Science topic

Molecular Biology - Science topic

PCR, Cloning, Restriction Digestion, Ligation, Transformation, Plasmid et al
Questions related to Molecular Biology
  • asked a question related to Molecular Biology
Question
3 answers
I am analyzing the differential expression of splicing variants, through capillary electrophoresis, of a DNA repair gene after treatment with a DNA-damaging drug. I am using gene-specific primers for cDNA synthesis, which makes it challenging to identify a suitable internal control. Using another region of the same gene might also be affected by the drug, while choosing a housekeeping gene would require a separate cDNA synthesis step and introduce variability. What would be the most appropriate internal control to ensure accurate normalization in this setup?
Relevant answer
Answer
Why are you using capillary gel electrophoresis instead of qPCR? The detection limits of capillary tubes is very limited - easy to saturate & minimal signal for detection is also high.
You can use a 1-step qPCR for your splice variants and 1-step for a housekeeping gene.
  • asked a question related to Molecular Biology
Question
2 answers
Hi,
I'm trying to differentiate iPSCs to Immune cells and looking for plates with low-attachment. I came across this reagent "Anti-Adherence Rinsing Solution" by stemcell technologies which is a surfactant solution for pre-treating cultureware to reduce surface tension and prevent cell adhesion.
But, my question is: can I use this solution on any TC treated plates or should it be only from aggrewell brand? Has anyone used it before?
Thanks.
Vertica
Relevant answer
Answer
Hi Vertica,
My name is Audrey and I am a Product Manager at STEMCELL Technologies.
The Anti-Adherence Rinsing Solution is effective when applied to any plate type, not just AggreWell™ plates. However, its performance is not optimal on tissue culture-treated plates and you may see failures after a couple of days. 
Our Product and Scientific Support Team would be happy to work with you directly and troubleshoot this issueyou can email them at techsupport@stemcell.com.
I hope this helps!
Kind regards,
Audrey
  • asked a question related to Molecular Biology
Question
7 answers
I have been using NEB Hifi Gibson assembly for a couple years now and I've been quite happy with it. I regularly make plasmid constructs with 4-8 fragments, and always >1/4 of the colonies are "perfect," while the remaining ones may have some SNPs at the joining sites, or be misassembled due to a repetitive region.
Some colleagues said that Golden Gate has even higher efficiency. That even with 8 fragments, it is normal for 50-100% of colonies to be perfect. Is that true? Is Golden Gate that perfect?
Relevant answer
Answer
Has anyone tried GenScript's GenBuilder DNA assembly kits?
  • asked a question related to Molecular Biology
Question
26 answers
I am using rosetta for protein expression and my recombinant protein is T7 promotor base. I used more than 3 protocols and different temperatures (16,30,37) and IPTG 1.0 mM and I also used 3% ethanol but I am not getting sds page or western but I am getting positive result in dot blot,but in dot blot my control also showing reaction also. Anyone can help me?
Relevant answer
Answer
Hi Can,
Howard Salis, 1st author of the manuscript I had linked in my prior response to the question 8 years ago, was at UCSF at the time his manuscript was published. The manuscript link to the prediction program is dead but he put it on a new server when he moved to Penn State.
I was using a prokaryotic GateWay Destination expression plasmid. The donor plasmid in which I had cloned the gene was primarily designed for recombining into eukaryotic expression vectors. Therefore, I added a prokaryotic Ribosome Binding Site (RBS or Shine-Dalgarno sequence) to the forward oligo to PCR and clone the gene I was expressing. Cloning was very straightforward (I actually had a high school student do it as a summer project). BUT it took almost 2 years to get expression (when I found Salis' program's new site). It correctly predicted that when transcribed, the RBS in the 5' leader sequence would form a very strong hairpin with an upstream NotI restriction site and flanking sequence (so lots of GCs in a row) in the original Donor plasmid. The hairpin sequestered the RBS, preventing the ribosome from assembling on it so translation was squelched. Eukaryotic ribosomes assemble at the mRNA cap and just plow their way to the Kozak site, secondary structure be damned. Prokaryotic ribosomes instead assemble on the mRNA on the RBS located directly before the initiating methionine, but only if the site is accessible.
Salis' program analyzed the inputted sequence and predicts expression levels but doesn't advise how to improve it. I used a separate program that maps RNA structure (I don't remember its name but there are certainly better programs now), and used intuition to manually tweak the sequence to eliminate secondary structure, and then tested it with Salis' program. I kept reiterating this process many times until I thought I had absolutely maxed predicted expression. I then repeated this for the initial 6xHis tag at the N-terminus because it too was predicted to form a very strong hairpin - that involved just changing some of the CAC codons to CAU, so pretty easy. I'm not sure if the His hairpin affected translation but as all the changes were incorporated into a single oligo, there was really no additional work.
The first time I tried the optimized vector, I got rid of all the desperation techniques I had tried - tight repression of toxic expressed proteins, medium formulations, temperature, inoculation protocols, fancy bugs. Also exotic purification techniques, protease inhibitors, and lysate formulations. Absolutely everything. Just 1 L of barebones LB on a shaker overnight and a simple lysate poured over a nickel column. Ended up with hundreds of milligrams of almost pure protein. I had *no* evidence of expression prior. So much protein that it overwhelmed the endogenous E. coli biotinylation system (I had included a C-tag for biotinylating the protein) so I had to add a 3rd plasmid to the system to express additional biotinylating enzyme (the 2nd plasmid expressed the 2 casein kinase subunits to phosphorylate the target expressed protein, so yeah, kinda complicated).
Quite long, but I hope this helps. I would certainly contact Howard Silas for advice - he's the expert. And I'm sure things have advanced significantly in the 8 years since this question was originally posted. https://www.researchgate.net/profile/Howard-Salis
  • asked a question related to Molecular Biology
Question
7 answers
Hello ResearchGate group,
As M.D.-Ph.D. scientist working on molecular biology, I am wondering if anyone is interested in joining efforts for creating PCR standardization statistical tools.
Best regards,
Alexios
Relevant answer
Answer
Othman Mueen Mohammed Many thanks for your insight and input; I deeply appreciate it! I can keep you posted on the project.
  • asked a question related to Molecular Biology
Question
1 answer
Is this toolkit only an extension for YTK (MoClo Yeast toolkit), does it assume that I have this kit and is based on YTK level 0 fragments?
Are there any regular YTK promoters or only the newly added inducible promoters?
Relevant answer
Answer
I think, newly added inducible promoter
  • asked a question related to Molecular Biology
Question
5 answers
molecular biology and molecular genetics laboratories. It is a reagent designed for the removal of ribonucleases (RNases) and other nucleic acid contaminants, such as RNA, that can degrade nucleic acid samples, such as DNA.
Relevant answer
Answer
From answers on the Internet, most of them reveal that RNAse Zap or other similar decon reagents use oxidants (e.g. Hydrogen Peroxide/H2O2 or Sodium Hypochlorite/NaClO(main ingredient of commercial bleach)) to inactivate RNAse.
Defeat RNAse Contamination Using Bleach in Your RNA Agarose Gel (bitesizebio.com)
10 Ways to Work RNase Free (bitesizebio.com)
Just bleach it – Pipette Jockey
  • asked a question related to Molecular Biology
Question
10 answers
In recent months, I've been running gels for a fragment around 430 bp on 1.2% agarose gel with a voltage of 120 and a 100 bp DNA ladder, and it has been working without any issues.
After a two-month break, I’ve returned to this routine, but now the runs under the same conditions are turning out as shown in the picture. Does anyone have suggestions on how to improve this? I don’t think it’s an issue with my samples, as even the DNA ladder itself is showing problems.
Relevant answer
Answer
It seems that your DNA samples and ladder are fine, but there might be a problem in the agarose gel preparation. Are you sure you have used 1X TAE buffer while preparing you gel and run the gel in 1X TAE buffer? I think the gel is made with water or run against water. TAE buffer provides ions that conduct electricity, which is essential for the DNA to migrate through the gel. This poor migration of DNA samples and and "smileys" are typical sign of low buffer capacity.
Best wishes.
  • asked a question related to Molecular Biology
Question
4 answers
the lab has minimum molecular biology setup like: DNA extraction, gel electrophoresis and thermal cyclers/
Relevant answer
Answer
Dear Dr. Suhail Ayesh
You may start with blood genomic DNA extraction by using a DNA blood kit followed by quantification using nanodrop and quality check by agarose gel electrophoresis.
This could be followed by a simple method namely, PCR-RFLP. In this method, known regions of DNA are amplified by PCR, and the PCR amplicons are then digested with restriction enzymes to produce distinct polymorphic fragments which are then subjected to electrophoresis. Ultimately, what you detect is nucleotide variation for a restriction enzyme recognition site. The variation must lie within the site.
So, PCR-RFLP employs four main steps:
(1) isolation of genetic material and PCR,
(2) restriction digestion of amplicons,
(3) electrophoresis of digested fragments, and
(4) visualization.
PCR-RFLP is a low-cost and low-throughput research method allowing for the analysis of SNPs in the absence of specialized equipment, and it is useful when there is limited budget.
Besides the above, there are many novel and reliable techniques used to assess gene polymorphisms such as Taqman assay, amplification refractory mutation system (PCR-ARMS), high-resolution melting and different types of mini-sequencing, which you could try, if you have enough funds.
Best Wishes,
Malcolm Nobre
  • asked a question related to Molecular Biology
Question
1 answer
Have a question on gene expression, protein synthesis, or molecular pathways? Post it here, and let’s explore molecular biology together!
Relevant answer
Answer
Myes, I am interested in genomic study of microbes
  • asked a question related to Molecular Biology
Question
6 answers
I ordered a primer when I received it where one nucleotide is different in the forward primer.
the original sequence is: CTCTTTGGGCTCAGAGTGAGTCTGG
the sequence which I get: CTCTTTGGGTTCAGTGTGAGTCTTG
Three nucleotides (Bold) are different
This primer will work?
while I have tried so many times but there is no result. If it is working, how can I fix my PCR protocol?
