another primer (GAPDH specific), the results of the amplification curve and melt curve were satisfactory.
Relevant answer
Answer
Are you using SYBR green? Anything that would show up in an electrophoresis should also show up on the real time results.
  • asked a question related to DNA
Question
3 answers
I would like to know the reason why DNA conc. in PCR supermix+nuclease free water = 907.3 ng/uL and A260/A280 = 2.98, A260/A230 = 0.83. So the result is very close to DNA conc. of my PCR product = 1045 ng/uL. (I use AccuStart II GelTrack® PCR SuperMix for doing PCR)
and I also would to know what does A260/A280 ratio higher than 2 means?
Relevant answer
Answer
This isn't surprising at all. The template DNA + primers + dNTPs should be the same "amount of nucleic acids" at the start and end of PCR.
  • asked a question related to DNA
Question
2 answers
I am looking for a protocol which I can get the purified DNA from FTA cards easily and inexpensively.
Then I want to use TE buffer instead of using buffer AT, ATL.
The thing I want to know is that if buffer TE can work instead of buffer AT, ATL or not.
Relevant answer
Answer
ATL is a lysis buffer which has SDS and EDTA in it and is not recommended for long term storage.
AL has guanidine hydrochloride and malic acid to denature proteins.
TE is Tris/EDTA so uses tris to maintain the pH and EDTA to chelate metal ions which would damage the DNA.
A crude method for getting some DNA for PCR might simply be to boil the cards in some distilled water for 15 mins.
  • asked a question related to DNA
Question
4 answers
I am working on some grape Vitis arizonica leaf extractions and after extraction they look great on nanodrop with 260/280 between 1.72 and 2.05 for all samples and 260/230 between 1.7 and 2.10 for all samples. All concentrations also looked good. Although on the gels there was a blob around or slightly less than 100bp which is RNA contamination. For this I was instructed to do an RNase A treatment (I had already done an rnase treatment during lysis). After doing another rnase A treatment post elution I re-precipitated the DNA using sodium acetate and ethanol to remove rnase (centrifuged @4c), then washed the pellet twice with 70% ethanol. Then I ran a gel and it looked good, high molecular weight gDNA (above 10000 bp) with no RNA blob at the bottom. However, the confusing part is my nanodrop now looks horrible on any sample that I treat with rnase A and re-precipitate. I get 260/280 of 1.4-1.7 and 260/230 under 1.0 for any of these samples. I am not sure what contaminant could be there?
Relevant answer
Answer
I agree that and increased 230 is usually due to salts and has a bad effect on the ratios. I think that ethanol is better than isopropanol because ethanol tends to preciptate fewer salts so all I can suggest is to check the rnase buffer for SDS and if it contains sds then do not add the buffer just run the rnase as Rohit Kumar suggests. Also check your ethanol volumes very carefully to avoid using too much ethanol. So If your dna volume is very small it may be more accurate to increase the dna volume so that adding the alcohol is very accurate...eg 5ul dna plus 10ul ethanol is prone to pipetting error but 50ul plus 100ul the accuracy of the pipetting should not be an issue. If the amounts of dna are very small then you may want to add a coprecipitant like glycogen or linear acrylamide to increase the precipitation yield of the dna.
It may be worth using your pipettes to weigh a few volumes of water to check that the pipette used for measuring the dna volume is not reading low and the pipette used to add the ethanol is not reading high
  • asked a question related to DNA
Question
1 answer
Do you think that DNA encoded libraries (DELs) holds promising future prospects and job opportunities ?
I will be very thankful to receive your suggestions
Relevant answer
Answer
DEL which entails a collection of small molecules covalently linked to DNA that has unique information about the identity and the structure of each library member.
In future, promising job opportunities will be beconing on those professionals who are clinically savvy especially in the pharmaceutical/drug and biomedical sciences. More human capital will be needed in numerous drug discovery programmes in future.
  • asked a question related to DNA
Question
4 answers
I have been using the DNeasy kit from Qiagen to extract DNA and, in the final step of the extraction, the product guide recommends to elute the DNA in AE Buffer that contains EDTA (0.5 mM). However, the Nextera protocol might be seriously affected by EDTA during tagmentation. Could I use TE buffer (0.1mM EDTA) for storing DNA samples in a stable manner for a long time? Or would I have to extract my samples only in nuclease free water for immediate use?
Relevant answer
Answer
Hi Moises , I agree with Jatesh , yes you can use EB without any EDTA
  • asked a question related to DNA
Question
5 answers
Hello!
I am using DNeasy PowerBiofilm Kit to isolate DNA from skin swabs. Now I am considering if I should use the provided collection tubes to final storage of extracted DNA. DNA can bind to plastic walls of the tube, so should I rather use low-DNA binding tubes from Eppendorf? On the other hand, the kit has been made for the extraction of DNA from various types of source samples, so it should be appropriate for DNA storage, despite nowhere is wrote that the tubes are DNA-low binding?
Any suggestions?
Martin
Relevant answer
Answer
In my opinion, kit tubes should be fine. If adherence is the problem, you can short-spin it after adequate tap mixing.
Hope, it helps.
  • asked a question related to DNA
Question
7 answers
what is difference between living and non living ?life and death?#molecular concept
Relevant answer
Answer
As part of my ongoing research within the Energy-Impulse-Information-Tensor (EIIT) framework, I am delving into the intricate characteristics that distinguish living and non-living systems, as well as the profound concepts of life and death. This framework offers a holistic perspective to comprehend these concepts, viewing life as a complex system that harnesses energy, responds to impulses, processes information, and evolves within the fabric of spacetime.
1. **Energy**: Living organisms actively harness and convert energy to carry out various functions such as growth, reproduction, and maintenance. This energy is typically derived from food (in animals) or sunlight (in plants) and is used to drive biochemical reactions. In the EIIT framework, this energy utilization can be represented as a flow of energy within the tensor, which is absent in non-living entities. Non-living entities, like a rock or a molecule of water, may contain potential energy or kinetic energy due to their physical state, but they do not actively convert or use energy for processes like growth or reproduction.
2. **Impulse**: Living organisms respond to stimuli or impulses from their environment. This could range from a plant bending towards light to a human responding to a complex thought. In the EIIT framework, these responses can be represented as changes in the impulse components of the tensor. Non-living entities, on the other hand, do not respond to impulses in the same way. They may change state due to external forces, but these changes are not active responses and do not involve information processing.
3. **Information**: Living organisms process information. This is evident in the genetic information carried in DNA, which guides the growth and development of an organism. Additionally, organisms process information from their environment to make decisions. In the EIIT framework, this information processing can be represented as changes in the information components of the tensor. Non-living entities do not process information in the same way.
4. **Tensor**: The tensor in the EIIT framework represents the spacetime in which the living organisms exist and evolve. They are born, they grow, they reproduce, and they die. Over generations, populations of organisms evolve, driven by the forces of natural selection and genetic drift. Non-living entities, while existing within spacetime, do not evolve in the same way.
The boundary between living and non-living can be blurry, especially when we consider entities like viruses. Viruses exhibit some but not all characteristics of life. They contain genetic information and can evolve over time, but they do not have their own metabolism and cannot reproduce without a host cell. In the EIIT framework, viruses could be considered as systems that can process information and evolve over time, but do not independently utilize energy or respond to impulses.
As for the difference between life and death, in the EIIT framework, death could be considered as a state where the organism no longer processes energy, responds to impulses, or processes information. The system that was once a living organism no longer evolves over time in the same way. However, it's important to note that while the individual organism ceases to function, the energy and matter it was composed of continue to exist and transition into other forms and systems. This aligns with the principle of conservation of energy and the concept of information conservation, suggesting that while the specific configuration of energy and information that represented the living organism is gone, the energy and information themselves persist.
Regards,
Alessandro Rizzo
  • asked a question related to DNA
Question
1 answer
Dear colleagues,
I am writing to you to request your assistance in evaluating the results of my research on DNA comparison and analysis. I am not an expert in genetic engineering and would like to receive expert feedback on my work.
The following tasks were performed as part of the study:
DNA comparison was performed for influenza viruses of segment A H1N1 H3N2, with the results presented in the bestmatch.json file. An example of element-wise comparison is provided in the pa_pb1.json file. The accuracy of the match is determined by weight, such as "w":0.472249629.
A search was conducted for identical segments in the DNA sequence. The original file is HA.seq. The results are presented in the following format: [{length, number of variations with this length, number of occurrences of these variations in the original larger sequence}].
[ {2, 25, 403380}, {3, 18, 114124}, {4, 16, 31748}, {5, 16, 7710}, {6, 16, 2893}, {7, 14, 685}, {8, 3, 282}, {9, 5, 137}, {10, 3, 3}, {11, 4, 5}, {12, 2, 2}, {13, 5, 135}, {14, 5, 6}, {15, 4, 132}, {16, 5, 6}, {17, 5, 134}, {18, 5, 6}, {19, 4, 132}, {20, 5, 134}, {21, 4, 5}, {22, 5, 132}, {23, 3, 130}, {24, 3, 130}, {25, 2, 2}, {26, 3, 129}, {27, 3, 129}, {28, 3, 129}, {29, 2, 128}, {30, 1, 127}, {50, 16, 32} ]
DNA was divided into "words." The results are presented in the HA_seq.json file.
I would be grateful if someone from the ResearchGate community could provide their professional insight into my results and assist in their analysis.
The source data was obtained from:
The result files can be downloaded via the link.
During the study, I used our data processing technology, KnoDL, which does not require knowledge of data structure, machine learning, or neural network technologies. All operations took an average of 1-2 minutes on a personal laptop.
