Science topic

Peptides - Science topic

Explore the latest questions and answers in Peptides, and find Peptides experts.
Questions related to Peptides
  • asked a question related to Peptides
Question
2 answers
I have a large number of peptides. Is it possible to save a peptide structure in Pymol in PNG formally without having any extra spaces around it? I am using Pymol in Python to save the peptide PDB file in PNG format.
Relevant answer
Answer
For making images I use the command:
ray 2400, 2400
png FileName, dpi=3000
This should save it without the background.
I try to minimize the extra space by focusing the GUI window on the protein/peptide. Alternatively, you can use an image editor. The simplest is to use PowerPoint, but you can also use SketchBook, Illustrator, Krita, Corel, Canva, etc., and crop out the background with their magic select tool.
I use SketchBook.
Note: You do need the PyMOL license to do this, especially if you want to publish the images.
  • asked a question related to Peptides
Question
2 answers
Hey, I have been trying to design a peptide for a surface receptor using the RF-diffusion, But every time I am getting a peptide that has only glycine as the residue in results. I have looked at multiple literature but could not find relevant explanation.
Relevant answer
Answer
Thank you Kais Khudhair al Hadrawi for the response. My initial inputs were not correct, I have resolved the issue.
  • asked a question related to Peptides
Question
3 answers
I am doing coarse-grained MD simulations using the CG-MARTINI forcefield of GROMACS and my system consists of a box with two different peptides in solution. Is there any way in which I can colour these two peptides differently (in order to distinguish between them in the same system clearly) while visualising using the VMD program?
Relevant answer
Answer
Say you want to color just 2 objects in VMD different colors, it is relatively simple. You can do it in two main ways.
1) The easiest is to go to the VMD main window, to the "Graphics" tab, and then click on the "Representations..." tab.
A window will appear. In this window, you should select the coloring method ColorID from the drop-down menu. The drawing method and other options are down to you.
Now you need to make selections of the proteins. Go into the GRO or PDB file and write down the atom number with which each protein starts and ends. Into the "Graphical Representations" window that previously opened write those numbers into the "Selected Atoms" line like this: index (first atom) to (last atom) and then press enter.
For example: index 0 to 273
Next, you are going to click on the "Create Rep" button in the same window, I should create a new representation by copying the previous one. Change it by writing the atom numbers of the second protein (example: index 274 to 547) and press enter.
Afterward, you just need to change the color ID number by choosing from the drop-down menu the color you like for each protein.
In summary: Graphics -> Representations...-> select "index (first atom) to (last atom) -> ColorID coloring method -> repeat process after creating new representation just with the second proteins atom numbers
2) The more advanced method is to go to "Extensions" in VMD Main -> Tk Console and then paste this into the command prompt and press enter:
mol delrep 0 top
#First protein
mol selection {index 0 to 273}
mol color ColorID 0
mol representation Lines
mol addrep top
# Second protein
mol selection {index 274 to 547}
mol color ColorID 1
mol representation Lines
mol addrep top
Just change the code by adding your atom ID numbers, ColorID numbers, and drawing methods. You can copy-paste the section of code that has "mol selection" to "mol addrep top" for any number of proteins you want to color. I even used Excel to create code to color 200 peptides with different colors automatically.
NOTE: Be careful when selecting atom indices in different files. GRO files count the first atom from 0, PDB files from 1.
  • asked a question related to Peptides
Question
2 answers
pi->pi* transitions are seen by a positive intensity around 190-195 ('perpendicular') and a negative intensity around 208 ('parallel') in alpha helices. I have a protein that displayed a helical style profile where the pi->pi* perpendicular (190-195) is much more intense and there is minimal pi->pi* parallel intensity (208). What conformation is likely to cause this and why? It would help to know where on the peptide backbone these transitions are taking place and why they would be more prevalent on certain structures.
Relevant answer
Answer
Possibly due to increased helicity and compact packing of the protein ligand complex
  • asked a question related to Peptides
Question
2 answers
I used NHS-PEG4-Azide to label protein and temporarily store the labelled protein at 4 degrees. Is it suitable for long-term storage at this temperature?