Relevant answer
Answer
Just buy a new primer! It's not worth your time or reagents to try and get a faulty primer to work. Primers are cheap.
  • asked a question related to Molecular Biology
Question
4 answers
Hi. I want to synthesize this blaTEM gene. This is the FASTA sequence, but I don't know which part is related to coding sequence, Can anyone help me?
(my major isn't related to microbiology or genetic, so I don't know the exact procedure of genetic basics,)
and one more thing, after synthesis how can I know how many copy numbers are there in my sample to generate qPCR standard curve based on that?
Relevant answer
Answer
The coding sequence start from the first "ATG" right after bacterial promoter sequence and end up at stop codon "TAA".
From 101 to 961.
  • asked a question related to Molecular Biology
Question
4 answers
Relevant answer
Answer
Science is making anti-aging progress. But do we want to live forever?
Scientists reprogram T cells to slow down and reverse aging
The Future Of Anti-Aging: Emerging Technologies And Trends ** ( Apr 12, 2024 )
Category : ANTI AGEING TREATMENTS
  • asked a question related to Molecular Biology
Question
23 answers
I have to estimate the growth of my bacterial cultures spectrophotometrically and I read articles of measurement at an OD of 600nm. Also what to do if the values exceed 1. What is the proper method for the measurement of the same.
Relevant answer
Answer
Different labs use different wavelengths to measure, usually from 590-650. They all work fine so it doesn't really matter so long as you consistently use the same. Traditionally 600nm has been used but not by everyone, I think because it matches the old color filters of Klett meters that used to be used.
When you get to optical densities above 1, the amount of light being scattered vastly exceeds the amount passing through, so the accuracy of the measurement goes way down. As the other responders said, the solution is to dilute your culture and take your reading so that it is below 1.
  • asked a question related to Molecular Biology
Question
3 answers
Exciting news! Users can now like projects on PeptiCloud. Check it out: www.pepticloud.com
Relevant answer
Answer
Yeah, it's absolutely right
  • asked a question related to Molecular Biology
Question
4 answers
I found that fingerprint analysis of oligonucleotide fragments was used in molecular biology in some top-tier journals in the 1970s, but it seems difficult to find such experimental methods in current journals. Why is that? Does this experimental approach have any defects? What methods can be used to replace it now?
Relevant answer
Answer
There are some challenging issues.
Need prompt expert in this field to reveal results, as there may be errors in indicating false results.
Contamination will collapse total protocol.
Environmental factors may change the originality of results
  • asked a question related to Molecular Biology
Question
2 answers
15 E. coli K12 promoters from EcoCyc are now accessible on PeptiCloud (www.pepticloud.com)! You can clone them into your projects to build constructs. Find them here: https://www.pepticloud.com/public-project/E.%20coli%20K12%20Promoters.
Relevant answer
Answer
Thanks for sharing
  • asked a question related to Molecular Biology
Question
7 answers
Dmt protection is a important step allowing protection of the hydroxyl group in the 5'OH in the nucleotides. This 5'OH specific protection allow a sequential coupling of the nucleotides, forming a obligonucleotide.
As we know DMT protection is follow the sn1 reaction manner. And for sn1 reaction, primary alcohol is less reactive than the secondary alcohol. Therefore, beside steric hindrance I would like to know why DMT is 5'specific. Thank you so much
  • asked a question related to Molecular Biology
Question
2 answers
Dear esteemed colleagues and professors,
I understand that this platform may not be the most appropriate place for this request, but as I was unsure of where to post this request where all respected professors and colleagues are present, I have decided to post it here.
I am a master's student in cellular and molecular biology and have been looking for a group or professor to collaborate with on writing a paper.
If you are willing and able to help, please send me an email at:
[ztaheri199@yahoo.com]
Thank you for your time and consideration.
Relevant answer
  • asked a question related to Molecular Biology
Question
4 answers
Greetings, recently I have acquired the pCDH-CMV-MCS-EF1-CopGFP-T2A-Puro vector (V012884). I would like to know whether the gene of interest's expression is regulated by the CMV promoter or not, given that this vector contains two sets of 5'LTRs. Specifically, I would like to know which set is integrated into the cell genome subsequent to transduction. Regards in advance,
Relevant answer
Answer
I also have the same question regarding the issue of two 5'LTR in this vector. Have you figured it out? If I want to construct a stable cell line by this vector, where should I put my gene of interest in? Between the first 5'LTR and 3'LTR or the second one and 3'LTR?
  • asked a question related to Molecular Biology
Question
12 answers
For researchers in molecular biology and genetic engineering, I would like to know the most successful genome sequencing technique(s) currently used worldwide?Thanks in advance.
Relevant answer
Answer
Dear Dr. Fetta Mehouel
I feel Next-generation sequencing (NGS) is currently most popular. The global next-generation sequencing market size was valued at USD 8.30 billion in 2023. The market is projected to grow from USD 9.29 billion in 2024 to USD 27.55 billion by 2032. Please refer to the article attached below.
It all started with the first-generation technology represented by Sanger sequencing, which was largely superseded by newer, higher-throughput sequencing technologies. The second generation introduced massively parallel sequencing with platforms such as Illumina and Ion Torrent, enabling high-throughput sequencing. Among the second-generation sequencing platforms,
1) Roche’s 454 sequencing method, which relies on pyrosequencing, where the sequence is determined by detecting the release of pyrophosphate when nucleotides are added to the DNA template, and
2) Illumina sequencing platform which utilizes a sequencing-by-synthesis method based on reversible dye terminators.
have emerged as the widely used second-generation sequencing platforms.
Third-generation sequencing technologies represent the latest advancements in DNA sequencing, offering new approaches that overcome the limitations of previous generations.
1) PacBio Sequencing, which uses a single-molecule, real-time (SMRT) approach with fluorescently labeled nucleotides, enabling long-read sequencing of DNA fragments up to tens of kilobases in length and
2) Oxford Nanopore sequencing, based on nanopore technology, where a single-stranded DNA molecule passes through a nanopore, and changes in electrical current are measured to determine the DNA sequence.
You could use Sanger sequencing when interrogating a small region of DNA on a limited number of samples or genomic targets. But NGS will allow you to screen more samples cost-effectively and detect multiple variants across targeted areas of the genome, which otherwise would be costly and time-consuming using Sanger sequencing.
Regards,
Malcolm Nobre
  • asked a question related to Molecular Biology
Question
4 answers
Good evening, can you help me interpret the results of the 1% agarose gel.
DNA was extracted from plant tissue using the CTAB protocol. In the wells (left to right) in well 1 and 4 is the molecular weight marker (1kb) with the amount of 1μl, well 2 has 1μl of sample + 1μl of loading buffer. well 3 μl of sample + 1μl of loading buffer.
Electrophoresis conditions: 80V x 30 min.
I am mainly interested in understanding the banding pattern of well 2.
Thank you.
Relevant answer
Answer
Most folks don't bother to run out gDNA on an agarose gel since it's not informative unless you are doing a very specific protocol like long-read DNA sequencing where you need to verify intact, large DNA molecules. If needed, measure the concentration in a spectrophotometer (aka nano drop) or Qbit.
Typically, you just proceed to the next step in the protocol.
Good luck!
  • asked a question related to Molecular Biology
Question
18 answers
In the identified case of familial desminopathy (T341P DES mutation in heterozygous state), the son has bradycardia, but the father did not have bradycardia. How can this fact be explained?
Relevant answer
Answer
Because of some autosomal dominant & others can be autosomal recessive
"Desminopathy is one of the most common intermediate filament human disorders associated with mutations in closely interacting proteins, desmin and alphaB-crystallin. The inheritance pattern in familial desminopathy is characterized as autosomal dominant or autosomal recessive, but many cases have no family history."
  • asked a question related to Molecular Biology
Question
2 answers
Hi all,
DNA sequences have traditionally been shared through literature, often scattered or hidden in appendices, making it difficult for the research community to access and use modular sequences.
To enhance collaboration and accessibility, PeptiCloud has introduced a new feature that allows researchers to effortlessly clone and import DNA sequences from other projects into their own. Inspired by open-source collaboration in the software industry, this feature aims to make it easy and error-free for researchers to utilize and build upon sequences created by others.
Check out the cloning feature at www.pepticloud.com!
Thank you,
Chris
Relevant answer
Answer
Hi Dr. Akter. It's completely free!
  • asked a question related to Molecular Biology
Question
5 answers
I am currently optimizing a RPA-based protocol. As everyone knows, this nucleic acid amplification technique is based on isothermal amplification (around 37-42˚C) in combination with recombinases and single-stranded DNA binding (SSB) proteins.
I have designed several candidate primers to optimize my RPA. All of them were searched in literature and designed considering the criteria to be used in an RPA reaction.
These same primers were verified by conventional PCR (At different Tm: 55-65ºC), obtaining a successful result.
The problem started when I used the RPA master mix (lyo version from Twistdx) and performed the RPA reaction with the same primers and samples used in the conventional PCR. ALL THE RESULTS BECAME NEGATIVE?!
Primer concentrations used in RPA reaction was 400nm (recommended by twistdx) and the reaction time was 30 minutes at 39 °C (conditions recommended for the set of primers tested). Visualization of the amplification was done on agarose gel (1.5%) and doing a posterior melting curve assay. In all cases, no amplification was detected.
Does any one have a clue on what is happening???
I don´t know which other variables I can change to obtain good results
Suggestions?
Thank you
Relevant answer
Answer
We manage to make it work with another kit! The problem emerges because we bought the exo kit by mistake which amplifies and degrades at the same time to produce signal with the prove. Thank you all for the replies. I am no longer working on that projeto neither on the company developing the method
  • asked a question related to Molecular Biology
Question
3 answers
Help me to find experienced supervisor with excellent lab studying plant abiotic stress physiology and molecular biology
Relevant answer
Answer
CCNU
  • asked a question related to Molecular Biology
Question
10 answers
When we perform statistical analysis on qPCR data, do we use fold change or ΔCt?