Sincerely,
Dmitriy Pospelov
Relevant answer
Answer
I am not a biologist, but I should think that hidden markov models comes out as a prerequisite to undertake this kind of tasks.DNA strands for various genes are aligned along a given strand sequence--the DNA of your mouth has a unique code, different from that of your legs. the mathematics which models this kind of sequence is operations research and biostatistics. to build these models one assumes/ presupposes that you are familiar with what stationary markov chains are, and that you are a bit familiar with bayesian statistics. This will give u added advantages u read and make efforts at understanding & building such sequence-based models.
a link such as the one presented hereunder for your perusal gives you a headstart:
  • asked a question related to DNA
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 DNA
Question
2 answers
Hello everyone,
I have issues with extraction of DNA from Pinus halepensis roots for the further Metagenomics research. My sample size is correct (about 250 mg). I followed the original protocol and obtained very low yield (10-20 ug/mL of DNA)...
I modified the protocol as it was suggested in FAQ of the kit's protocol :
- Increase the volume of DNA/RNA Shield (up to 800 uL in my case)and/or DNA/RNA Lysis Buffer to ensure complete lysis and homogenization. Be sure to centrifuge any cellular debris and then process the cleared lysate.
- To increase yields, heat the ZymoBIOMICS™ DNase/RNase Free Water to 60ºC before use. Additionally, users can reload the eluate onto the column matrix, incubate at room temperature for 3 minutes, and centrifuge again to increase yield without further dilution
- For high density samples, ensure lysate is centrifuged properly to pellet insoluble debris following bead beating. Ensure that none of the debris is transferred to the ZymoSpin™ III-F Filter in the next step.
- Beat beating : using MP fast prep24; 4 cycles of 1 min with liquid nitrogen and 1 cycle without. ( adding liquid nitrogen helps to prevent overheating of the sample).
Funny thing is that I obtain a very good RNA yield, but almost nothing for DNA.
I don't know where the problem is... Maybe some of you had the same issue in the past and could share the solution?
I appreciate a lot your help in advance.
Relevant answer
Answer
There is no specific reference available that discusses the issues with extraction of eDNA using ZymoBIOMICS DNA/RNA Mini Kit from roots. However, there are several studies that have evaluated different DNA extraction methods for eDNA analysis from various environmental samples, including water, soil, and human milk. These studies have used different DNA extraction kits and protocols and have reported variations in DNA yield, quality, and taxonomic composition. For example, [10] found that filtration through a 0.2-0.6 μm pore size PC filter, followed by extraction with MoBio PowerSoil DNA Isolation Kit was optimal for quantification of eDNA from fish. [11] reported that eDNA collection by filtration and subsequent extraction/purification using a DNeasy Blood and Tissue DNA extraction kit (Qiagen) or PowerWater DNA Extraction Kit (Qiagen) is the most common procedure for detection of aquatic macroorganisms. Ojo-Okunola et al. (2020) found statistically significant differences in taxa prevalence from DNA extracted using ZymoBIOMICS™ DNA Miniprep kits compared to other kits for human milk bacterial profiling. [12] tested nine different DNA extraction methods using three commercial kits and compared them in terms of DNA extraction ability from human fecal sample. [13] proposed an optimized protocol for eDNA extraction from oligotrophic and degraded water samples using a specific kit. [14] evaluated the performance of four commonly used DNA extraction kits, including ZymoBIOMICS DNA Miniprep Kit, for studying cervical microbiota. [15] reported that different DNA extraction methods may cause changes in yield and stability, resulting in an inaccurate estimation of eDNA. [16] compared different methods for extraction and purification of environmental DNA from soil and sludge samples and focused on methods that are appropriate for the extraction of eDNA from very limited sample sizes (0.1 g) to enable a highly parallel approach. Therefore, it is important to carefully select the appropriate DNA extraction method and kit based on the sample type, target organism, and downstream analysis.
  • asked a question related to DNA
Question
6 answers
For the past few months, I've been plagued with this fluorescence that refuses to leave the wells of my Agarose gels. I'm running a nested PCR, so there are primary and secondary reactions with the same thermocycling conditions. I'm trying to purify Cryptosporidium parvum DNA. I've used the Qiagen DNA Stool kit to extract my samples. However, I don't think it's a DNA issue since even the negative controls are fluorescing!
N1 is the first negative control. I replace the template DNA with H20 in only the primary reaction. I then run the PCR and put that product (which contains water instead of DNA) into the secondary reaction and run the nested PCR. As you can see in the picture, it fluoresces just as brightly as the positive control. N2 is the other negative control in which I add only water to the secondary reaction and put that on a gel. No fluorescence at all. This leads me to believe that the cause of my sample hangup occurs because of the nested PCR. When I run a gel of just the PRIMARY PCR product, I don't get this "well fluorescence".
In terms of troubleshooting, I have replaced the primers, used 2 different Taqs (MyTaq Red & Thermopol), tried gel/PCR purifications, 2 different extraction kits (Qiagen & Omega), and tried 2 different annealing temperatures. However, the problem persists! I use a 1% Agarose gel run at 120V. I feel that I've exhausted almost every option. Does anyone have suggestions as to how I can avoid this pattern?
Relevant answer
Answer
Did you manage to fix this issue? How? I am experiencing this problem, too. Except I ethanol precipitate my amplified DNA before resuspending it in water REALLY WELL and running it on the gel. The samples just hang around in the wells and do not migrate down.
  • asked a question related to DNA
Question
3 answers
I'm working on Helvella phylogeny. I obtained pure DNA from isolation. I amplyfied four DNA regions (ITS, LSU, TEF, RPB2). However I usually see the non-specific bands (double bands, low quality bands especially in ITS region). Although I sequenced the good bands, the results of the sequences are very weird. My raw sequence data are very clear and there are not noisy peak. Interestingly, my sequences matched with other genus such as Hebeloma.
Helvella is a ascomycota and Hebeloma is a basidiomycota you know. I repeated my experiments and the results was not changed. I can't solve this problem. Can you help me.
Relevant answer
Answer
Believe the data. If the sequence data is good quality and it is matching up with something other than what you expect, you should believe that data but figure out why. It could be the organism you think you are working on is not really that organism but is something else. Or it could be that your culture is contaminated.
  • asked a question related to DNA
Question
4 answers
I am currently undergoing my end of year research project which is testing if DNA can be found in a secondary transfer when multiple transfers have occurred, one of my replicates have a Ct value of 0, do I include this or find an average of the remaining replicates as when I have calculated out the fold change of my replicates it has a value of 20.29
Relevant answer
Answer
You might need to provide more experimental design information.
Are you dealing with very low expected [template], such that Cqs are often 30+ (in which case "no amplification" genuinely implies "no target"), or are you dealing with fairly robust [template], i.e. Cqs of 20-30 (in which case "no amplification" means "freak weird event")?
If the former, the extent of quantitative info you can glean is going to be more limited anyway (stochastic template numbers will be inherently variable), and you should flag the well as "no amplification".
If the latter, you just ignore it.
  • asked a question related to DNA
Question
1 answer
The slides are not deparaffinized yet. I'm using a scalpel to scrape off the tumor area from it, however, the area of interest is very small. I had a hard time transferring the scrapping piece into the tube. I saw some youtube videos, they pre-fill the Eppendorf tube with 100% ethanol, then immerse the scalpel into the liquid to ensure all the pieces are collected. Is this a proper way to do it? , if so, how would I get rid of ethanol and proceed with the deparaffinized process?
My next step is to deparaffinized FFPE and extract RNA and DNA with Qiagen kit. Do you think I can pre-fill deparaffinized solution into the tube, and then immerse my scalpel into the solution to collect all the tissue pieces?
Relevant answer
Answer
Pure ethanol will evaporate if you leave the tube open. It does take a while though.
  • asked a question related to DNA
Question
1 answer
I have a structure I want to make, a tetrahedron with 3 full turns between the vertices, made of tri-star motifs. And I want to implement 3 nucleotides for a sticky ends to save one more for the motif binding, but strangely I don't see other people using 3. It's rather 2, or 4, or more. Is there a particular reason why?
Relevant answer
Answer
Dear friend Grigorii Rudakov
The choice of the number of nucleotides in a sticky end depends on several factors, including the size and complexity of the DNA molecule, the stability of the hybridized DNA, and the specificity of the interaction between the complementary sticky ends.
While it is possible to use three nucleotides for a sticky end, it may not be the most common choice because it can result in a weaker interaction compared to using two or more nucleotides. This is because a longer sticky end with more nucleotides provides a greater number of hydrogen bonds between the complementary base pairs, resulting in a stronger binding affinity.
Additionally, the length of the sticky end can affect the specificity of the interaction. Using a longer sticky end with more nucleotides can increase the likelihood of non-specific interactions between complementary sequences, leading to off-target binding and reduced efficiency in constructing the desired structure.
In your case, using a three-nucleotide sticky end may be sufficient for your specific design, but it is important to consider the potential trade-offs between specificity and binding strength. You may also want to consider using other strategies to increase the stability and specificity of the interaction, such as incorporating modified nucleotides or optimizing the sequence of the sticky ends.
  • asked a question related to DNA
Question
5 answers
I've been looking for methods for immobilizing 5'-Thiol modified dsDNA on gold slides. Most of what I've come across suggests using DTT followed by a desalting step or using TCEP. However, I haven't had any luck reducing and then conjugating the DNA onto the gold slides. I've seen some methods use DTT1,2, and some use TCEP3, and some include an incubation step with MCH2,3 after the thiol reducing step. It's been difficult finding a complete and satisfactorily detailed method online.
The DNA I am using is a 1 kb long double-stranded molecule with a 5'-Thio modification at one end and a 5'6-FAM modification at the other end. I need to synthesize the strands myself via PCR using two modified primers and a template strand rather than ordering the fully modified 1 kb strand, so my working concentrations are usually fairly low (~70-100 ng/uL after PCR and spin-column purification).
Can anyone provide a detailed method that has worked for them for reducing thiol modified DNA and immobilizing it onto gold surfaces or gold monolayers?