Relevant answer
Answer
In general, one of the commonly used storage conditions for azide-labeled proteins or peptides is to keep them in buffer solution at a low temperature, usually at -20°C or even lower, such as -80°C. This helps slow down any potential degradation reactions and maintain the integrity of the labeled molecules. In addition, storing them in the dark can prevent any photochemical reactions that may occur... On the other hand, storing azide-labeled proteins or peptides at low temperatures in a suitable buffer solution and protecting them from light should help maintain their preservation. Its stability over time.
  • asked a question related to Peptides
Question
2 answers
Due to factors outside of my control, peptides in my ESI-MS data have been ionised normally by protons ([M+2H], [M+3H]...) but also by sodium ([M+2Na], [M+3Na]...).
Is there a way to configure MaxQuant's andromeda search engine to look for the sodium-ionised peptides as well?
Thanks!
Relevant answer
Answer
James M Fulcher Thanks for your answer! I agree it is probably not trivial, because you cannot assume that *all* ionisations are with sodium rather than a proton, so it sort of explodes combinatorically at the MS2 level.
Unfortunately, I cannot rerun the samples, because I am trying to apply a new analysis pipeline to existing data on PRIDE, and I have found this phenomenon in other researchers' published data.
I have tried setting up sodiation as a PTM on MaxQuant, and I think it could work, but it increases the analysis time from minutes per sample to days per sample, so might be useful for a proof-of-concept, but if anyone has suggestions for a better solution – or knows of a way to this in MaxQuant already – I would love to hear them!
  • asked a question related to Peptides
Question
4 answers
Hello!
I am working on investigating the effect of freeze and thaw on the peptides stability. The peptide is relatively short less than 50 AA so my question is would this have an impact at all? Is it work exploring this? Does controlled freeze thaw a valid solution?
Relevant answer
Answer
I have also heard that the pH of phosphate buffer changes when freezing, so it is not preferred for a storage solution with sensitive materials.
If the solution is flash-frozen, the solutes do not have time to separate from the ice, so this method is preferable to slow-freezing.
  • asked a question related to Peptides
Question
2 answers
I am planning to do mass spectrometry for protein hydrolysate. How can I desalt the peptide mixture without affecting hydrophilic and hydrophobic peptides?
Relevant answer
Answer
To desalt peptide mixtures while preserving their hydrophilic and hydrophobic properties, you can use a reversed-phase solid-phase extraction (SPE) method. This method separates peptides based on their hydrophobicity using a hydrophobic stationary phase, while the mobile phase is a water-miscible organic solvent. The method allows for the elution of hydrophilic peptides first, followed by hydrophobic peptides using a stepwise increase in the organic solvent concentration. Alternatively, size exclusion chromatography (SEC) can also be used to separate peptides based on their size and hydrophilicity. SEC separates peptides by size, with larger peptides eluting first, and smaller peptides eluting later. This method can also be used to desalt peptide mixtures without affecting their hydrophilic and hydrophobic properties.
  • asked a question related to Peptides
Question
1 answer
Hi all,
Kindly I have questions about peptide design. I'm now in the process of designing a peptide based on protein structure and the length of peptide is 45 a.a . I have predicted the structure by many tools but my problem the two catalytic residues in peptide are not located in front of each other so kindly is there any advice or tool we can detect the two catalytic residues in peptide before build the structure. Or is better to use scaffold? if yeah are there any mini or small scaffold for peptide.
The picture attached below is just to make it clearer (this is the protein that has catalytic residues, I need to make two catalytic residues of my peptide in front of each other)
thanks
Relevant answer
Answer
What do you plan to do with your 45mer peptide? Are you hoping it will have catalytic activity?
  • asked a question related to Peptides
Question
1 answer
We have the peptide and mRNA sequences; is it possible to create an antibody?
Relevant answer
Answer
You don't really "design" an antibody. You grab your peptide or your mRNA and you vaccinate a critter. Its immune system then designs the antibody for you. Yes it really is possible. About $5k to hire a service to do it all for you, just off the sequences.
  • asked a question related to Peptides
Question
1 answer
We have mRNA sequence and peptide sequence if possible design antibody?