Relevant answer
Answer
you mean -ΔΔCt?you could use either -ΔΔCt or the fold change ( 2 to the power of -ΔΔCt).
  • asked a question related to Molecular Biology
Question
3 answers
I am reaching out to seek your expertise on the use of different culture media for growing gene-transformed Agrobacterium K599.
In my current work, I am utilizing both TY and YEP media to culture Agrobacterium rhizogenes K599 that has been transfered DNA plasmid pAGM4723 contain RUBY reporter gene. However, I am unsure how these media might affect the growth and gene expression in this particular bacterial strain.
1. Are there any known effects of using TY medium versus YEP medium on the expression of the introduced genes in Agrobacterium K599? How might the choice of medium influence gene stability or expression levels?
2. Are there specific experimental conditions or practical considerations that might make one medium preferable over the other for optimal culturing of gene-transformed Agrobacterium K599?
Relevant answer
Answer
Differences:
Nutrient Content: YEP is richer, promoting higher bacterial density.
Growth Rate: TY supports faster growth, while YEP yields more biomass.
Application: TY for quick, routine growth; YEP for robust growth and higher biomass.
In summary, choose TY for faster growth and YEP for larger biomass, depending on your experimental needs.
  • asked a question related to Molecular Biology
Question
5 answers
I was wondering if it is possible to form a permanent open "ssDNA bubble" similar to a transcription bubble (>13 nucleotides) within E. coli. These criteria are important:
1. Open ssDNA bubble within replicable (in E. coli) genetic element. So no C-Traps under force.
2. No proteins, nucleic acids, or other toxic chemicals supporting the bubble. Can help during nucleation, but bubble has to be accessible for protein interaction.
3. Stable in bioorthogonal conditions. Physiological pH, salt, 37 °C, etc.
Relevant answer
Answer
Well, creating a semi-permeable transcription bubble can be challenging in the context of the structural stability of DNA as it tends to reanneal to its double-stranded form. Next is the concern of replication, which involves the fact that semi-permanent unwinding can potentially hinder the replication machinery from proceeding with DNA replication. Lastly, to maintain the transcription bubble to its semi-permanent unwound state, RNA polymerase is required to be halted in its activity at the transcription site, which could, in turn, lead to instability and interference in the replication of the plasmid. Considering these aspects, I believe three probable yet theoretical strategies can be adopted in this regard. First is genetically engineering a modified RNA polymerase, which can maintain the plasmid DNA at its single-stranded state by getting associated at a precise plasmid location without hindering the transcription process. Second is implementing genetically engineered single-strand binding (SSB) proteins, which can keep the plasmid DNA at its unwound state without interfering with RNA synthesis. Lastly, chemical molecules such as intercalating agents are introduced, which can develop proximal unwinding by being inserted at the nitrogenous base pairs of plasmid DNA; Molecules that are enhancers or activators of helicases; Hydrogen bond destabilizers like Di-Methyl Sulfoxide (DMSO), Urea or Formamide which can perform denaturation of double-stranded DNA; Cross-linking agents like Psoralens which forms covalent cross-linkages between single-stranded DNA molecules and DNA or RNA polymerases; Ligands which associate with single-stranded DNA such as Peptide Nucleic Acids (PNAs) and nucleic analogs which stabilizes the single-stranded structures of DNA; Alkylating agents such as Nitrogen and Sulfur derivatives of Mustard gas, Ethyl Methanesulfonate (EMS), Methyl Methanesulfonate (MMS), N-Nitrosoureas and Temozolomide. Nevertheless, besides being hypothetical, all these strategies have cons of their own.
  • asked a question related to Molecular Biology
Question
2 answers
Hi all,
Editing DNA sequence has been done traditionally in a word doc or google sheets, which often leads to mistakes and limits the ability to create combinations using various regulatory elements in one location (ex: trying out different promoters or signal sequences for a certain ORF).
To solve such issues, PeptiCloud has created a brand new feature called Sequence Playground. Inspired by visual programming or block-based coding, Sequence Playground treats each DNA segment as a block and allows users to mix and match various blocks to combine multiple sequences in an efficient manner. It also allows users to deconstruct a sequence into smaller segments to be used as components.
Please check out the new feature at www.pepticloud.com!
Thank you,
Chris
Relevant answer
Answer
Husni Mubarok Yes, it's completely free!
  • asked a question related to Molecular Biology
Question
7 answers
Hi,
I'm transducing Ly1 and L363 cell lines using our standard protocol for retroviral transduction. The cells are successfully transduced as evidenced by GFP expression. However, after 4-5 days they start dying off and look really stressed. I'm suspecting polybrene since we've got a new batch. The cells look really weird, irregular and start forming clumps which they don't normally do in standard cell culture. I've tried using the same polybrene concentration (8ug/ml) in standard culture medium without the virus to check toxicity and it appears that it is decreased. Which concentrations do you normally use? Should I make a polybrene concentration curve to find the minimal nontoxic condition?
Relevant answer
Answer
Dear Colleague,
I hope this message finds you well. Polybrene (hexadimethrine bromide) is commonly used to enhance the efficiency of viral transduction, particularly with retroviruses and lentiviruses, by reducing the charge repulsion between the virus and the cell surface. However, polybrene toxicity can sometimes be an issue, manifesting as reduced cell viability or altered cell behavior. Here are some insights and troubleshooting tips to address polybrene toxicity during viral transduction:
Understanding Polybrene Toxicity
  1. Mechanism of Toxicity:Cell Membrane Interaction: Polybrene interacts with the cell membrane to facilitate viral entry. At higher concentrations, this interaction can disrupt membrane integrity, leading to cell toxicity. Cell Type Sensitivity: Different cell types have varying sensitivities to polybrene. Some cells tolerate higher concentrations, while others are more susceptible to its toxic effects.
Troubleshooting and Mitigating Polybrene Toxicity
  1. Optimizing Polybrene Concentration:Concentration Range: Polybrene is typically used at concentrations ranging from 2 to 10 µg/mL. Start with a lower concentration (e.g., 2 µg/mL) and gradually increase it while monitoring cell viability. Cell Type Specificity: Determine the optimal concentration for your specific cell type. Perform a titration assay to find the highest concentration that enhances transduction efficiency without causing significant toxicity.
  2. Shortening Exposure Time:Reduced Exposure: Limit the exposure time of cells to polybrene. Shorter incubation periods (e.g., 2-4 hours) can reduce toxicity while still enhancing transduction efficiency. Wash Steps: After the desired exposure period, wash the cells thoroughly to remove any residual polybrene, replacing it with fresh culture medium.
  3. Using Alternatives or Supplements:Poloxamer 407 (Pluronic F-68): This non-ionic surfactant can be used as an alternative to polybrene to enhance viral transduction with reduced toxicity. Serum Supplementation: Ensure that your culture medium contains serum (e.g., 10% fetal bovine serum), which can help mitigate the toxic effects of polybrene by providing protective factors.
  4. Monitoring and Validating:Cell Viability Assays: Use viability assays (e.g., MTT, CellTiter-Glo) to quantitatively assess the impact of polybrene on cell health. This helps in optimizing the concentration and exposure time. Transduction Efficiency: Evaluate the efficiency of viral transduction by measuring the expression of the reporter gene or transgene (e.g., GFP, luciferase) to ensure that reduced polybrene toxicity does not compromise transduction efficacy.
Example Protocol for Polybrene Optimization
  1. Cell Seeding: Seed your cells in a 24-well plate at a density that allows them to reach 70-80% confluency the next day.
  2. Polybrene Titration: Prepare a series of polybrene concentrations (e.g., 0, 2, 4, 6, 8, 10 µg/mL) in your viral supernatant or culture medium.
  3. Viral Transduction:Add the viral supernatant containing the different concentrations of polybrene to the cells. Incubate the cells for 2-4 hours at 37°C. After incubation, replace the medium with fresh culture medium without polybrene.
  4. Assessment:Cell Viability: Assess cell viability 24-48 hours post-transduction using a viability assay. Transduction Efficiency: Measure the expression of the transgene 48-72 hours post-transduction to determine the optimal polybrene concentration.
By carefully optimizing the polybrene concentration and exposure time, you can enhance viral transduction efficiency while minimizing toxicity to your cells.
Should you have any further questions or require additional assistance, please feel free to reach out.
Reviewing the protocols listed here may offer further guidance in addressing this issue
  • asked a question related to Molecular Biology
Question
4 answers
I have used ssDNA oligo and plasmids for HDR. I am wondering whether anyone has tried using PCR product directly as HDR template. 
Relevant answer
Answer
you can use a PCR product as a CRISPR HDR (Homology-Directed Repair) donor, but there are several considerations to ensure its effectiveness:
**1. Quality of the PCR Product
**1.1 Purity:
Ensure Purity: The PCR product should be pure and free from contaminants, such as residual primers, enzymes, and other by-products. Use a purification step like gel extraction or commercial PCR clean-up kits.
**1.2 Length and Integrity:
Check Integrity: Verify that the PCR product is of the correct size and does not have unintended bands or degradation products. This can be assessed by running an aliquot on an agarose gel.
**2. Design Considerations for the HDR Donor
**2.1 Homology Arms:
Include Homology Arms: The PCR product should contain homology arms flanking the desired insertion or modification site. The homology arms should be sufficiently long (typically 500-1000 bp) to promote efficient HDR.
**2.2 Donor Sequence:
Incorporate the Desired Sequence: Ensure that the sequence you want to insert or modify is correctly included in the PCR product. Verify the sequence by sequencing if necessary.
**2.3 Avoiding Repeats:
Minimize Repeats: Avoid long repeats or sequences that might cause unwanted recombination or instability.
**3. Transformation and Transfection
**3.1 Transformation Efficiency:
Assess Efficiency: If using bacteria, check the efficiency of transformation. High-quality DNA is essential for efficient transformation or transfection.
**3.2 Transfection:
Optimize Conditions: When using the PCR product in mammalian cells, optimize transfection conditions to ensure efficient delivery of the donor DNA. Methods include lipofection, electroporation, or viral delivery systems.