  1. Hegner, M., Wagner, P. & Semenza, G. Immobilizing DNA on gold via thiol modification for atomic force microscopy imaging in buffer solutions. FEBS Letters 336, 452–456 (1993).
  2. Ladik, A. V., Geiger, F. M. & Walter, S. R. Immobilization of DNA onto Gold and Dehybridization of Surface-Bound DNA on Glass. 5.
  3. Das, J. et al. Reagentless biomolecular analysis using a molecular pendulum. Nat. Chem. 13, 428–434 (2021).
Relevant answer
Answer
Is it usual to amplify DNA with thiolated Primer? Otherwise, you may need to check how thiolated primer affects PCR. I am not sure but thiolated primers might form di-thiol between themselves.
The immobilizing DNA amount might not be enough ( I did not check it).
During incubation of your DNA on the gold surface, it should not be dried. TCEP makes the solution acidic. I recommend you need to mix TCEP and DNA in a proportionate buffer. Usually, I used PB as the article. I am not sure that such long incubation. Anyhow, if you add salt such as NaCl to the incubating solution. It helps to increase the immobilized density of DNA because It reduces electrostatic repulsion between DNA. You might refer to these two articles and 10.3390/bios10050045.
Good luck
  • asked a question related to DNA
Question
1 answer
I have unstained FFPE tumor tissue, with the area of interest graphing on the back of the slides. The goal is to scrap off the area of interest, then extract both DNA and RNA.
What's the good order of the procedures?
Should I scrape the tissue first and then deparaffinized it? Someone also suggests me to bake the slides first and then scrape them.
Appreciate any pov!
Relevant answer
Answer
Hi Yun,
Generally, when using the whole slide, scraping is done after rehydration (post-xylene etc.). However, based on your question it looks like you do not want the whole section.
Right at the beginning before deparaffinization, you could use a very sharp blade and then cut around the area you want. Don't scrape it or anything and leave the area still attached. Make sure you make a good cut through the tissue. This would the best time to make the cut when the slide is untouched and section is firm. Then you can do deparaffinization with xylene etc. and when you finish, the rehydration step, it will be easier to just remove that area by scraping, or use a regular sharp shaving blade to "lift" off the area.
Trying to cut after the rehydration step you will definitely be dealing with more fragile tissue and cutting may not be that easy. Don't do too much baking etc. this will jsut degrade the RNA/DNA that much more.
Hope this helps!
Good luck!
  • asked a question related to DNA
Question
3 answers
Hi, I would like to ask if anybody has positive experiences with single primer PCR ? Can you recommend me any proven protocol of this type of PCR ? Thank you for all recommendations. Bohuš
Relevant answer
Answer
Hi , in selection of mismatches (SNPs) it easily works. Coupling flourcent dyes to such primers can convert PCR to RT PCR .
  • asked a question related to DNA
Question
4 answers
I want to synthesise mRNA using in vitro transcription. I am trying to the best way to design my cDNA template to be used for this purpose. I read lot of papers where some 5' and 3' UTR sequences are added to the coding sequence to enhance the stability of the mRNA. For example these sequences from human beta globin genes are often used. Can someone tell me howto chose these sequences ?
Relevant answer
Answer
Just curious, did you get good protein expression using beta globin gene UTRs instead of the genes specific UTR? I am thinking of using alpha or beta globin UTR too. Dolon Maji
  • asked a question related to DNA
Question
5 answers
While I am docking a metal complex with DNA in autodock I am getting following error:
I did DNA docking with this complex and not getting any error. But in running autodock (HSA docking) I am getting below error.
autodock4: too few values read in. Check grid map 'a' ! Real= 2.53, CPU= 0.63, System= 1.89 autodock4: FATAL ERROR: ERROR in readmap autodock4: Unsuccessful Completion Thank you.
Relevant answer
Answer
Could you write, please, which software you can recommend for docking including metal ions? Thank you very much in advance.
  • asked a question related to DNA
Question
2 answers
Folks,
Does anyone know of a paper showing binding affinity data of DNA oligos for their complement? Ideally it would show data from a 'non-molecular biology method' such as ITC, SPR, or MST. Even better would be a calculator for Tm to KD, and vice versa.
Thanks in advance,
Rob
Relevant answer
Answer
Thanks Adam, very useful. Not sure how I missed this one in my searching.
  • asked a question related to DNA
Question
9 answers
Using AGE, we are trying to check whether our DNA samples were amplified following PCR.
Here are the details of our gel run:
> Lane 1 is loaded with an expired 100 bp DNA ladder (for testing purposes, i.e. will expired ladders still present with distinct bands).
> Lane 2 is supposed to be a 236 bp product.
> Lane 4 is supposed to be a 667 bp product.
> Lanes 3 and 5 are each supposed to be 86 bp products after PCR using b-Actin primers.
Lanes 2 and 3 underwent the same PCR conditions (we followed the protocols stated in a published study that was able to produce the 236 bp product).
Lanes 4 and 5 were run in the same PCR conditions (according to published protocols that successfully produced a 667 bp product).
My question is (without minding the ladder) why does the supposed 236 bp product appear to align with the 86 bp product, considering their huge size difference? And why does the supposed 667 bp product appear only a small distance away from both of the aforementioned?
We know that the smaller sequences should travel the gel considerably further than larger ones.
Our predicament is that we are not sure if the appearing bands are really our amplified samples or just primer dimers/noise/whatever they are.
Thus, 1) may any of you please confirm if those bands are really amplified DNA. If they are, 2) what can we do to improve the distance/distinction between the bands besides increasing the agarose concentration?
FYI, gel is 3% w/v agarose. Samples were ran for 30 mins on 100 V.
Much thanks in advance to whoever answers!
Will happily share our PCR mastermixes and protocols if necessary.
***
Note: The images are of the same gel, just at varying contrasts.
Relevant answer
Answer
I would use a 1% agarose gel . Heat your ladder to 89c and allow to cool. The fragments sometimes reanneal to give better bands. I do not like the situation where the ladder runs slower than the product so borrow another ladder and run it as well in case your ladder has too much salt added and is running slowly. If the ladder is running well then all of your bands are the wrong size and too small so the bands may even be primer dimer. Run a pcr again but include one sample that has NO added dna and see if these sample generate a band in which case you either have contamination amplifying or you have a primer dimer in which case use a hot start Taq and use much less primer
  • asked a question related to DNA
Question
5 answers
Hello!
I am preparing a protocol to analyse some food and in the supplier user guide in the dilution step the say that you should mix the initial solution with water by vortexing for 10s after that you should spin briefly and store at 4°C.
my question here is: when I will take a volume from that tube for the next steps of my protocol I think that by spining the tube the DNA can form a pellet and this of course can affect my dilution and my concentration in the volume that I will use next?
Thank you so much
Relevant answer
Answer
If your dna is already in solution then spinning down will move all of the wetting on the tube sides into the solution at the bottom which will minimise the loss of water to evaporation in storage although small volumes will evaporate even when stored in a freezer.
Your dna will remain in solution during spinning and storage because addition of salt and alcohol is needed for dna precipitation. Once your dna is dissolved it should stay dissolved even in freezing and thawing
  • asked a question related to DNA
Question
4 answers
Good afternoon,
I am carrying out a monthly invertebrate sampling for future molecular studies (DNA). I am euthanizing my arthropods with 70° ethanol right after the capture and then store them in a freezer. Would it be better for DNA preservation using 96° or pure ethanol?
Relevant answer
Answer
I have heard, but do not remember the source, that 100% alcohol/absolute alcohol can be used for DNA studies. Normally 70% alcohol is used for storage in alcohol. Some recommend 70% with 5% glycerol for long-term storage. If possible, freezing, as mentioned, is best for DNA studies. The colder the better. -18 degrees C, is standard in household freezers. Dry ice holds - 78o and can be used for short-term freezing. Liquid nitrogen is even colder, but requires special equipment.
  • asked a question related to DNA
Question
3 answers
I have done a blue-white screening with E.coli and pUC19 with lambda DNA. Then, I selected some white colonies (and blue colonies as a control) and gel electrophoresed them next to pure pUC19. All lanes have a band similar to pure pUC19. I am stuck on why this is the case. Any help is appreciated!
Relevant answer
Answer
Dear Emilie,
The solution may be very simple: if you clone in Lambda DNA digested with PstI into PstI-digested pU19, and then digest all of the plasmid DNA samples produced from cultures inoculated with blue or white colonies and selected with ampicillin, then all of your samples will include a band at 2.7 kb corresponding to ... pUC19 ... which is the source of the ampicillin resistance and will turn the colonies blue if there is no insert of DNA at the PstI site.
Your gel images suggests that a number of your samples contain Lambda DNA inserts, but since the band intensities vary a lot, I'd be a little careful in interpreting some lanes with bands above the pUC19 band, as this may represent partially-digested plasmid DNA. Lanes 3, 6, 7, 9, 10, 13, 15 and 16 look interesting. You could look at a Lambda restriction map to see the range of PstI fragments it would produce, and map them onto the fragments you can see in your gel.
Regards, Andrew
  • asked a question related to DNA
Question
4 answers
For context, I am trying to digest my vector with HindIII-HF (from NEB) and then I will be phosphatase treating it with Antarctic Phosphatase (from NEB). For the digestion, I've prepared my samples as shown below:
  • DNA = 600 ng
  • Cutsmart Buffer = 5 uL
  • Restriction Enzyme = 1 uL
  • H2O = fill up to 50 uL
I digested my samples at 4 different incubation time at 37C: 15 minutes, 30 minutes, 1 hour, and 2 hours (some photos are included) . But what I noticed was 15 minutes and 1 hour, the DNA wasn't there and the gel was smeared. However, for the 30 minutes and 1 hour, the digestion worked?
Does anyone know why my gel might be smearing and why my DNA seems to be disappearing? I created my DNA fresh (1 day old - stored in -20C and thawed only once). For anyone suggesting that maybe I don't have any DNA, I ran 1 uL of each onto a gel and DNA were present.