Relevant answer
Answer
Designing antibodies involves several approaches aimed at generating antibodies with desired properties, such as specificity, affinity, and functionality. Here are some key methods and strategies used in antibody design:
  1. Monoclonal Antibodies (mAbs) vs. Polyclonal Antibodies (pAbs):Monoclonal antibodies are derived from a single clone of B cells and are highly specific to a single epitope on the target antigen. They are typically produced by hybridoma technology or recombinant DNA technology. Polyclonal antibodies are produced by multiple clones of B cells and recognize multiple epitopes on the target antigen. They are generated by immunizing animals with the target antigen and collecting serum containing a mixture of antibodies.
  2. Antibody Selection:Select the appropriate antibody type (e.g., IgG, IgM, IgA) based on the intended application and desired antibody characteristics. Choose the antibody species (e.g., mouse, rabbit, human) that best suits the experimental system and minimizes potential cross-reactivity.
  3. Antigen Selection and Immunization:Select the antigen or antigenic epitope of interest based on its relevance to the research question or application. Design and synthesize peptides or recombinant proteins representing the antigenic epitope for immunization. Consider factors such as antigenicity, immunogenicity, and conservation of the target antigen when selecting the immunogen.
  4. Hybridoma Technology:Generate monoclonal antibodies using hybridoma technology by fusing antibody-producing B cells (isolated from immunized animals) with immortal myeloma cells to create hybridoma cell lines. Screen hybridoma clones for antibody production and specificity against the target antigen. Expand and characterize selected monoclonal antibody-producing hybridoma cell lines.
  5. Recombinant Antibody Technology:Generate recombinant antibodies using phage display, yeast display, or other display technologies. Construct antibody libraries using synthetic or natural antibody repertoires. Select antibody variants with desired binding properties through iterative rounds of selection and screening against the target antigen.
  6. Antibody Engineering:Engineer antibody fragments (e.g., Fab, scFv, nanobodies) with improved characteristics such as smaller size, enhanced stability, or altered effector functions. Introduce mutations into the antibody variable regions to modulate affinity, specificity, or other binding properties. Conjugate antibodies with labels (e.g., fluorophores, enzymes, nanoparticles) for detection or therapeutic applications.
  7. In silico Antibody Design:Use computational methods such as homology modeling, molecular docking, and molecular dynamics simulations to design antibodies with improved binding affinity or specificity. Design antibody-antigen complexes to predict antibody-antigen interactions and optimize binding interfaces.
  8. Functional Antibody Screening:Characterize antibody function through in vitro assays (e.g., ELISA, Western blot, immunoprecipitation) and in vivo assays (e.g., cell-based assays, animal models). Assess antibody specificity, affinity, and functionality under relevant experimental conditions.
By combining these methods and strategies, researchers can design and generate antibodies tailored to their specific research needs, facilitating a wide range of applications in basic research, diagnostics, and therapeutics.
  • asked a question related to Peptides
Question
1 answer
Hi,
I am looking for computational tools to get a score/measurement of goodness of fit/binding affinity/any other metric to present a binding between a protein and a peptide, preferably via Pymol.
Any help will be greatly appreciated!
Best,
Chen
Relevant answer
Answer
I think you can use molecular docking tools like AutoDock or AutoDock Vina to calculate the binding affinity between a protein and a peptide. PyMOL can be used for visualization but not for calculating affinity. I hope this helps!
  • asked a question related to Peptides
Question
3 answers
I am planning to synthesize a peptide :YSTCDFIM (MW 978). However, no matter how I did the synthesis, the LCMS result always showed peaks at 529, 644, 791, 1035. It seems like I always missed STDF residues in my sequence. I don't know why it happened.
Relevant answer
Answer
Okay, no problem.
  • asked a question related to Peptides
Question
2 answers
I recently encountered an intriguing situation while examining a plasmid constructed by someone else for a eukaryotic expression system. This plasmid contains a unique arrangement of open reading frames (ORFs) that has sparked several questions regarding the potential outcomes of their translation.
In this plasmid, there is an ORF near the 5' end, where the translation initiation site is quickly followed by a stop codon, potentially resulting in a very short peptide. More interestingly, nested within this first ORF is a second ORF that begins inside the first ORF and could potentially translate into a much longer protein, consisting of 500 amino acids.