**4. Validation
**4.1 Confirm Insertion:
Validation: After introducing the HDR donor into the cells, validate successful insertion or modification by PCR, sequencing, or other molecular assays.
**4.2 Screening:
Screen for Correct Integration: Use appropriate screening methods to confirm that the HDR process occurred correctly and that the donor DNA has been properly integrated.
**5. Considerations for Using PCR Products
**5.1 Length Limitations:
Product Length: PCR products are typically short (e.g., 200-1000 bp). For larger insertions or modifications, consider using plasmid-based donors with larger homology arms.
**5.2 End Modification:
Blunt Ends vs. Overhangs: Ensure that the ends of the PCR product are compatible with the CRISPR-generated DSB (Double-Strand Break) ends. Depending on the CRISPR system used, you might need to modify the ends (e.g., to be blunt or to have overhangs).
**5.3 PCR Artifacts:
Check for Artifacts: Ensure that the PCR product does not contain artifacts such as nonspecific bands or residual primers that could affect the HDR process.
Alternative Approaches
**1. Plasmid-Based Donors:
Consider Using Plasmids: For larger modifications or more complex constructs, using plasmid-based HDR donors with appropriate homology arms may be more effective.
**2. Commercial Donors:
Pre-made Donors: Commercially available HDR donor plasmids often come with optimized designs for efficient integration and can save time and effort.
In summary, while PCR products can be used as HDR donors, their effectiveness depends on proper design, purification, and delivery. For larger or more complex modifications, plasmid-based donors or commercially available options might be preferable.
l Reviewing the protocols listed here may offer further guidance in addressing this issue.
  • asked a question related to Molecular Biology
Question
3 answers
I am trying to extract DNA from serum. I found an article that claimed simple extraction can be done in a single tube -
Here they used a solution containing 6M NaI/13mM EDTA/0.5% sodium N-lauroylsarcosine/10 µg glycogen as a carrier/26mM Tris-HCl, pH 8.
But currently, I don't have NaI and glycogen. So, I am thinking of making a solution with KI + EDTA + sodium lauroyl sulfate + Tris-HCl, pH 8. And finally, use Na-acetate and absolute ethanol for the precipitation of DNA.
What consideration should I take into account to use alternative reagents?
And, in their protocol does it mean the final solution containing all reagent should have a pH 8 or it just means the use of Tris-HCL with pH 8?
Relevant answer
Answer
I think KI should work as Nal replacement, but it might behave slightly differently, so I would keep an eye on how it affects the results, I shall for sure check for any potassium salts deposits in the solution before proceeding with it. The replacement of sodium lauroyl sulfate should still work but just be mindful, that it can be a bit stronger or much aggressive for cell lysis or protein denaturing, so I would just adjust the concentration accordingly. Without any glycogen, the DNA quantity might be dropping but you should still be fine.
I shall use Tris-HCL at pH8, since the protocol specifies that the extraction solution is maintained at pH8, which is said to be adjusted by Tris-HCl at pH 8. They might be suggesting this pH throughout the process for better results.
If possible, just test the adjusted formulation on a small scale, with minimal serum sample to see how well it works and how good the DNA quantity would be.
  • asked a question related to Molecular Biology
Question
3 answers
Hello,
We found three packages of Illustra™ MicroSpin™ G-25 columns in the cabinet of an unused lab. They are very old but have never been opened. I have never used this kit before, and I couldn't fully understand what it is used for from my internet search. Is it just a simple DNA purification kit, or is it something more functional? I am interested in recombinant protein production. Can I purify my ligation product with this before cloning into bacteria, or can I purify my PCR product with this before ligation? In which scenarios is this kit indispensable?
Thank you.
Relevant answer
Answer
These are sephadex spin columns and work on the basis of size exclusion. So dna of more than 10 bases long will flow round the beads and elute early while small salts will have to flow through the pores of the beads and will elute much later. They can be used for dna purification from most smaller molecules and have been used to remove radioactive salts from end labelling of oligos with the labelled oligo eluting first off the column. The G25 is an indication of the size range that can be separated on these columns
  • asked a question related to Molecular Biology
Question
1 answer
Dear Scientists and Researchers,
Problem Statement: Managing multiple versions of biological sequences can be challenging, often leading to compromised data integrity and collaboration issues.
Introducing PeptiCloud's Version Control:
  • Create and Manage Versions: Start with an initial sequence and create multiple versions without losing the original or compromising on different visions.
  • Organize Changes: Implement changes and create new branches for different versions, keeping everything organized in one place.
  • Flexible Modifications: Add various elements like promoters, terminators, and tags as new branches to any version of the sequence or go back and edit previously created sequences.
  • Track History: Maintain a detailed history of all modifications, including who made changes and when.
  • Collaborative Projects: Share version trees with others, allowing collaborative viewing and editing. Public projects let non-members view version trees while restricting editing to added members.
Sign up for free at www.pepticloud.com and reach out at pepticloud@outlook.com with your thoughts, suggestions, or questions.
Thank you for your time!
Sincerely,
Chris Lee Bioinformatics Advocate & PeptiCloud Founder
Relevant answer
Answer
Thanks for your discussion and interest in our research paper
  • asked a question related to Molecular Biology
Question
1 answer
Apparently the lack of tyrosinase:
"Inhibition of tyrosinase can reduce the production of melanin and achieve skin whitening, effectively solving pigmentation (Lall and Kishore, 2014). Therefore, the development of antioxidants, tyrosinase inhibitors, and elastase inhibitors play important roles in solving skin aging and pigmentation" ( https://www.google.com/url?q=https://www.sciencedirect.com/science/article/abs/pii/S0926669020309766&sa=U&ved=2ahUKEwjE4pTd0KyHAxUzHEQIHTzCCpIQFnoECAEQAw&usg=AOvVaw0gD_VQbHW1t1Go0zkPQyIW ).
Ming-Xiang Li, Jing Xie, Xue Bai, Zhi-Zhi Du,
Anti-aging potential, anti-tyrosinase and antibacterial activities of extracts and compounds isolated from Rosa chinensis cv. ‘JinBian’,
Industrial Crops and Products,
Volume 159,
2021,
113059,
ISSN 0926-6690,
Abstract: Rosa chinensis cv. ‘JinBian’, a cultivar of Rosa chinensis Jacq., is one of major raw material of rose tea and possesses sufficient plant resources in China. However, the studies on the chemical constituents and cosmetic activities of R. chinensis cv. ‘JinBian’ are almost blank. The main purpose of this study was to evaluate the anti-aging, skin-whitening, and antibacterial potentials of extracts and chemical constituents of the flower by 1,1-diphenyl-2-picrylhydrazyl (DPPH) radical scavenging, elastase inhibition, anti-tyrosinase, and antibacterial assays. Bioassay results suggested both 95 % and 65 % ethanol extracts possessed significant antioxidant, elastase inhibition, and anti-tyrosinase activities. The combined active extract was studied with bioassay-guided fractionation to give a new compound, kaempferol 3-O-α-l-rhamnopyranosyl (1→6)-(2”,3”-O-digalloyl)-β-d-glucopyranoside (1) and fourteen known compounds (2–15). All compounds were firstly isolated from this species and subjected to the above mentioned bioassays. Ten compounds exhibited antioxidant activities with DPPH radical scavenging rate from 63.40 %–94.04 % under the concentration of 100 μg/mL. The antioxidant activities of 1, 2-phenylethyl 1-O-β-d-(6'-O-galloyl)-glucopyranoside (12), vomifoliol (14), and 4, 4'-dimethoxy-3'-hydroxy-7, 9': 7', 9-diepoxylignan-3-O-β-d-glucopyranoside (15) were firstly found with DPPH radical scavenging rate of 83.24 %, 91.10 %, 63.40 %, and 77.75 %, respectively. The moderate elastase inhibitory activities of 12, ethyl gallate (13), and 15 were firstly found with the inhibitory rate of 43.69 %, 43.25 %, and 35.34 % at the concentration of 100 μg/mL. Multiflorin B (3), 12, and 13 showed strong tyrosinase inhibitory activities with the inhibition rate at 43.83 %–55.80 %, comparing with the positive control, α-arbutin (22.15 %). In addition, 1 showed significant antibacterial activity against Staphylococcus aureus with the MIC50 of 8.51 ± 0.26 μg/mL. Compounds 2–4 and 12–14 showed moderate antibacterial activities against S. aureus. Compounds 6 and 13 also exhibited moderate inhibitory effects against Klebsiella pneumoniae. Above results manifested that R. chinensis cv. ‘JinBian’ possessed potential application values in the development of natural anti-aging, skin-whitening and antibacterial products.
Keywords: Rosa chinensis cv. ‘JinBian’; Antioxidant; Elastase inhibitory activity; Tyrosinase inhibitory activity; Antibacterial activity; Cosmetic potential
Relevant answer
C-SHOT SERUM contains a combination of two molecules with a proven anti-ageing activity: a high percentage (30%) of a more stable vitamin C derivative, 3-O-ethyl-l-ascorbic acid, and lactic acid (1%).
  • asked a question related to Molecular Biology
Question
2 answers
Relevant answer
Answer
Definitely yes
  • asked a question related to Molecular Biology
Question
8 answers
If we take a gel picture (Image attached) then in lane 1 and 2, all the bands are Polymorphic bands? or they are monomorphic? 
Relevant answer
Answer
I think, In lane 1 and 2, there is monomorphic bands but in case of lane 3 and 4, we can observe polymorphic band pattern.
  • asked a question related to Molecular Biology
Question
4 answers
Cellular damage, NOT mutation, causes aging. Cellular error is difficult to define.
Relevant answer
Answer
Aging is a multifaceted process influenced by various biological factors, with significant research pointing to the role of telomeres. Here's an explanation of how telomeres contribute to aging and the discussion surrounding cellular damage:
Telomeres and Aging
1. Role of Telomeres:
  • Telomeres are repetitive nucleotide sequences at the ends of chromosomes that protect them from degradation and fusion with neighboring chromosomes. Each time a cell divides, telomeres shorten slightly, which is a natural part of the aging process.