Relevant answer
Answer
My question will be, why do you have such a strong smear below your DNA bands?
Is this a plasmid?
Looks like your samples are contaminated with DNAase.
Depending on the host bacteria and extraction kit you might need to do a washing step.
  • asked a question related to DNA
Question
1 answer
Good day! The question is really complex since CRISPR do not have any exact sequence - so the question is the probability of generation of 2 repeat units, each of 23-55 bp and having a short palindromic sequence within and maximum mismatch of 20%, interspersed with a spacer sequence that in 0.6-2.5 of repeat size and that doesn't match to left and right flank of the whole sequence, in a random sequence.
Relevant answer
Answer
First, I'd re-state the question to assure that I understood it correctly. A nucleotide sequence of length l contains a palindrom with unit of length k. The palindrom is not exact; there can be from kmin to k matches between units. The distance between palindrom units can be from smin to smax. First and last sub-sequences of length k are not exact matches of any palindrom unit.
My solution. Let's omit the last condition for now. How we search for a palindrom with unit of length k? Take any subsequence of length k and search for a 'match'. Searching for a 'match' is equal to checking (l-k-smin) subsequences, because the unit itself occupies k nucleotides and a spacer can't be shorter than smin nucleotides. In each window the probability of hit is (1/4)^(kmin), if every nucleotide has equal probability of occurrence. The probability of having 1 or more hits then is equal to binomial cdf with the number of attempts equal to n-k-smin, the probability of success equal to (0.25)^kmin and number of successes equal to 1. For example, GSL function gsl_cdf_binom_Q(n-k-smin,0.25^kmin,0) would give the answer. The last paramerter is zero, because the function computes the probability of more than x successes, i.e. 1 and more in this case.
Now, let's include the last condition. It is important to define what 'does not match' mean. I suppose that it means that we can't find the second palindrom unit at postions 1 and l-k. So, the number of windows that we check has to be decreased by 2. The final answer would be:
F(n-k-smin-2,0.25^kmin,0), where F - binomial cdf.
For varying length the answer would be a weighted sum of those propabilities, with weights equal to the probability of observing given legnth. So, if all lengths have equal probability, this is the mean.
I checked the answer on a synthetic set and it seems it is correct or close to being so.
  • asked a question related to DNA
Question
9 answers
I am having problem with 16S amplification on colon content DNA from DSS treated group. I have found that DSS is a polymerase inhibitors and tried to purify DNA with LICL and Glycogen-ethanol precipitation. I have also tried to dilute my stock DNA before PCR. However, there is no successful 16S amplification. I have attached a gel electrophoresis result ( tapestation report) on PCR product of my samples and E.coli as control for your reference. Only control showed 200bp band but all my samples showed no amplification. Any suggestion would be greatly appreciated.
Relevant answer
Answer
Hello all,
Thank you so much for your answer. Lately I have tried DNeasy PowerClean Pro Cleanup Kit for an additional clean up process of DNA samples and ran PCR. I think I saw some bands in the region of 200-300bp.
  • asked a question related to DNA
Question
9 answers
Well,
I am a very curious person. During Covid-19 in 2020, I through coded data and taking only the last name, noticed in my country that people with certain surnames were more likely to die than others (and this pattern has remained unchanged over time). Through mathematical ratio and proportion, inconsistencies were found by performing a "conversion" so that all surnames had the same weighting. The rest, simple exercise of probability and statistics revealed this controversial fact.
Of course, what I did was a shallow study, just a data mining exercise, but it has been something that caught my attention, even more so when talking to an Indian researcher who found similar patterns within his country about another disease.
In the context of pandemics (for the end of these and others that may come)
I think it would be interesting to have a line of research involving different professionals such as data scientists; statisticians/mathematicians; sociology and demographics; human sciences; biological sciences to compose a more refined study on this premise.
Some questions still remain:
What if we could have such answers? How should Research Ethics be handled? Could we warn people about care? How would people with certain last names considered at risk react? And the other way around? From a sociological point of view, could such a recommendation divide society into "superior" or "inferior" genes?
What do you think about it?
=================================
Note: Due to important personal matters I have taken a break and returned with my activities today, February 13, 2023. I am too happy to come across many interesting feedbacks.
Relevant answer
Answer
It is just coincidental
  • asked a question related to DNA
Question
4 answers
Hi all.
I am investigating the impact different medication has on the gut microbiota in a range of samples. From what I understand, a qPCR would be an appropriate method followed by quantification. What method of quantification would be best for this study?
Would using gel electrophoresis be best, or using a fluorometer, or something completely different?
Or would a ddPCR be a more suitable method to quantify and compare gut microbiota concentrations?
Relevant answer
Answer
First you should specify if your are interested in a particular gene, its gene expression, or just genomic DNA. If you are interested in a gene or a specific genomic or plasmid region, then qPCR is a good method. Just design primers for that, and for a control gene or region, and build a standard curve with known concentrations or copy numbers to extrapolate fluorescence intensity values
  • asked a question related to DNA
Question
3 answers
I have run an agarose gel electrophoresis for DNA and RNA, and I was wondering if ImageJ or Fiji can measure the purity of the sample, as well as quantify it. I was planning on using a standard UV-Vis spectrophotometer, unfortunately, the UV-Vis broke down. Hoping someone could enlighten me.
Relevant answer
Answer
If you have a plasmid or PCR product, you can estimate the concentration by comparing your sample to a known DNA ladder. The product insert will tell you the specifics for the ladder. ImageJ could make it a bit more quantitative, but you still need samples with a known concentration as a reference. Also, know that ethidium bromide (or SYBR Safe or whatever stain you use) will have an upper saturation limit.
No one every bothers to quantify a DNA extraction from eukaryotes on an agarose gel. All you would ever see is a faint smear. If you really need to know the concentration of genomic DNA, ask to borrow a spec or nano drop. The chemistry department should have one.
You will get a rough estimate of the "quality" of your RNA by looking for 2 bright rRNA bands + a faint tRNA smear on an agarose gel. In fact, it's best practice to use an agarose gel rather than a spec since the spec won't tell you if the RNA is degraded.
Good luck!
  • asked a question related to DNA
Question
4 answers
thanks.
Relevant answer
Answer
Sure Azka Saleem
Steps u can consider following.
1. Download the SGD data as a fasta file/if separated as many files, u can merge all into single fasta file.
2. Make a list of IDs in MS excel whose sequences you need to extract.
3. Install TB tool in your dekstop, and search for fasta extract option, where u need to provide the database fasta file and the IDs and click ok.
4. In the output file you will have your required IDs sequence data.
If you still find it difficult to perform, u can mail me in [email protected]
  • asked a question related to DNA
Question
3 answers
Certain softwares and sites allow to calculate a DNA hairpin Tm depending on the size of the loop and the stem sequence. For example, Gene Runner. Yet the calculation method or citation is not provided. Is there a formula that could help?
Relevant answer
Answer
DOI: 10.1039/b804675c
This paper explains very well how unfolding and melting of DNA hairpin works. kindly have a look.
  • asked a question related to DNA
Question
3 answers
I'm facing small issues in understanding the topic of Genetic Recombination, please guide me to understand the topic.
Thank you.
Relevant answer
Answer
The process or act of exchanges of genes between chromosomes, resulting in a different genetic combination and ultimately to the formation of unique gametes with chromosomes that are different from those in parents is what is called genetic recombination. In this process, there is rearrangement of DNA sequences by the breakage and re-joining of chromosomes or chromosome segments.
Two very close-together genes will have very few recombination events and be tightly linked, while two genes that are slightly further apart will have more recombination events and be less tightly linked.
So, the linkage is indirectly proportional to the distance between genes, the less the distance between the genes, the less is the cross over frequency between them and the more is the linkage frequency between them.
Best.
  • asked a question related to DNA
Question
2 answers
For windows I usually use Bioedit for alligment and sequence analysis but I'm looking for a free alternative for mac
Relevant answer
Answer
hi Jessica
you can also use chromas which is worlwidely used (http://technelysium.com.au/wp/chromas/).
all the best
fred
  • asked a question related to DNA
Question
3 answers
We know that Restriction enzymes cut/destroy the DNA, but "Does it cut the DNA of the same organism that makes them?"
If they cut so, How and where does it cut?
If they don't cut, Why wouldn't they?
Relevant answer
Answer
Don't use this website to ask for help on your homework.
  • asked a question related to DNA
Question
8 answers
I'm looking for a method to extract DNA from desiccated crocodile scutes to be used in a genomic DNA study. Is there a best method to extract DNA from these tissues?
Relevant answer
Answer
This is very good and relevant question. I think same with extraction procedure tissues of animals.
  • asked a question related to DNA
Question
4 answers
Hi,
I need to buy the spectrophometer and after research I have selected three items:
1. Uv/vis spectrophotometer named Nabi by Biogenet;
2. NanoReady F-3100 by Syngen;
3. NanoReady F-2100by Syngen.
Do anyone has an experience with some of them? Can anyone share the opinion? Currently, I am thinking about Uv/vis spectrophotometer named Nabi by Biogenet and NanoReady F-3100 by Syngen as both has similar performance but Nabi is significantly cheaper. Do anyone has Nabi spectrophotometer? Please share opinion.
Thank you in advance!
Relevant answer
Answer
I have similar question. we would like to buy spectrophotometer for plant phyisology lab. what is your opinion about Thermo Scientific
GENESYS Vis and UV-Vis Spectrophotometers? or any other suggestion?
to measure chlorophyll, some enzyme, protein and ...
  • asked a question related to DNA
Question
4 answers
I have YOYO-1 dye and I want to add dye at 5:1 ratio. And I am using a plasmid of length 5411 bp. Please tell me how to calculate this ratio to get a volume of dye to be added to DNA of 1 microgram.