Given the common understanding that eukaryotic transcripts typically feature a single ORF, the discovery of this arrangement has led me to ponder the following questions about the translational dynamics in this specific scenario:
  1. In the context of this plasmid, will the translation machinery be capable of bypassing the short ORF to translate the longer protein, or will it prioritize the translation of the short peptide due to its proximity to the 5' end?
  2. If both peptides are indeed translated, what might be the expected ratio between the production of the long and short peptides?
  3. Is there a possibility that only the short peptide will be translated, effectively ignoring the translation potential of the longer, nested ORF?
Furthermore, I'm curious about how this scenario might differ if the plasmid were used in a prokaryotic system, which is known for its ability to translate multiple ORFs within a single transcript.
I'm seeking insights, experiences, or any relevant literature that could help shed light on the translational strategies employed by cells when faced with plasmids containing nested ORFs, especially in the context of eukaryotic expression systems.
Thank you in advance for sharing your knowledge and experiences.
Relevant answer
Answer
There are a myriad examples of this sort of ORF structure in eukaryotic viral systems (particularly RNA ones), where it has evolved to diversify/expand coding capacity and/or regulate viral gene expression. At first pass you can predict relative expression levels by examining the Kozak consensus around each AUG codon. We usually consider the -3 and +4 positions to categorise them as strong, middling and weak. If the first AUG is strong, you typically won't get much of the second product. If the first AUG is medium or weak and the second is strong, you'll get lots of the second product. Confounders to look out for; 5'-UTR length and protein stability. AUGs very close to the 5'-cap are often not translated well. Shorter proteins are harder to detect and if they're too short (or the wrong sequence) to fold up into a coherent structure, they'll be turned over rapidly and can be hard to detect unless (or even if) the proteasome is turned off.
After that, it can get properly complicated with all sorts of ribosomal gymnastics depending on the exact sequence of the transcript.
Bugs - need to consider Shine-Dalgarno (sp?) sequences or lack of, I think. But my undergrad lectures on this were many years ago :)
Cheers
Paul
  • asked a question related to Peptides
Question
1 answer
Hi all, I am a PhD student working on the effect of Ang (1-7) on Alzheimer's disease. I am using Ang (1-7) peptide in electrophysiology and Western blot techniques. I could not see any effect with this peptide. More likely something related to the preparation!!
I prepare it in a distilled water ( Mass: 25 mg
Molecular weight: 899 Daltons
Concentration: 25 mM
Volume of water= 1.112 ml
And allocate them in 20ul tubes.)
Any tips to make the peptide work please !!!
Thanks a lot
Relevant answer
Answer
I might be a little concerned about the pH of the solution, since you dissolved the peptide aty high concentration in water with no buffer. Spot a few microliters of the stock onto a pH strip to see if it is strongly acidic, which might affect the stability of the peptide.
  • asked a question related to Peptides
Question
2 answers
BSA assay and Fluorometric peptide assay do not work for the quantification of the Pepstatin A (aspartic protease/cathepsin D inhibitor/peptide). Does anybody have an idea to quantify it besides using LC-MS?
Relevant answer
Answer
Somewhat less complicated than LC_MS I would suggest reversed-phase LC in combination with UV detection at 218 nm employing a proper (thus not to steep!) acetonitril gradient using 0,1% TFA.
  • asked a question related to Peptides
Question
2 answers
Hi All,
This is a question regarding calculation of m/z values of fragment ions from peptide/proteins in mass spectrometry analysis:
1. Does anyone have a recommended tool for calculating m/z values of fragment ions from peptides (such as: input=peptide sequence, output= all y and b-ions, with charge states +1 and +2, etc)?
2. I have previously been using the Proteomics Toolkit on the server: https://db.systemsbiology.net/
This tool is now terminated.
Instead, I have now used the MS/MS fragmentation calculator tool on the server https://proteomicsresource.washington.edu/protocols06/.
However, the two tools do not provide the same m/z values for the same peptide!?