  • When telomeres become critically short, they trigger cellular senescence or apoptosis (programmed cell death), which prevents cells from dividing further. This is a protective mechanism to avoid the propagation of damaged DNA.
2. Telomere Shortening and Cellular Aging:
  • Telomere shortening is a key indicator of cellular aging. As cells reach their replicative limit (the Hayflick limit), the accumulation of senescent cells contributes to tissue dysfunction and age-related diseases.
  • Factors that accelerate telomere shortening include oxidative stress, inflammation, and lifestyle factors such as poor diet, lack of exercise, and chronic stress.
Cellular Damage and Aging
1. Beyond Telomeres: Cellular Damage:
  • While telomere shortening is a significant factor, aging is also driven by accumulated cellular damage. This includes oxidative damage to DNA, proteins, and lipids, leading to impaired cellular function.
  • Cellular damage can result from environmental factors (e.g., UV radiation, pollutants), metabolic processes (e.g., production of reactive oxygen species), and lifestyle choices (e.g., smoking, excessive alcohol consumption).
2. Antifragility and Cellular Resilience:
  • The concept of antifragility suggests that exposure to mild stressors can enhance cellular resilience and repair mechanisms. For instance, intermittent fasting, exercise, and certain hormetic stressors may promote telomere maintenance and overall cellular health.
  • Strategies to protect telomeres and reduce cellular damage include maintaining a healthy lifestyle, consuming antioxidants, and potentially using telomerase activators (though this area requires more research).
  • asked a question related to Molecular Biology
Question
1 answer
Hello, I am Master student under molecular biology.
I want to find the Shine-Dalgarno sequence after performing a BLAST search in TargetRNA2. Could you please suggest a web tool to predict the Shine-Dalgarno sequence?
I would also like to know if there are any papers that discuss the minimum base pair distance between flanking genes required to categorize them as trans-acting or cis-acting.
Relevant answer
Answer
Unfortunately, TargetRNA2 itself doesn't predict Shine-Dalgarno (SD) sequences. However, following a BLAST search on TargetRNA2, you can use several web tools to analyze the resulting sequence and predict the SD sequence:
This tool from the Center for Phage Technology (CPT) specifically focuses on Ribosome Binding Sites (RBS), which includes the SD sequence. You can paste your sequence from the BLAST result and choose to "Report best hits only" to get the most likely SD sequence.
This website offers various tools for analyzing bacterial sequences. It includes a "Shine/Dalgarno Sequence Finder" where you can paste your sequence and get potential SD motifs identified.
  • asked a question related to Molecular Biology
Question
4 answers
Hi all,
I have a question regarding proper dilutions and conversions of my RNA.
  • According to my protocol, my cDNA kit is optimized for 1ug of RNA and calls for this amount.
  • I would like to dilute all of my RNA samples to the same concentration so I can pipette the same amount of RNA into tubes to make cDNA.
  • I understand how to dilute the RNA samples (adding x amount of water depending on ng/ul) but what concentration should I dilute them to? This is where I get confused. What is the proper quantity of RNA that I should use for cDNA synthesis? How do I get 1 ug from each sample?
This might be a silly question, but I am getting confused by the conversions and wording. Thank you!
Relevant answer
Answer
The amount of RNA you can add will be dependent on your cDNA synthesis kit or RT enzyme. For example in many enzymes you will preform the reaction at a final volume of 20µl and will need to add 4µl of buffer, 1µl of primer and 1µl of enzyme which leave you 14µl free for your RNA.
The concentration you prepare your RNA at will depend on the concentration of your RNA prep. I generally bring all my sample to the concentration of the sample with the lowest concentration.
Once you have that you can calculate how much to take in order to have 1µg in each reaction. For example if your concentration is 200ng/µl than you will need to take 5µl in order to have 1µg of RNA.
  • asked a question related to Molecular Biology
Question
7 answers
It is known that patients with desminopathy often die from pneumonia. Have pathomorphological studies of the lungs been performed in patients with desminopathy?
Relevant answer
Answer
Dear Sagar Nanaso Salunkhe, thank you very much for your detailed answers!
  • asked a question related to Molecular Biology
Question
6 answers
Hi! I'm doing a renilla luciferase assay with coelenterazine and HEK293T cells transfected with AP-1 renilla luciferase. I'm wondering if the cells need to be lysed prior to adding coelenterazine and measuring the luminescence. Coelenterazine is cell-permeable, so I'm wondering if/why the lysis step would be necessary.
I understand that the proteins would be more exposed--would the luminescence be not as great through the cell membrane if we didn't do the lysis?
Relevant answer
Answer
Sindhu Kondath ... Dual Luciferase assay is not to test cell viability. among other things, it is mainly used to test DNA repair capability. For cell viability, I think MTT or WST-1 would be the best assay
  • asked a question related to Molecular Biology
Question
5 answers
Hi there,
I have transfected these two plasmids into 293T packaging cells to encapsulate a pLVX-M-Puro coding plasmid. I had already validated the stability of the pLVX-M-Puro insert by transient transfection. However, after producing the particles, infecting the cells, and selecting with puromycin, I can't observe the expression of my gene of interest by Western Blot.
Any idea what might be happening?
Best wishes,
Relevant answer
Answer
Do you know for sure that all of the plasmids you are using look okay and are not degraded? I would run the psPAX2 and pMD2G on a gel to make sure.
  • asked a question related to Molecular Biology
Question
8 answers
Hi everyone,
I have the problem in endogenous nuclease contamination in my protein preparation purified from E. coli BL21(DE3). I would like to ask your experts in avoiding or removing such contamination. Can you suggest me the protocols or an alternative E. coli host strain with tagged-nucleases?
Thank you in advance.  
Relevant answer
Answer
Gel filtration towards the end removes minute nucleases also.
  • asked a question related to Molecular Biology
Question
3 answers
I will be running my first luciferase assay and the protocol I am using does not specify what filters or filter settings I should use. The plate will be read on a lumometer from Biotek, Synergy HT. What would a general filter setting be to measure the luminescence?
Relevant answer
Answer
Hello Dear Cody,
This is my question too. I am also working with Luciferase for the first time and I have questions about the plate, the device and the wavelengths, but the answers to your questions cannot be found here. Please tell me the answer too.
  • asked a question related to Molecular Biology
Question
10 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
you're right, maybe one day there will be nobody in front of the screen
IA...loss of humanity
fred
  • asked a question related to Molecular Biology
Question
2 answers
Hello,
In the literature, there are some MS/MS results that include hypothetical proteins, which can be shorter than 40 amino acids. I can also find these when I search for an organism in the protein section of NCBI. My question is, would it be absurd if I synthetically synthesize these peptides called hypothetical proteins and test them as drug candidates in certain disease models? Or are studies like the one I mentioned feasible and being conducted? If so, what procedure should I follow? For example, when I find a hypothetical protein, should I first perform a blast and then synthesize and use it if it meets certain conditions?
Is there any chance you could share some references with me that have been done in this manner?
I hope I have been able to convey what I want to ask.
Thank you for your answers.
Relevant answer
Answer
I cannot really answer your questions, but wondered, if this will be of some help. I have seen a review article about how lactobacilli degrade milk protein, and the resulting short peptides have medicinal properties. For instance: "Many studies focused on ACE inhibitor peptides, probably
due to the ease of use of in vitro anti-ACE assays. The well-known
Val-Pro-Pro and Ile-Pro-Pro peptides are produced during milk
fermentation by some Lb. helveticus strains. ... An additional
ACE inhibitory peptide sequence (Ala-Ile-Pro-Pro-Lys-Lys-Asn-
Gln-Asp) was also identified in milk fermented by Lb. helveticus."
Raveschot, C., Cudennec, B., Coutte, F., Flahaut, C., Fremont, M., Drider, D., & Dhulster, P. (2018). Production of bioactive peptides by Lactobacillus species: from gene to application. Frontiers in Microbiology, 9, 409606.
  • asked a question related to Molecular Biology
Question
12 answers
I would like to do genome assembly for the bacteria isolate from the environment. Can any provide me the information or any tutorial? It would be helpful!
thanks!
Relevant answer
Answer
KBase is an online platform with many implemented tools and also examples to follow. Easy for starting as you don't need local resources and can piece together your pipeline or follow published ones.
  • asked a question related to Molecular Biology
Question
13 answers
We are preparing to write some review articles on molecular biology of human diseases. Is there anybody expert who wants to participate?
My e-mail address: zbshirvani@gmail.com
Please write to me and give some info about yourself.
Relevant answer
Answer
Yes I am interesting, Researcher from Sudan work on oncology centre
  • asked a question related to Molecular Biology
Question
5 answers
What are the 3’ end modifications that prevent DNA from being extended by DNA polymerase? Which one has the best blocking effect? Leakage is minimal?
Relevant answer
Answer
Hi there,
A 3'H instead of the 3'OH is 100% efficient to block extension! This is the principle of Sanger sequencing...
  • asked a question related to Molecular Biology
Question
5 answers
Hello everyone,
I hope this message finds you doing well.
​I am writing to ask you a significant question about whole-virus ELISA and its procedure.
Honestly, it seems that coating a microplate with viruses is not as convenient as some papers mentioned, especially when high accuracy is needed. Now, my question is, is there any particular procedure in order to enhance the efficiency of the coating? For instance, what would we do if we tended to expose viral protein to the microplate better than before, based on your experience?
Thank you
Relevant answer
Answer
Coating a microplate with viruses for a whole-virus enzyme-linked immunosorbent assay (ELISA) requires precision and adherence to a standardized protocol to ensure reproducibility and accuracy. Here is a detailed, step-by-step guide on how to coat a microplate with viruses for this purpose:
  1. Virus Preparation:Propagation and Purification: Propagate the virus in a suitable host cell line until you achieve a high titer. Subsequently, purify the virus using techniques such as ultracentrifugation through a sucrose gradient or other appropriate methods to remove cell debris and enhance purity. Quantification: Determine the viral concentration using a method such as plaque assay or TCID50. It's crucial to have an accurate measurement of the virus titer to ensure consistent coating across wells.