Relevant answer
Answer
How it being calculated attached file please?
  • asked a question related to DNA
Question
8 answers
Kindly discuss your ideas and viewpoints on the origin of life and the RNA world hypothesis.
What are the contradictory views on why researchers are still unsure about the origin of life through RNA or such analogous molecular intermediate pre-cursors preceding its existence?
"The general notion of an “RNA World” is that, in the early development of life on the Earth, genetic continuity was assured by the replication of RNA and genetically encoded proteins were not involved as catalysts. There is now strong evidence indicating that an RNA World did indeed exist before DNA- and protein-based life. However, arguments regarding whether life on Earth began with RNA are more tenuous. It might be imagined that all of the components of RNA were available in some prebiotic pool and that these components assembled into replicating, evolving polynucleotides without the prior existence of any evolved macromolecules. A thorough consideration of this “RNA-first” view of the origin of life must reconcile concerns regarding the intractable mixtures that are obtained in experiments designed to simulate the chemistry of the primitive Earth. Perhaps these concerns will eventually be resolved, and recent experimental findings provide some reason for optimism. However, the problem of the origin of the RNA World is far from being solved, and it is fruitful to consider the alternative possibility that RNA was preceded by some other replicating, evolving molecule, just as DNA and proteins were preceded by RNA." - Robertson and Joyce
[This is as per the explanation by Michael P Robertson and Gerald F Joyce in the article: "The origins of the RNA world." published in the Cold Spring Harb. Perspect. Biol. 4, a003608 (2012).]
The scientific community must resolve this contradicting conjecture through rational discussion and debate backed by strong experimental evidence on what must be the pre-cursor molecule to the Origin of Life if it is not RNA!
Relevant answer
Answer
One of the issues that is holding the concept of an RNA world back from being more scientifically useful - irrespective of whether there ever was such a thing - is that we don't use the idea in the scientific way it was intended. Just like any other prebiotic scenario, it is not (nor has it ever been) a scientific hypothesis. In fact, scenarios are usually not intended as such. Scenario authors from all niches (including RNA world) have pointed out that scenarios themselves are untestable. However, they guide thinking and allow to conceive of hypotheses that are testable. If we go through the old literature we find very explicit passages to support this fact.
For the specific authors advanced in the question, G.F. Joyce and L.E. Orgel, we have a passage from 1999 in "prospects for understanding the origins of the RNA world". (The RNA World 2nd ed. 49-77).
"The presumed RNA World should be viewed as a milestone, a plateau in the early history of life on earth. So too, the concept of an RNA World has been a milestone in the scientific study of life's origins. Although this concept does not explain how life originated, it has helped to guide scientific thinking and has served to focus experimental efforts."
You can find this point of view expressed in foundational work for all niches related to the popular scenarios today. But you can also find it for scenarios most people in origins have never heard of. E.g. the idea that celllular life started with terpenoids found in G. Ourisson and Y. Nakatomi's "the terpenoid theory of the origin of cellular life: the evolution of terpenoids to cholesterol. (1994) Chem & Biol. 1 11-23".
"The hypothesis provides an attractive way of ordering the terpenoids: like all evolutionary theories, it cannot be tested directly. The ideas summarized here do, however, suggest a multitude of experiments having some bearing on the fundamental and fascinating question: how did the first cells appear? We hope to carry out some of them."
A related line of thought - but highly influential - is the exposition by Harold J. Morowitz from 1992 in his book "Beginnings of Cellular Life: Metabolism Recapitulates Biogenesis". If we go to the conclusion, we find this explicit clarification on the distinction between a genuine scientific theory and a scenario:
"at this stage of the thought process, it is important to focus on the hypothesis that intermediary metabolism recapitulates prebiotic chemical evolution. This hypothesis is not a strictly vulnerable theory in the Popperian sense, but it does provide us with a valuable heuristic method for using modern knowledge of biochemistry to search for events that have left their trace. If the intermediary metabolism of autotrophs does not recapitulate biogenesis, then the discontinuities will have to be explained."
More than 2 decades back, many authors made a clear distinction regarding this nuance. Scenarios are here to help: they guide thinking and design experiments. They only guide thinking in a scientifically meaningful direction as long as we can easily abondon scenarios and enthusiastically continue replacing them with new, more informed scenarios. A situation where a scenario gets entrenched and where researchers treat it as a scientific hypothesis is - by construction - hard to escape.
In fact, this is exactly the situation that many researchers have described around the 80s, when criticism mounted against the prebiotic broth scenario. The passage from Wächtershäuser's 1988 "Theory of a Surface Metabolism" is telling:
"The prebiotic broth theory has received devastating criticism for being logically paradoxical (11, 135), incompatible with thermodynamics (11, 144, 160), chemically and geochemically implausible (134, 136, 144), discontinuous with biology and biochemistry (160), and experimentally refuted (135, 160). The reason for the tenacity with which it is retained as accepted dogma has been forcefully and clearly stated by Scherer (126): "If this rejection is substantiated, there will remain no scientifically valid model of the selforganization of the first living cells on earth."
Clearly, the broth scenario had overstayed its welcome. One reason for this is that its 'claims' (which for a scenario can only be speculations) were too much in contradiction with claims from fields of science that do not suffer the same restrictions when it comes to testing and refuting their theories. One example of a very controversial idea that can be found in Haldane's formulation of a broth scenario, is the purported necessity of a long, highly functional protein randomly emerging from a soup, as an extremely rare event: we expect this to be prohibitively unlikely and hence a far from parsimonious explanation.
Quite a few of the critiques voiced against the prebiotic broth scenario are equally valid critiques of some scenarios we have today, including RNA world.
The RNA world is an old and multifaceted concept. There are contrasting formulations that make different claims (to be interpreted as speculations) about history. As with the prebiotic broth scenario (and any scenario), it has raised genuine scientific objections. These have remained largely unadressed, in spite of its long dominance.
It is instructive to bear in mind that scenarios don't come from nowhere. They're fairly detailed speculations about purported historical events. To make them, each author makes assumptions. Some of these concern speculations that later became testable, e.g. about chemistry and physics. You will find different scenario authors make different assumptions and different arguments (and flaws therein). There's an inevitable bias here with respect to the fields an author is trained in. Some of the foundational assumptions in popular scenarios like RNA world are at least 50 years old, but some unchallenged assumptions date back to a literature that is more than a century old. A time before IUPAC, modern quantum mechanics, genetics, and so forth.
That has been enough time to forget that scenarios like RNA world are by construction not testable hypotheses and that they were not intended as such. Scenarios are here to guide thinking, to inspire experiments. The best thing a scenario can do for us, is generate insights that spur us to change the way we think and thereby necessitate replacing our old scenarios with new ones, and repeat the cycle. The science coming out of the community today is a lot more conducive to doing that than previously.
The same cannot be said for the rather myopic RNA-centric framing of a question in the cited passage, which attempts to elevate RNA world to more than a scenario. Rather than forcing ourselves to think about the rather narrow and outdated proposal by Joyce and Robertson, ("consider the alternative possibility that RNA was preceded by some other replicating, evolving molecule"), it is more productive to critically revisit all the things that have been assumed and argued when the concept of an RNA world was conceived and how which of these premises are considered valid or plausible today, and which ones back then. Is there a formulation of RNA world for abiogenesis that is logically sufficient? And if so is it logically necessary that abiogenesis proceeded this way?
It is also instructive to check how much of the logic was sound. e.g. the rhetorical tricks employed in RNA world introduce all sorts of hidden assumptionsm.
As an example of the latter: some still justify an RNA world by the party trick 'chicken-and-egg' question 'protein or RNA, which came first?', only to conclude with 'RNA, it encodes proteins' and hastily conclude with an even stronger 'RNA-first' for abiogenesis. 'chicken-and-egg' fallacies are nothing new in origins. In fact, they were already identified as such long ago. E.g. in chapter 8 of "Seven Clues to the Origin of Life (1985)" by Cairns-Smith, there's an illustrated passage detailing that these types of paradoxes in origins frame the question in a manner that prevent us from considering scaffolds.
"
The fact is that even the so-called simple organisms such as E. coli are very complex enterprises with all sorts of things going on together. There is plenty of scope for accidental discoveries of effective new combinations of subsystems. It seems inevitable that every so often an older way of doing things will be displaced by a newer way that depends on a new set of subsystems. It is then that seemingly paradoxical collaborations may come about.
To see how, consider this very simplified model - an arch of stones: This might seem to be a paradoxical structure if you had been told that it arose from a succession of small modifications, that it had been built one stone at a time.
scaffolds that starts like this:
This might seem to be a paradoxical structure if you had been told that it arose from a succession of small modifications, that it had been built one stone at a time. How can you build any kind of arch gradually? The answer is with a supporting scaffolding. In this case you might have used a scaffolding of stones. First you would build a wall, one stone at a time:
Then you would remove stones to leave the 'paradoxical' structure.
"
It should be noted that in 2022, even in RNA-world, very few scholars remain that find RNA-first a convincing idea. As a scenario, however, it is not useless: it is instructive to consider what the underlying ideas are that at some point in time made such a highly specific idea compelling to so many of us.
A fixed motif in scenario papers is to start explicitly and implicitly assuming a few things about what chemistry can and cannot do and some properties of abiogenesis. These sort of assumptions used to be spelled out routinely, also outside scenario papers. Let me give two examples.
The original 1953 paper for the "Frank Model" "on spontaneous asymmetric synthesis", has the passages
".. the defining property of a living entity the ability to reproduce its own kind ...
confining attention to chemical molecules, the complexity of any having this essential property of life is likely to be great enough to make it highly improbable that it has a centre of symmetry."