  2. Microplate Selection:Choose a high-binding ELISA plate designed for protein interaction. These plates are generally treated to enhance protein (virus) binding and are critical for the stability of the coating.
  3. Virus Dilution:Dilute the virus in a coating buffer, typically carbonate-bicarbonate buffer (pH 9.4), which helps maintain the structural integrity of the virus and promotes optimal adsorption to the plate surface. The concentration of the virus in the dilution should be determined empirically, but typically ranges from 1x10^6 to 1x10^8 particles per mL depending on the virus and the assay sensitivity required.
  4. Coating the Plate:Add the virus dilution to the wells of the ELISA plate. Usually, 50-100 µL per well is sufficient. Ensure that the distribution is even across all wells to prevent variability in assay results. Cover the plate to prevent contamination and evaporation, and incubate overnight at 4°C. This temperature stabilizes the virus and promotes consistent binding.
  5. Blocking:After the incubation, wash the plate 3-4 times with PBS containing 0.05% Tween-20 to remove any unbound virus. This step is critical to reduce background noise in the assay. Block the remaining protein-binding sites on the wells with a suitable blocking buffer, typically 3-5% non-fat dry milk or BSA in PBS, for 1-2 hours at room temperature. This prevents nonspecific binding of antibodies in later steps.
  6. Washing:Wash the plate again as described after blocking to remove any excess blocking agent.
  7. Storage:If not used immediately, the coated plates can be dried and stored at 4°C, sealed to prevent contamination and dehydration. For longer storage, freezing at -20°C or -80°C may be necessary.
By following these detailed steps, you ensure that the virus is properly adhered to the microplate, maximizing the sensitivity and specificity of your whole-virus ELISA. Each step, from the preparation of the virus to the final storage of coated plates, is designed to maintain the functional integrity of the viral antigens and provide reliable, reproducible assay results.
Check out this protocol list; it might provide additional insights for resolving the issue.
  • asked a question related to Molecular Biology
Question
4 answers
Currently, phenotypic characterization is routinely used in diagnostic laboratories for antibiotic resistance measurement due to its cost-effectiveness. With the advent of molecular diagnostic technologies that offer shorter turnaround times and more comprehensive data on antibiotic resistance, is there any chance that they will replace phenotyping and become standardized in practice?
Relevant answer
Answer
The forecast suggests that molecular-based methods will increasingly become standard procedures in monitoring antibiotic resistance. These methods offer advantages such as speed, accuracy, and the ability to detect multiple resistance genes simultaneously. As technology advances and becomes more accessible, these methods are likely to become more widely adopted in clinical and public health settings for monitoring antibiotic resistance trends and guiding treatment decisions.
  • asked a question related to Molecular Biology
Question
8 answers
I am trying to stitch in a 38 amino acid tag to the N-terminal end of my protein (3200bp) to be cloned into a lentiviral vector (~7000bp). The forward primer for the same, along with the overhang and the restriction site, comes about 150bp long. The first round of amplication gives me a band close to about 3000-3500bp along with a lot of other non specific bands at the higher molecular weight range. I then gel elute this specific band and reamplify using it as a template with the same primers but i end up getting a smear on the gel. I have also tried using this gel eluted sample to proceed with the digestion and ligation with my vector but in vain.
My PCR parameters are as follows:
1. 98 degC- 2min
2. 98 degC- 10s
3. 65 degC- 30s (2-4: x25 cycles)
4. 72 degC- 2min
5. 72 degC- 5min
6. 4 degC- hold
I use Q5 polymerase (strangely, I do not get any amplification with Phusion). I have tried a gradient PCR and it generally works in the range of (58-68 degC). I use about 50ng of the plasmid template for amplification. I understand that really long primers hamper the quality of amplification but unfortunately, this is a necessity right now.
I would really appreciate if anyone with experience can help me out here. My molecular biology is not THAT strong so please point out if I am committing any obvious mistakes.
Thanks in advance!
Relevant answer
Answer
Using primers longer than 100 base pairs (bp) for cloning purposes is not a common practice, but it can be necessary for certain applications, such as incorporating large tags, mutagenesis of multiple sites simultaneously, or cloning sequences with high secondary structure. Long primers allow for the introduction of complex modifications and can facilitate the assembly of sequences with precise control over the genetic architecture. However, working with long primers presents unique challenges and considerations.
Design Considerations
  1. Sequence Accuracy: Longer primers have a higher likelihood of containing errors. It's crucial to use high-fidelity synthesis methods and possibly perform sequencing verification after synthesis to ensure accuracy.
  2. Secondary Structure: Analyze the potential for secondary structures within the primer sequence that might hinder hybridization to the template. Software tools can help predict these structures and guide the design to minimize such issues.
  3. Melting Temperature (Tm): The Tm of long primers can be significantly higher than shorter ones, affecting PCR conditions. Ensure that the Tm is compatible with your PCR protocol and adjust annealing temperatures accordingly.
  4. Cost: Synthesis of long primers is generally more expensive. This cost increases with the need for purifications such as PAGE or HPLC to ensure primer quality.
Synthesis and Purification
  1. High-Fidelity Synthesis: Opt for synthesis services that offer high fidelity for long primers, as the likelihood of errors increases with length.
  2. Purification: Standard desalting might not be sufficient for long primers. Consider HPLC or PAGE purification to ensure the removal of truncated products and synthesis errors.
PCR Optimization
  1. Annealing Temperature: Due to the higher Tm, optimize the annealing temperature, possibly using a gradient PCR to find the ideal conditions.
  2. Extension Time: Longer primers may require longer extension times to ensure full-length product synthesis.
  3. Polymerase Selection: Use a high-fidelity DNA polymerase suitable for long amplifications, which can reduce errors introduced during PCR.
Cloning Strategy
  1. Overlap Extension PCR: For assembling fragments or introducing large modifications, consider using overlap extension PCR, where the long primers contain overlapping sequences for subsequent assembly steps.
  2. Gibson Assembly or Similar Methods: Techniques like Gibson Assembly, which can join multiple DNA fragments in a single, isothermal reaction, may be particularly suited for cloning strategies involving long primers.
Troubleshooting
  1. Poor Amplification Efficiency: If amplification is inefficient, assess the primer design for secondary structures or re-optimize PCR conditions.
  2. Non-specific Amplification: High-fidelity polymerases and careful primer design can minimize non-specific products. Additionally, touch-down PCR protocols can improve specificity.
Conclusion
While using primers longer than 100 bp for cloning is challenging, it is feasible with careful design, high-quality synthesis, and optimization of PCR conditions. These primers offer flexibility for complex cloning projects but require meticulous planning and execution to ensure success. Always verify the final construct sequence to confirm that the intended modifications have been accurately incorporated.
Take a look at this protocol list; it could assist in understanding and solving the problem.
  • asked a question related to Molecular Biology
Question
1 answer
Hi!
I am staining iNOS and CD163 in my cells so I am looking for a minus control, expressing neither iNOS nor CD163. But the cell familiar in our lab including panc-1 and 293T seems at least expressing 1 of them. Which cell line is known to not having observable level of these 2 proteins?
Thanks!
Relevant answer
Answer
A cell line that does not express both iNOS (inducible nitric oxide synthase) and CD163 (cluster of differentiation 163) could be a cell line derived from a tissue or cell type that does not typically express these markers under normal conditions.For example, many cancer cell lines, such as HeLa cells or MCF-7 cells, may not express iNOS or CD163 unless they are specifically induced to do so under experimental conditions. Additionally, certain immortalized cell lines derived from non-immune tissues, such as fibroblasts or epithelial cells, may not express these markers.However, it's important to note that the expression of iNOS and CD163 can vary depending on the experimental conditions and the specific context in which the cells are studied. Therefore, it's always a good idea to confirm the expression profile of a cell line using experimental techniques such as immunostaining, Western blotting, or flow cytometry.
  • asked a question related to Molecular Biology
Question
4 answers
I know, IRES enables the coordinated co-expression of two genes with the same vector, used for the expression of two proteins separately.
But I found two kinds of IRES sequences in my plasmid database and literature. Here it is:
IRES:
TCCCTCCCCCCCCCCTAACGTTACTGGCCGAAGCCGCTTGGAATAAGGCCGGTGTGCGTTTGTCTATATGTTATTTTCCACCATATTGCCGTCTTTTGGCAATGTGAGGGCCCGGAAACCTGGCCCTGTCTTCTTGACGAGCATTCCTAGGGGTCTTTCCCCTCTCGCCAAAGGAATGCAAGGTCTGTTGAATGTCGTGAAGGAAGCAGTTCCTCTGGAAGCTTCTTGAAGACAAACAACGTCTGTAGCGACCCTTTGCAGGCAGCGGAACCCCCCACCTGGCGACAGGTGCCTCTGCGGCCAAAAGCCACGTGTATAAGATACACCTGCAAAGGCGGCACAACCCCAGTGCCACGTTGTGAGTTGGATAGTTGTGGAAAGAGTCAAATGGCTCTCCTCAAGCGTATTCAACAAGGGGCTGAAGGATGCCCAGAAGGTACCCCATTGTATGGGATCTGATCTGGGGCCTCGGTGCACATGCTTTACATGTGTTTAGTCGAGGTTAAAAAACGTCTAGGCCCCCCGAACCACGGGGACGTGGTTTTCCTTTGAAAAACACGATGATAA
IRES2:
CCCCTCTCCCTCCCCCCCCCCTAACGTTACTGGCCGAAGCCGCTTGGAATAAGGCCGGTGTGCGTTTGTCTATATGTTATTTTCCACCATATTGCCGTCTTTTGGCAATGTGAGGGCCCGGAAACCTGGCCCTGTCTTCTTGACGAGCATTCCTAGGGGTCTTTCCCCTCTCGCCAAAGGAATGCAAGGTCTGTTGAATGTCGTGAAGGAAGCAGTTCCTCTGGAAGCTTCTTGAAGACAAACAACGTCTGTAGCGACCCTTTGCAGGCAGCGGAACCCCCCACCTGGCGACAGGTGCCTCTGCGGCCAAAAGCCACGTGTATAAGATACACCTGCAAAGGCGGCACAACCCCAGTGCCACGTTGTGAGTTGGATAGTTGTGGAAAGAGTCAAATGGCTCTCCTCAAGCGTATTCAACAAGGGGCTGAAGGATGCCCAGAAGGTACCCCATTGTATGGGATCTGATCTGGGGCCTCGGTACACATGCTTTACATGTGTTTAGTCGAGGTTAAAAAAACGTCTAGGCCCCCCGAACCACGGGGACGTGGTTTTCCTTTGAAAAACACGATGATAATATGGCCACAACC
Somehow, I want to know what is the difference on the expression level of these two sequences. Someone said IRES2 will decrease the expression of the second gene compared with IRES, is it true? Could IRES keep same expression level of two genes (I know people will suggest 2A peptide, but I do not want to introduce any amino acids on my protein)?
Relevant answer
Answer
The difference between using an IRES (Internal Ribosome Entry Site) and an IRES2 lies in their efficiency and specificity in driving gene expression in a bicistronic mRNA.IRES (Internal Ribosome Entry Site): IRES is a sequence element within the mRNA that allows ribosomes to initiate translation internally, bypassing the requirement for a 5' cap structure. When an IRES is present in a bicistronic mRNA, it enables translation initiation of the downstream gene even if the ribosome is still translating the upstream gene. However, IRES elements are generally less efficient than cap-dependent translation initiation, leading to lower expression levels of the downstream gene compared to the upstream gene.IRES2: IRES2 is an improved version of IRES that has been engineered to enhance its efficiency and specificity. IRES2 sequences have been optimized to increase translation initiation rates and reduce leaky scanning (initiation at inappropriate start codons). As a result, IRES2 elements typically lead to higher expression levels of the downstream gene compared to traditional IRES elements.In summary, while both IRES and IRES2 facilitate translation initiation of downstream genes in bicistronic mRNAs, IRES2 generally offers higher expression levels due to its improved efficiency and specificity.
  • asked a question related to Molecular Biology
Question
7 answers
Greetings, dear colleagues!
Our team conducts research on newly discovered SIRC elements in plant genomes ( , which are thought to be MITE transposons losing inverted repeats products, which could influence genome regulation) using bioinformatics, and we plan to conduct experimental molecular biology studies to elucidate the functions of SIRC. The problem is - our team is specialized in molecular bology experiments aiming to reveal the functions of genes, not non-coding DNA elements. That's why I want to ask your expert opinion - what experimental techniques would help to reveal the functions of abundant DNA elements of repetitive nature?
What comes to mind is the creation of mutant lines without several of these elements, but such experiments are too large-scale and can last for years, which is too complicated at the moment.
Another technique that comes to mind is the amplification of certain sequences and examination using circular dichroism spectroscopy to reveal whether given elements have unusual secondary structure like G-quadruplex of triplex DNA etc that could influence processes of genome transcription or replication.
And one more - we thought it could be possible to capture and identify plant proteins that specifically recognize SIRC via some modification of EMSA (electroforetic mobility shift assay) method. Unfortunatelly, up to date we didn't find any mentions of EMSA variant that uses not single purified protein, but whole DNA-free nuclear lysate, with subsequent identification of binding proteins via MALDI-TOF.
What other in vitro experiments could be useful?
Relevant answer
Answer
@Robert Adolf Brinzer The question is, what is the possible structural and functional role of newly discovered SIRC (Short Interrupted Repeat Cassettes) elements in plant genome. The point is that in bacterial genomes there are CRISPR cassettes which are looking similar to SIRC but have nothing common.
  • asked a question related to Molecular Biology
Question
11 answers
A patient with desminopathy (mutation Thr341Pro DES in a heterozygous state) with the progression of the disease has a decrease in taste and smell, immunosuppression, and an increase in IgA in the blood.
Oddly enough, but all this is characteristic of infections, including viral ones. For example, it is known that if the hepatitis C virus is not treated, then death will occur in 20 years.
In the identified case of late onset desminopathy, muscle weakness manifests itself at the age of 30, and death occurs 20 years after the onset of the disease.
Could the desmin mutation in myofibrillar myopathy be caused by an infection?
Perhaps the infection contributes to the progression of desminopathy?
Relevant answer
Answer
Dear Esteemed Colleague,
Greetings. I trust this message finds you deeply engaged in your research and seeking answers to complex questions within the realm of genetics and molecular pathology. Your inquiry regarding the potential role of infection in causing desmin mutations in myofibrillar myopathy is both intriguing and indicative of a keen scientific mind exploring the multifaceted nature of genetic disorders.
To address your question with the precision and clarity it deserves, it is crucial to first understand the nature of myofibrillar myopathies and the role of desmin within this context. Myofibrillar myopathies are a group of neuromuscular disorders characterized by the progressive weakening of muscles and the disintegration of muscle fibers at a cellular level. Desmin, a type of intermediate filament protein, plays a pivotal role in maintaining the structural integrity and function of muscle cells. Mutations in the DES gene, which encodes the desmin protein, are directly linked to certain forms of myofibrillar myopathy.
The genesis of these mutations, particularly those affecting the desmin protein, is primarily genetic, resulting from inherited or de novo mutations in the DES gene. These mutations lead to the production of an abnormal desmin protein, which disrupts the normal architecture of muscle cells, leading to the symptoms associated with myofibrillar myopathy.
Addressing the specific question of whether an infection could cause desmin mutations, it is essential to differentiate between the origins of genetic mutations and factors that may exacerbate the phenotype of a genetic disorder. Genetic mutations, including those affecting the desmin gene, arise from alterations in the DNA sequence. These alterations can be inherited from parents, occur spontaneously during DNA replication, or be induced by certain environmental factors, such as exposure to specific chemicals or radiation. Infections, while capable of causing a wide array of health issues, do not directly induce genetic mutations in the DNA sequence of the genes like DES. However, it is conceivable that certain infections could exacerbate the clinical manifestations of myofibrillar myopathy in individuals already predisposed or carrying a desmin mutation, by stressing the muscular system or triggering inflammatory responses that may further compromise muscle function.
In conclusion, while infections can have significant impacts on overall health and may interact in complex ways with genetic disorders, the mutations in the DES gene that cause myofibrillar myopathy are not directly caused by infections. The mutations are genetic in origin, and the relationship between infections and the severity or progression of myofibrillar myopathy would be more accurately viewed through the lens of infection exacerbating pre-existing conditions rather than causing the genetic mutation itself.
I hope this elucidation addresses your inquiry comprehensively. Should you have further questions or require additional clarification, please feel free to reach out.
Warm regards.
This protocol list might provide further insights to address this issue.
  • asked a question related to Molecular Biology
Question
4 answers
I just found the Platinum SuperFi II DNA Polymerase, which should simplify PCR protocol as it allows uniform annealing temperature of 60°C, but it should also have very high fidelity; >300× higher than Taq Pol, that should be even more than Q5, reported by NEB to have 280× higher fidelity than Taq Pol.
The SuperFi II DNA Polymerase should even allow amplification up to 40 kbp, while Q5 only up to 20 kbp.
This looks like we have new Queen in the HighFidelity DNA Pol area, don't we? Does somebody have experience with this enzyme?
Relevant answer
Answer
Yes and no.
60°C is used for almost all reactions, but not all. For example, in my case, I amplified some genes for Gibson cloning, so my oligos have a high size (50/60bp). In these cases, the annealing step is not necessary and you just proceed to the extension step. The enzyme buffer permits annealing at 72°C. In all my reactions, I always had high amplification and I always followed the protocol parameters.
You just need to be careful with the correct design of your primers and with your DNA sample. Your sample needs to be as pure as possible, and you should use the amount recommended by the protocol in each reaction, which is 10ng if the plasmid gene is being amplified. When I used a bit more sample, the amplification didn't occur.
I do find it reliable, much more so than other enzymes I've used. We haven't had any problems with genes amplified with it and used in cloning.
  • asked a question related to Molecular Biology
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 Molecular Biology
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 Molecular Biology
Question
7 answers
Dear ResearchGate Community, I am reaching out to express my interest in collaborating on the write-up of a research paper related to Cancer Biology, Telomere Biology, and Gut Microbiome. If anyone is working on research based on this field and you are looking for a co-author or contributor to help in the writing process, I am more than willing to be a part of that. With a strong background in biochemistry and molecular biology, I believe I can bring valuable insights and a fresh perspective to the work. I am committed to maintaining the highest standards of research integrity and would be thrilled to contribute to a meaningful project in this vital area of science. If you are interested or know someone who might be, please do not hesitate to connect with me directly or drop a comment below. Thank you for considering this collaboration, and I look forward to potentially working with some of you soon! Best regards, Mashal Naeem #collaboration #researchpaper #research
Relevant answer
Answer
AOA
Dear Mashal
I am interested in collaboration with you. Thank you.
  • asked a question related to Molecular Biology
Question
3 answers
I am currently doing my PhD project which consists of a lot of cloning of new plasmids I am assembling. Our laboratory generally maintains the collection on JM109 strain. But since I am doing a lot of Gibson Assemblies, I have been using electrocompetent DH10B cells for higher efficiency. My question is, can I use standard protocol of preparation of electrocompetent E. coli on JM109 instead of DH10B?
Relevant answer
Answer
Yes, you can adapt the protocol for preparing electrocompetent E. coli cells from DH10B to JM109. However, it's important to note that different strains of E. coli may have slightly different requirements for optimal transformation efficiency, so you may need to optimize the protocol for JM109 cells.
Here's a general outline of how you can adapt the protocol for preparing electrocompetent JM109 cells:
  1. Start with a fresh overnight culture of JM109 cells grown in LB medium at 37°C with shaking.
  2. Inoculate 50-100 mL of LB medium with the overnight culture and grow at 37°C with shaking until the culture reaches an OD600 of around 0.4-0.6. This typically takes 2-3 hours.