(*I should point out that Frank makes an important error here: the capacity for molecular reproduction is not a molecular property but a property of a reaction network. If we add an additional thermodynamic criterion this property is autocatalysis and we can then check this claim from the IUPAC definition: https://goldbook.iupac.org/terms/view/C00876. It turns out there are trivial ways to make small networks that have this property https://chemrxiv.org/engage/api-gateway/chemrxiv/assets/orp/resource/item/60c74d67469df42226f44295/original/emergent-autocat-animation.gif.)
The point to retain here is that Frank considers it to be generally accepted that one can assume this property to be prohibitively rare in chemistry. This belief was wideheld, and we can e.g. read in "the units of selection" (1970) by Lewontin a summary on scientific views on abiogenesis
"The present view ... Since there was no autocatalysis, there was no reproduction or heredity and so no possibility of natural selection."
The coacervates in Oparins scenario were notably invoked to adress this issue.
When it comes to assumptions in scenarios, this systematically involved conjecturing that chemistry 'in the wild' intrinsically and deterministically becomes a 'mess', undergoing no meaningful complexification, and for which no reproduction and evolution can reasonably be expected. From there, it appears that no process of abiogenesis should conceivably occur naturally, and thereafter some specific sequence of exceptional events is proposed as plausible, because it appears to be the sole contender.
Let us make more explicit why this is not an innocent procedure:
We still find our understanding of 'basic chemistry' to be plagued with limitations and long-lived misinterpretations (e.g. 2 days ago we learned that methyl substitution destabilizes radicals instead of the textbook knowledge that it stabilizes them ).
Moving beyond the basics, we by and large lack a lot of formal theory, experiment, or even a simple reference frame for the things that happen then. Joyce and Robertson honor the tradition of purporting from the outset that 'chemistry in the wild' becomes an intractable mess. The issue is that we don't know at all if that's the case. We cannot assume this from the outset, we need to extensively study it. We require extensive experiments and theory and a reference frame for all the phenomenology associatied with complex systems (e.g. multiple components, compartments, multiple forms of nonequilibrium driving, length scales, time scales).
In making the routine assumption of 'messy, intractable chemistry that can neither complexify nor multiply', we have decided in advance that, once we finally understand 'chemistry in the wild' with its 'so-called intractible mixtures', it cannot have any bearing on abiogenesis. Let alone explain it.
That is a disproportionately bold conjecture about fundamental science, and a very consequential one: all historical scenarios - RNA world being one out of many - have been justified by formulating conjectures of this sort (many authors also insist on other properties, e.g. chemistry being deterministic). Clearly, it should be the first priority of everyone in the field to test this conjecture, by extensively and rigorously studying complex chemical systems as an end in itself. If the conjecture is correct, it provides an important validation for historical scenario approaches. If the conjecture turn out wrong, we are in a much better position to conceive of more scientifically informed scenarios, but potentially the approach will change entirely.
In presenting it as such, I am making it appear as if it could be an open question whether the chemical conjectures underpinning our scenarios in origins may be true or not. In fact, we have learned quite a few things in the meantime. And some clumsy mistakes were made elsewhere.
- Determinism:
When it comes to chemistry being deterministic (a key tennet of e.g. Sutherlands scenario and Wächtershäusers surface metabolism): upon critical evaluation of what is known of basic chemistry this idea becomes unacceptable, especially when considering the chemical processes on the surface of a planet, as opposed to a quick reaction in pyrex.
1) insofar as it is reproducible, modern chemistry owes much of it to big strides of standardization in glassware, methodology, synthesis protocols (e.g. usage of stirring bars).
2) lab chemistry exhibits many forms of contingency. This is particularly the case when it comes to phase behavior, e.g. habit modification, polymorphism. Aspirin purportedly has 8 reported polymorphs, phenobarbitone 13.
3) glassware is cleaned between reactions, thereby making successive reactions in the same glassware independent. In nature, this property of independence is absent. In fact, effort to make an evolutionary classification of minerals are rooted in the opposite: that certain minerals start to form conditional on the presence of certain others. (https://pubs.geoscienceworld.org/msa/ammin/article/104/6/810/570840/An-evolutionary-system-of-mineralogy-Proposal-for)
- Autocatalysis:
A first issue to get out of the way is the misconception that autocatalysis is prohibitively rare. A prominent PI in origins (RNA world, not a chemist) told me that chemists throughout history have found exactly one example. Claims about the contents of a literature one cannot realistically have read in a lifetime is a common error we can find in the origins literature. Below are some reviews.
I should stress that these reviews discuss examples from a few niches in chemistry. These reviews do at least allow to have 100s of counterexamples to dubious claims about no autocatalysis in chemistry, but it's only a small fraction. Virtually all branches of chemistry have regular reports of autocatalysis, but very few focus on autocatalysis in its own right. And hence most branches do not review their reported examples.
By critically examining the IUPAC definitions, one can show that autocatalysis is dramatially more widespread than long thought. In part, this is because the definition applies to a wealth of situations where the term is not routinely employed. By examinging the requirements of autocatalysis as an emergent network property, one can demonstrate that this property emerges particularly readily in a heterogeneous / multicompartment context. With the disclaimer that I'm an author I refer to the following:
- Messy chemistry:
Refreshing counterexamples are afforded by the literature on systems chemistry and dynamic combinatorial libraries.
In the context of origins, a recent work that is greatly aiding in fixing our misconceptions is : https://www.nature.com/articles/s41557-022-00956-7
Where a reaction of purported immense complexity is found to exhibit highly reproducible and ordered behavior as function of environmental inputs. How chemistry exactly works on this level is still poorly understood. I think I do, but it'll have to await peer review. But we cannot in good scientific conscience take for granted anymore that chemistry becomes messy and intractable. When we do the experiments, we see something very different.
in conclusion, I want to come back to the final point of the question
"The scientific community must resolve this contradicting conjecture through rational discussion and debate backed by strong experimental evidence on what must be the pre-cursor molecule to the Origin of Life if it is not RNA!"
No. The sientific community should strive to do what it can justify scientifically. Those that find it fruitful to relegate the RNA world - which is not a hypothesis - are justified in doing so. Notably because it is is founded on scientifically refuted premises and logical errors.
Those that find ways to make it fruitful to keep it are justified in doing so: it's a scenario, one can draw inspiration from it. Perhaps a thoroughly altered version can be developed that fixes previous issues.
Above all else, RNA is an amazing molecule that has been used for fundamental research that concerns everyone in origins, and will continue to do so irrespective of how serious the RNA world scenario is still taken.
What the origins of life community needs, first and foremost, however, is concern itself with more important matters.
Complex chemistry needs to be studied thoroughly on an experimental and theoretical level.
New scenarios are needed. And these scenarios should no longer require chemistry to have properties it doesn't have, and vice versa. These scenarios should also explicitly be appraciated for what they are, an for what they're not. They're here to help, to guide thinking, inspire experiments, produce testable predictions, update our beliefs. They are not scientific hypotheses in and of themselves.
  • asked a question related to DNA
Question
3 answers
Hi all,
I generated mutant lines using the CRISPR/Cas9 method, and my plants are in the T1 generation.
I am trying to send PCR products for Sanger sequencing, but the problem is, the positive control that shows me two bands. I borrowed wild-type genomic DNA from another colleague and got the same result.
I used my primers for genotyping in the previous generation without any problems, so my primers are specific.
I attached my results: Pic 1 shows extracted DNA from leaves by the CTAB method. Pic 2 shows the PCR result of WT and one of the mutant lines. As you can see, I have two bands in the WT lane but only one band in the mutant lane, and Pic3 displays PCR results of other mutant lines with one/two bands. Besides, I did not get any bands for some lines. To check those lines, I used cas9 primers which were successful.
The NC lane is clear, so I don’t have contamination.
I did gradient PCR, changed enzyme, and changed denaturing, annealing and extending time, but they did not work.
I am running out of ideas. What could cause this issue and any suggestions to address it?
Thank you so much in advance for your help!
Relevant answer
Answer
Looks like you have non-specific bands. And you are really over-loading your gel - try loading less PCR sample in the wells.
I second the vote to design some new primers. Primers are cheap, endless trouble-shooting is expensive.
Good luck!
  • asked a question related to DNA
Question
5 answers
Hi,
I am using a technique in C2C12 muscle cells called DamID to tag DNA in close proximity to the nuclear envelope in undifferentiated myoblasts and differentiated myotubes. However, following sequencing of the tag-enriched material amplified by the final PCR step, 80% of the DNA sequences identified come directly from the mitochondrial genome.  So it looks like the mtDNA is being tagged and because per copy it is more numerous than any genomic sequence, gets preferentially amplified.
I have tried removing the circular and supercoiled mtDNA by;
  1. subtractive hybridization (purifying mtDNA, fragmenting it, biotinylating it, hybridizing it material to my sample and then pulling down the hybrid duplexes with Dynabeads)
  2. Nuclear isolation (hypotonic lysis, dounce homoginisation, centrifugation)
  3. CsCl gradients (using the density difference acquired by linear and supercoiled DNA following addition of ethidium bromide)
However, none of these have worked. Does anyone have any suggestions on easy ways to remove the mtDNA from the genomic DNA? The last thing I am thinking of trying is gel filtration?
Any help would be great!
Relevant answer
Answer
I find one paper use Cas9-assisted removal of mtDNA.
  • asked a question related to DNA
Question
3 answers
I performed a double digest of my PCR products and vector in CutSmart buffer followed by heat inactivation.
Unfortunately, the PCR purification kit is empty and ordering takes longer than expected, so I couldn't perform a purification/gel extraction of my DNA for downstream cloning application directly after the digest.
The products were stored at -20 °C directly after heat inactivation for over a week now.
How long can I store the DNA under these conditions and still use it reliably for ligation afterwards? Or is it advisable to start over again from scratch.
Thank you for your help.
Best,
Fabio
Relevant answer
Answer
I would expect dna frozen in buffer to be stable for many years/decades
  • asked a question related to DNA
Question
3 answers
Hi,
I wonder if anyone knows how to use sample release reagent from Sansure Biotech ?