  3. Chill the culture on ice for 15-30 minutes to stop growth.
  4. Pellet the cells by centrifugation at 4°C for 10 minutes at 4000 rpm.
  5. Remove the supernatant carefully and resuspend the cell pellet gently in an ice-cold solution of 10% glycerol using a small volume (typically 10% of the original culture volume) to concentrate the cells.
  6. Centrifuge the resuspended cells again at 4°C for 10 minutes at 4000 rpm.
  7. Repeat the wash step with ice-cold 10% glycerol one or two more times to ensure the removal of any remaining LB medium.
  8. After the final wash, resuspend the cells in a small volume of ice-cold 10% glycerol to achieve a concentrated cell suspension.
  9. Aliquot the electrocompetent cells into small volumes suitable for single-use transformations (typically 50-100 µl).
  10. Flash freeze the aliquots in liquid nitrogen and store them at -80°C for long-term use.
  11. To use the electrocompetent JM109 cells, thaw an aliquot on ice, add your DNA (e.g., plasmid DNA for transformation) to the cells, perform the electroporation, and recover the transformed cells in SOC medium before plating onto selective agar plates.
By following this adapted protocol, you should be able to prepare electrocompetent JM109 cells for your Gibson Assembly experiments. It's always a good idea to perform optimization experiments to determine the optimal conditions for your specific application.
  • asked a question related to Molecular Biology
Question
4 answers
Molecular biology
Relevant answer
Answer
Dear Colleague,
Among the structures listed — plasmid, pilus, capsule, and plasma membrane — the entity that contains genes for enzymes and antibiotic resistance is the plasmid. Plasmids are small, circular, double-stranded DNA molecules that are distinct from a bacterial cell's chromosomal DNA. They are capable of autonomous replication and often carry genes that may confer advantageous traits to bacteria, such as antibiotic resistance and the production of enzymes that degrade toxic compounds.
  • Plasmids play a pivotal role in horizontal gene transfer among bacterial populations, significantly contributing to the spread of antibiotic resistance. This characteristic makes them subjects of intense study in the context of infectious disease research and microbial ecology.
  • Pili (plural of pilus), on the other hand, are filamentous structures on the surface of bacterial cells that facilitate attachment to surfaces and other cells. While they are crucial for processes such as bacterial conjugation, during which plasmids can be transferred between cells, they do not themselves contain genes.
  • Capsules are gelatinous layers that encase some bacterial cells, providing protection against desiccation and phagocytosis. Although capsules play a role in bacterial virulence, they do not contain genetic material.
  • The plasma membrane is a phospholipid bilayer that encloses the cell, controlling the passage of substances in and out of the cell. While essential for numerous cellular functions, it does not house genes.
In conclusion, plasmids are the structures among those listed that contain genes for enzymes and antibiotic resistance, underscoring their significance in bacterial adaptation and survival, especially in environments with selective pressures such as antibiotics.
Yours sincerely,
Perhaps this protocol list can give us more information to help solve the problem.
  • asked a question related to Molecular Biology
Question
5 answers
An unopened Sigma-Aldrich (P4557) phenol solution bottle was shaken (prior to the addition of the Equilibration Buffer) and a gel-like layer formed at the bottom of the bottle. The upper phase is still liquid. The bottle was shaken briefly after the phenol solution was taken out of +4 C. What should be done? Should it be heated in order for it to return to liquid?
Relevant answer
Answer
All you need to do is to heat it up slightly, but not with real flame, do that by gently stirring it until it returns to liquid. If this does not work, contact the supplier/company for assistance. Thank you.
  • asked a question related to Molecular Biology
Question
1 answer
I have molecular data (0,1) and a trait with continuous variables. My goal is to detect the significance of markers associated with the trait. Which statistical analysis should I perform? Should I use a t-test, logistic regression, or something else?
Relevant answer
Answer
Can you clarify the roles of the variables you mentioned? If one of them is a dependent variable, for example, which one is it? Thanks for clarifying.
Please clarify too what "a trait with continuous variables" means. Perhaps if you just said what the trait is (and what the continuous variables are), it would help readers to understand better. Thanks.
  • asked a question related to Molecular Biology
Question
6 answers
Q1
We have animal behavior scores of 4 group, Normal+ctrl virus, Normal+down-regulation virus, Model+ctrl virus and Model+down-regulation virus. It has two factors(Independent Variable): Model and virus. Editors suggested we use two way ANOVA to analyze, and now we obtained main effects of Model (F(1, 56)=201.18, P<0.0001) and virus (F(1, 56)=11.17, P=0.00427), as well as Model × virus interactions (F(1, 56)=16.13, P=0.0007).
If we should continue to calculate? For example, Model+ctrl virus vs. Model+down-regulation virus. We want to confirm the role of virus in Model animals.
Q2
Next, we used chemical drug to treat the Model animals and Normal animal. It has 4 drug concentration. Should we still use two way ANOVA to analyze the behavior scores? We want to know the role of different drug concentration in Model animals. And what do we do after two way ANOVA?
Thanks very very much!!!
Relevant answer
Answer
Dear Esteemed Colleague,
Following the completion of a two-way ANOVA, which serves to ascertain the effects of two independent variables on a dependent variable, as well as any interaction between these independent variables, your subsequent steps should be methodically oriented towards a comprehensive interpretation and validation of the results obtained. Here is a structured approach to guide your post-ANOVA analysis:
  1. Examine ANOVA Assumptions: Prior to delving into further analysis, it is crucial to ensure that the assumptions underlying the two-way ANOVA have been met. These include the assumptions of normality, homogeneity of variances (homoscedasticity), and independence of observations. Tools such as the Shapiro-Wilk test for normality and Levene's test for equality of variances can be employed to assess these assumptions. Should any assumptions not be satisfied, corrective measures such as data transformation or the use of non-parametric tests may be considered.
  2. Interpret Main Effects and Interaction Effects: The core of your analysis will involve interpreting the main effects of each independent variable and any interaction effects between them. A significant main effect indicates that different levels of an independent variable have significantly different impacts on the dependent variable. A significant interaction effect, on the other hand, suggests that the effect of one independent variable on the dependent variable varies depending on the level of the other independent variable. It is essential to carefully interpret these effects in the context of your research question.
  3. Conduct Post Hoc Tests for Multiple Comparisons: In the event that your ANOVA results indicate significant effects, post hoc tests are necessary to determine which specific groups differ from each other. Techniques such as Tukey's HSD (Honestly Significant Difference) test, Bonferroni correction, or Sidak adjustment are commonly employed for pairwise comparisons while controlling for the family-wise error rate. The choice of post hoc test depends on the specific characteristics of your data and the comparisons of interest.
  4. Evaluate the Magnitude of Effects: Beyond statistical significance, assessing the practical significance of your findings is vital. This can be achieved by calculating effect sizes, such as partial eta squared (η²) or Cohen's d, which provide insight into the magnitude of the differences or relationships observed. These measures help to contextualize the importance of your findings in real-world terms.
  5. Graphical Representation of the Results: Visualizing your data and the results of the ANOVA can greatly aid in their interpretation. Interaction plots, for example, are particularly useful for visualizing how the levels of one independent variable affect the outcome across the levels of another independent variable. Box plots and bar charts can also be effective in displaying the central tendencies and variabilities within and across the groups.
  6. Report Your Findings: The final step involves a detailed and coherent reporting of your methodology, analysis, results, and interpretations. This should include a summary of the ANOVA results, post hoc tests, effect sizes, and any graphical representations. It is crucial to discuss the implications of your findings in the context of existing literature and your research objectives, including any limitations and suggestions for future research.
By following these steps, you will ensure not only the rigorous analysis of your two-way ANOVA results but also the meaningful interpretation and reporting of these results within the broader context of your research field.
Should you require further assistance or clarification on any of these steps, please do not hesitate to reach out.
Warm regards.
This protocol list might provide further insights to address this issue.
  • asked a question related to Molecular Biology
Question
8 answers
It is known that in the early stages of desminopathy the muscles most often affected are: Semitendinosus, Gracilis and Sartorius. What is the reason for the damage to these particular muscles?
Relevant answer
Answer
Desminopathy, also known as desmin-related myopathy (DRM), is a rare genetic muscle disorder that affects the protein desmin. Desmin is an essential component of the intermediate filaments that provide structural support within muscle cells. Mutations in the DES gene, which codes for desmin, lead to disruptions in the normal structure and function of muscle fibers.
The muscles you mentioned - Semitendinosus, Gracilis, and Sartorius - are often affected at the onset of desminopathy due to their specific characteristics and biomechanical roles.
1. Semitendinosus: The semitendinosus is one of the three hamstring muscles located in the back of the thigh. It plays a key role in knee flexion and hip extension. The semitendinosus muscle is frequently involved in desminopathy due to its high proportion of slow-twitch muscle fibers, which are more vulnerable to desmin-related abnormalities.
2. Gracilis: The gracilis muscle is a long, thin muscle located in the inner thigh region. It is involved in hip adduction and knee flexion. Similar to the semitendinosus, the gracilis muscle also consists of a high proportion of slow-twitch muscle fibers, making it susceptible to desmin-related abnormalities.
3. Sartorius: The sartorius muscle is a long, strap-like muscle that runs diagonally across the front of the thigh. It plays a role in hip and knee flexion and also assists in thigh rotation. The sartorius muscle is affected in desminopathy due to its similar composition of slow-twitch muscle fibers.
The predilection for these specific muscles in desminopathy may be attributed to their fiber type composition and the mechanical stress they experience during certain movements. However, it is important to note that desminopathy can affect other muscles as well, and the degree and pattern of muscle involvement may vary among individuals with the same genetic mutation.
It is advised to consult with a medical professional or genetics specialist for a more accurate assessment of muscle involvement and management of desminopathy.
  • asked a question related to Molecular Biology
Question
1 answer
User-friendly software tool designed for analyzing the final images generated from Gel Electrophoresis.
Relevant answer
Answer
Hi!
Please send
Email Address: rukwazir@gmail.com