Relevant answer
Answer
Hi,
The supplier claims that their "sample release reagent" (nucleic acid release technology), can lyse pathogens at room temperature very fast, with no need to heating, centrifuging or replacing tubes, the sample DNA/RNA can be extracted quickly through simple operations. The reagent is applied for the pretreatment of nucleic acid molecules, to release them from specimens, then the released nucleic acids can be used for clinical in vitro diagnosis or for the detection through appropriate apparatus.
  • asked a question related to DNA
Question
196 answers
Does the DNA remain stable or degrade at this temperature? Would there be any difference in thermal stability between supercoiled and linear forms of say, 3 kb plasmid.
Relevant answer
Answer
If we heat up a tube of DNA dissolved in water, the energy of the heat can pull the two strands of DNA apart (there's a critical temperature called the Tm at which this happens). This process is called 'denaturation'; when we've 'denatured' the DNA, we have heated it to separate the strands. The two strands still have the same nucleotide sequences, however, so they are still complementry. If we cool the tube again, then in the course of the normal, random molecular motion they'll eventually bump into each other ... and stick tightly, reforming double-stranded DNA. This process is called 'annealing' or 'hybridization', and it is very specific; only complementary strands will come together if it is done right. This process is used in many crime labs to identify specific strands of DNA in a mixture. Now, when we've denatured the two strands, there's something else we can do - replicate the DNA. The key here is that any single-stranded piece of DNA can only hybridize with another if their sequences are complementary. If we have just one strand, we can actually buildanother strand to match it. Here's how it's done, either in a test tube or in a live cell: The DNA strands are separated (for example, by heating them in a test tube). For each strand, we provide a primer, which is a short piece of DNA that sticks to one end of the strand. An enzyme is added. This is a specific type of protein called a "DNA polymerase" that can "read" the bases on one strand and can attach the complementary base to the growing strand. The polymerase "walks" down the template strand and creates its exact complement as it goes. The same thing happens to the other original strand. When we started, we had one double-stranded piece of DNA. After polymerase is done, we've got twoidentical pieces - exact copies of each other.
  • asked a question related to DNA
Question
2 answers
Hello, all! I'm performing an experiment in which I'm doing a CoIP in the presence or absence of RNase to see if a protein-protein interaction is dependent on RNA. I am trying to identify a protein pair that is known to interact in an RNA-dependent manner, particularly in mouse embryonic stem cells (mESCs). I was thinking of trying to identify some proteins involved in translation initiation or spliceosome assembly/function, but am unfamiliar with the biology of these complexes.
Any and all help is greatly appreciated!
Relevant answer
Answer
In case anyone is curious about this question, I've found that the U1 snRNP complex components U1A and U1C interact in an RNA dependent manner and give a great Western blot signal. I'd recommend them as a control for RNAse degradation.
  • asked a question related to DNA
Question
3 answers
I tried testing for qPCR inhibition in my samples by running a dilution series of my original sample and four ten fold dilutions. All series have a similar curve as seen in the three images I included in this question. Is this a normal result to have when you expect inhibition? I read somewhere that while the efficiency may be higher due to inhibition, the R2 still needs to be 0.98/0.99. Do the curves look like this due to pipetting errors or is there another explanation?
If this is not the best way to test for inhibition, what could I do instead?
Relevant answer
Answer
All the curves are sloping off at cycle ~30 because at this point you've diluted your sample so much that there's almost no target there: the assay isn't sensitive (or accurate) enough to quantify reliably at this dilution.
Basically, don't do it this way. Assume neat cDNA will always be inhibitory, but any cDNA diluted by a factor of 10 or more (I use 20-fold dilution, personally) will be fine.
And you'll have more cDNA (in volume terms) to play with, because you've diluted it.
  • asked a question related to DNA
Question
1 answer
Dears researchers,
Has anyone had the error below when calibrating the Step One Plus equipment (Thermo Fisher Scientific)?
"Spatial Calibration failed: Well locations are not evenly spaced.
System will revert to previous calibration.
Exit the calibration wizard and refer to the Hrlp to troubleshoot the calibration failure.
Error Code 1302"
I can't find the error code in the troubleshoot. Company support has not found a solution yet.
I already did the decontamination and the Backgroud calibration worked.
Regards,
Relevant answer
Answer
We also had a problem and after consulting with the experts of Kiagene Fanavar Company, who are based in our university, we finally decided to buy a MIC machine. I will send you the company link and email, maybe they can help you
  • asked a question related to DNA
Question
12 answers
I am not being unable to isolate DNA from J2 stage nematodes. The nematodes have been extracted from gall and currently are in water. I used extraction methods using sonnicator, glass beads, mortar pestle using liquid nitrogen and even qiagen blood and tissue DNA isolation kit, but no luck so far. I must be doing something wrong somewhere. I will be highly obliged if someone can share a working protocol for DNA isolation from juvenile nematodes or some tips how I can achieve this. Thanks
Relevant answer
Answer
Hi, you could use nanodrop
  • asked a question related to DNA
Question
8 answers
I am trying to convert a published sequence of mitochodrial DNA from the PDF file to fasta format in order to use it for primers design.
Relevant answer
Answer
good question
  • asked a question related to DNA
Question
5 answers
A number of samples were analyzed for isolated colonies of bacteria. However, in more than one sample, Total score was identical between several strains.
How can I choose the right strain?
Relevant answer
Answer
Thanks alot for your replies. I will try to use these options surely.
  • asked a question related to DNA
Question
8 answers
Hello,
I am working on a project where I need to recover RNA and DNA from the retina and RPE of mice (later I will likely test in rabbits and NHP's). I have been trying to find published data on the average level of nucleic acid recovery from these tissues in order to determine how efficient/inefficient my current approach is.
So far I have not found a publication listing such figures. Does anyone have personal experience or know of such a resource?
Thanks!
Relevant answer
Answer
Hey Salvador,
Its been awhile since I worked with mouse retina RNA extraction and I'm locked out of my old lab protocol I developed. From my memory I used a 250 uL proteinase K digestion for ~10 minutes on frozen tissues, afterwhich I added 750uL of TRIzol LS and further dissociated with bead beating. I followed the standard TRIzol extraction protocol since I found I obtained better total RNA yields. I used an overnight incubation at -80 during the precipitation (which could have just yielded more ribosomal RNA) but I was just doing rt-qPCR. I don't have any published protocol since I left that lab prior to any publications on the approach and no one picked up the project. As I said, this is from my memory of ~6 years ago so you'll definitely have to troubleshoot some. That said, at the time I found that yields by conventional TRIzol extractions yielded better extractions than column based on total recovery, but that could easily be skewed by ribosomal RNA.
Sorry I can't provide you with a more refined protocol to follow!
  • asked a question related to DNA
Question
5 answers
Is it possible that a monoclonal cell line expressing gfp in a stable way can lose or decrease its expression in the course of the passaging (example: after 20 passaging).
Because of for example a methylation of the promoter etc.?
Relevant answer
Answer
Hi Thomas,
I think this is possible and in fact I had a similar situation with HEK cells expressing GFP under a CMV promoter. After acoulpe of passages one could see the GFP signal was getting dimmer and it worsen with susesive passages. A similar line with EF1a as driver didn't have this issue. I was told a that time that it was either the promoter was getting metilated and inactivated overtime or that alternatively a regulatory element in the locus where the transgen got inserteg was getting modified and shutting down the expression of neighbouring genes.
Maybe you could check for methylation in your line within the area holding the GFP cassette.
Share if you find out.
Cheers
  • asked a question related to DNA
Question
14 answers
In human pregnancy, the constant turnover of placental trophoblast results in extrusion of apoptotic material into the maternal circulation. This includes cell-free (cf) DNA commonly referred to as "fetal", but is actually derived from the placenta. But are these DNA sequences as strongly correlated to preeclampsia as PlGF, sFlt-1, PAPP-A and other markers?
Relevant answer
Answer
There is strong genetic relation to this disorder. The STOX1 gene, the ERAP1 and 2 genes, the syncytin envelope gene, and the −670 Fas receptor polymorphisms are involved in the development of preeclampsia. The ACVR2A gene on chromosome 2q22 is also implicated.
  • asked a question related to DNA
Question
4 answers
After adding resuspension buffer and lysis buffer to the cell pellet, the next step is the addition of neutralisation buffer which requires immediate mixing. The expected outcome is the formation of large, fluffy and white precipitate. Slight delay in mixing results in the formation of small floating precipitate which remains suspended despite doubling of centrifugation duration, resulting in the decrease of A260:A280 eventually.
What are the remedies to solve this issue and improve the DNA purity? The DNA is plasmid DNA. Thank you.
Relevant answer
Answer
Honestly, it might be faster to just start over and follow the instructions. I'm sure you have more overnight culture than you needed.
  • asked a question related to DNA
Question
1 answer
Nat Protoc 16, 86–130 (2021).
Table 2 of this article mentioned that DNA nano barrel structure is "Difficult to increase lumen diameter beyond ~3 nm". I do not really understand the reason for this phenomenon.
Relevant answer
Answer
Dear jiahoa
I recumend this document for you
  • asked a question related to DNA
Question
3 answers
I am conducting a research using CRISPR/cas9 (gene knockout)
I am using PDR274 (#42250, addgene) to clone gRNA, transformation, in vitro transcription and pT3TS-nCas9n (#46757 , addgene) in vito transcription
After the isolation and purification of the plasmids the concentrations were quite low
[PDR274] 50 - 170ng/ul (purity 1.7 ~1.82)
[pT3TS-nCas9n] 30 - 100ng/ul ( purity 1.7 ~ 1.87)
Which is not enough for the following step
I used QIagen spin miniprep kit for purification
Any tips to increase the concentration up to 200 - 500 ng/ul
Thank you very much
Relevant answer
Answer
The normal way to concentrate DNA like plasmids would be just to do an ethanol precipitation and then resuspend in a smaller volume. Although since you are really only 2-3 fold below your target concentration you might just be able to add more DNA to the next steps and proportionally reduce any water in the reaction volume to keep overall volume similar.
  • asked a question related to DNA
Question
5 answers
Can anyone help me... I'm trying to purify DNA from whole blood w/ magnetic beads but yield and ratio 260/280 260/230 are very low.
My lysis/binding buffer is made with 4M thiocyanate guanidine, 50mM Tris-HCl, 2% Triton X-100 and 20 mM EDTA. Is there a difference about using SDS, Triton and Tween?
Relevant answer
Answer
you may also want to use Sodium deoxycholate to puncture the nuclear membrane for your lysis buffer. use at 1% w/v
  • asked a question related to DNA
Question
5 answers
Hey,
I have a question regarding Host-DNA-depletion and microbiome enrichment methods. To reduce the amout of Human-DNA in sulcular fluid samples from periodontitis patients I am looking for a microbiome enrichment method. The reason is that Host-DNA contanmination can overwhelm low biomass of microbial signals in Next-Generation-Sequencing.
I know that there are different kits for pre-extraction like MolYsis complete 5 kit, Molzym Ultra-Deep Microbiome Prep, QIAamp DNA Microbiome kit and Zymo HostZERO microbial DNA kit, but I am looking for a kit for Post-extraction. I only know one Post-extraction kit and that is NEBNext Microbiome DNA Enrichment kit, but do you know any other kits that work like the NEBNext kit?
I want to seperate the host-DNA to have my bacterial DNA that I can use for the library prep. So now I want to compair different kits for post-DNA extraction for host -DNA depletion and then check my results with qPCR and NGS, to make sure that the amout of human-DNA-reads are reduced.
The reason why I can‘t use a kit like MolYsis complete 5 kit or Zymo HostZERO microbial DNA kit (pre-extraction) is, because our company bought a beckman i5 workstation and also chemicals for the extraction (GenFind V3) that we have to use. So there is no way to change the DNA-extraction method. I have the extracted DNA, human and microbacterial DNA, and afterwards I want to seperate human and microbaterial DNA. So does somebody know a DNA-enrichment kit for microbiome enrichment after DNA-extraction?
Thank everbody for the help. Have a nice day!
Relevant answer
Answer
Hi Stephan,
Thank you! We are testing saponins as well, however we have solid tissue samples, not liquid, so I am not convinced about its effectivity for our purposes. Maybe some pre-treatment steps for tissue lysis would help. In addition, we also have total DNA extracted already which I wanted to use.
  • asked a question related to DNA
Question
16 answers
I am extractioning DNA from blood but my concentrations is low (about 20 ng/ul)..... can you help me? please...
my peotocol is:
1) add 500 ul lysyse buffer and 200 ul blood to a microtube.
2) 20s vortex and 10 min incubation in room temp and then centrifuge (1min/8000rpm).
3) transfer to new microtube.
4) add 100 ul of NaCl and 20s vortex.
5) add 500 ul isopropanol.
6) mix slowly and incubation 10 min in freezer.
7) mix slowly again and transfer to filtration columns and incibation 2 min in room temp.
8) centrifuge (40s/ 10000 rpm)
9) add 500 ul wash buffer and centrifuge (40s/ 10000 rpm)
10) transfer column to new microtube and add 40ul DNase free water pre_heated (60 c).
11) incubation 1 min and centrifuge (1 min/ 10000 rpm)
Relevant answer
Answer
Dear Amin Morshedi .The causes are: poor culturing conditions and plasmid propagation, excessive amounts of starting material resulting in insufficient bacterial cell lysis and column overloading. See the link: https://www.qiagen.com/us/resources/faq?id=82d92bd4-125f-4f54-88d1-d90fbe0fb090&lang=en
  • asked a question related to DNA
Question
4 answers
Component: Pure water, Bsm buffer, Bsm polymerase, primer, MgCl2, dNTP, DNA Salmonella
Heat 60 Celcius 1h and 80 Celcius inactivation 10min
Sometimes the results from gel electrophoresis appeared DNA bands but sometimes less band intensity. What happen? Not suitable primer or expired chemical?
Relevant answer
Answer
@Chayanit Thairat, Thanks for the protocol. I wanted to know if your are following a published protocol, then you may be able to compare your results with the reported observations. If you are optimizing your protocol, some parameters may have to be adjusted to get the desired gel profile
  • asked a question related to DNA
Question
6 answers
When run a LAMP reaction, sometimes false positive appeared in agarose gel electrophoresis. What are factors to create contaminations to false positives? What contaminations do in LAMP?
Relevant answer
Answer
Are you sure that you did not have any smpling problem such as insufficient amount of primer or different sources of genome! If not, you might optimiza the amount of enzyme, temperature and Incubation time. Also, i highly reccommend you in order to observe better amplification in LAMP assay do not forget using betain and MgCl2, I used it, it works😊
  • asked a question related to DNA
Question
11 answers
Hi,
I am trying to assemble a gene into a plasmid under a new promoter, is it possible to add the promoter sequence to the Forward primer to insert it? Or would I have to get a synthetic DNA, PCR it up and then insert it as a new fragment via Gibson?
Thanks
Relevant answer
Answer
Yes but you’ve to check with snapgene the freeware will tell you if the insertion will be in frame or not, alternatively if this not possible, having a good budget in mwg site you can order your gene optimized for e. Coli, adding your promoter immediately and then with the adaptors for the MCS site
  • asked a question related to DNA
Question
7 answers
Yesterday I have read a news stating that The embryo fossil, nicknamed “Baby Yingliang,” was discovered in Ganzhou, Jiangxi Province in southern China, and is believed to be at least 66 million years old. Researcher Dr. Fion Waisum Ma told the AFP news agency that the discovery is “the best dinosaur embryo ever found in history” (globalnews, 2021).
Although there were several discoveries of Dinosaur components such as:
Eggs
DNAs from thier remains
are frequently being discovered, Since the biotechnology development is in its Zenith at 2022, Why nobody has attempted to create a dinosaur?
What type of scientific constraints would be encountered in such a laboratory experiment?
Relevant answer
Answer
The science fiction book Jurassic Park and the movies based on it are about recreating dinosaurs by extracting their DNA from the guts of dinosaur-biting insects trapped in amber.
DNA is not sufficiently stable to survive for the necessary 65 million years or longer since dinosaurs roamed the Earth, so dinosaur DNA is not available. What you would do with it, if you could get it, in order to recreate dinosaurs is another issue.
The oldest DNA ever recovered was recently reported from 1.2 million year old mammoth teeth in Siberia.
It has been seriously proposed to recreate mammoths, which went extinct several thousand years ago. Mammoth DNA is available from animals preserved in permafrost.
  • asked a question related to DNA
Question
2 answers
Hello,
I am looking for a lab that can process insect DNA samples for me. More precisely, I would like to do hyRAD on my samples (they are not good enough for ddRAD). I am struggling to find one.
Thank you!
Sophie
Relevant answer
Answer
Tyler Chafin thank you very much !
  • asked a question related to DNA
Question
5 answers
Hi., I have been using phenol chloroform extraction protocol for DNA extraction but the solution is turning milky immediately after absolute ethanol precipitation and a low concentration and shearing is observed after running it in the gel. I am not using any vortex in order to avoid shearing , I have checked and changed the extraction buffer too, and have been carefully handling the procedure.
Protocol:(PDF) A modified efficient protocol for DNA extraction in Silkworm (Bombyx mori L.) (researchgate.net)
I am clueless at this point, kindly help me.
Thank You.
Relevant answer
Answer
As it's fat tissues, the ethanol can give cloudy/ milky appearance due to the lipids.
  • asked a question related to DNA
Question
3 answers
Some drugs/ carcinogen cause mutation in DNA, Which carcinogen causes highest mutation in genome?
Relevant answer
Answer
Hi Amrit,
The most commonly mutated gene in people with cancer is p53 or TP53. More than 50% of cancers involve a missing or damaged p53 gene. Most p53 gene mutations are acquired. Among most active carcinogenic mutagens the championship title belongs to polycyclic aromatic hydrocarbons (PAHs). These are the most abundant indirect-acting carcinogens to which humans are exposed to on a daily basis. Exposure has been associated with the development of breast, skin or lung cancer. Bioactivation of PAHs is required in order for these agents to exhibit mutagenic properties, which is primarily mediated by cytochrome P450 enzymes (among many others, our data on this subject - in http://www.xenobiovir.com/). Bioactivated metabolites target multiple genomic sites, including guanine and adenine bases via PAH diol epoxides. This results in the generation of bulky BPdG chemical DNA adducts; examples include quinone-mediated cross-linking of N7 position of guanine and N3 of adenine.
All the best,
Ilya
  • asked a question related to DNA
Question
6 answers
How does circular DNA benefit Epstein-Barr virus (EBV)?
Relevant answer
Answer
EBV circular DNA benefit establishing latent infection and long _term persistance for viral genome.the also the circular DNA infected cells may well be ignored by the immune system.the attached ref.illustrate all the request:
Annual Rview of Virology.Vol.3:359_372
Epsten_Barr virus is another herpes simplex 1
  • asked a question related to DNA
Question
6 answers
Hi all,
I have a six BP oligo consisting of GGTGGT (glycine - glycine) that I need to add a a restriction site to for an experiment we're carrying out using mutant restriction nucleases. I was going to design a primer that bound to these residues so I could add the desired sequence but the Tm is only ~20 degrees. Any recommendations would be very welcome.
Thanks in advance
John
Relevant answer
Answer
Assuming that the GTTGTT and then the restriction site are added to a longer specific oligo then you shoud check how many additional bases you need to add as well as the restriction site because many enzymes need up to 3 extra bases to bind to dna in the cutting process. New england biolabs publish a spreadsheet of many common enzymes and their need for extra bases