Please see the list below for a direct comparison of the values obtained with the two tools:
Example:
When asking for fragmentation of the peptide with sequence:
GYSEKCCLTGCTKEELSIACLPYIDF
, I get the following results:
Tool 1, Proteomics toolkit calculations (ave mass):
Precursor (M): 3115.34528
Precursor (M+H)+: 3116.35256
Precursor (M+2H)2+: 1558.7
Precursor (M+3H)3+: 1039.45573
Precursor (M+4H)4+: 779.84363
Example of a few selected fragment ions:
Y17(+2): 1008.45486
Y16(+2): 979.94412
Y11(+2): 656.32562
Tool 2, MS/MS fragmentation calculator (ave mass):
Precursor (M): 3115.727440
Precursor (M+H)+:3116.734716
Precursor (M+2H)2+:1558.870996
Precursor (M+3H)3+:1039.583090
Precursor (M+4H)4+:779.939136
Example of a few selected fragment ions:
Y17(+2): 1009.203266
Y16(+2): 980.677606
Y11(+2): 656.804096
As you can see, the two tools do not provide the same values? Which tool provides the correct m/z values and what is going on? :-)
Thank you for your help!
Relevant answer
Answer
Hi,
Why not look at monoisotopic mass?
The difference in masses obtained can be due to the way the elemental masses are set in the algorithm of the calculator: a difference in digits/rounding can result in large differences in peptide masses, let alone that of complete proteins..!
There is no 'correct' answer, I guess, it's more about the accuracy taken into consideration.
Hope this helps.
  • asked a question related to Peptides
Question
4 answers
Hi,
I did dG MM/GBSA calculation for a protein-peptide complex. I want to know the numerical value of contribution of each residue of peptide to that total dG binding energy. How can I do that?
Relevant answer
Answer
okay. Thanks for your reply.
  • asked a question related to Peptides
Question
2 answers
Hi there, I used thermo's TMT 10plex kit to label peptides and pool them together for LCMS/MS analysis. I am analyzing my data in Proteome Discoverer 2.3 and wondering how to quantify the number of peptides that were labeled vs unlabeled to make sure my labeling strategy worked.
When I run the processing and consensus workflows recommended by thermo for this data, I get protein IDs and quantitation for most of the proteins- I have the TMT label set as a static modification for N-terminus for the peptides and on lysine residues. So, all of my identified peptides and proteins have the TMT label identified as a modification. Surely there are some peptides that the program can identify that weren't labeled, so how do I find those peptides and determine the ratio of labeled:unlabeled peptides in my sample?
Also, even though all of the identified peptides in this workflow are marked as having the TMT label, not all of them are quantified. Does anyone know why this happens?
Thank you!
Talia
Relevant answer
Answer
Hi Talia,
I have looked into this particular question in the Methods paper I have published ; please see the link below. I did not use Proteome discoverer - I used MaxQuant and configured each of the TMT channel modification as a variable modification so that I could search my mass spec file to see how many peptides were not labeled.
Let me know if you can access the paper link below, if not I can also just email it to you.
All the best,
Hediye.
  • asked a question related to Peptides
Question
6 answers
Hi,
I performed a competition assay of serially diluted concentrations of the unlabeled peptide by adding a constant concentration of the FITC-labeled peptide. I have observed higher fluorescence intensity in higher concentrations of unlabeled peptide compared to the lower concentrations of the unlabeled peptide. As per literature we should have lower intensity in higher concentration so of unlabeled peptide. Could you kindly clarify why this is happening or should it be the opposite.
Relevant answer
Answer
Thank you for your suggestions.
  • asked a question related to Peptides
Question
2 answers
Hi everyone,
I'm part of a team keen on developing a deeper understanding of protein interactions within a cellular context. Our primary goal is to identify and implement the most effective assay for measuring interactions between:
  1. Two proteins
  2. A protein and a peptide
We are specifically looking for methods that:
  • Function within a cellular context (not in-vitro)
  • Provide quantifiable affinity scores for comparison
  • Allow high-throughput analysis, potentially comparing approximately 500 proteins against a comprehensive proteome or an extensive peptide library
We are navigating through the initial stages of this project and would immensely appreciate any insights, experiences, or recommendations on methodologies, technologies, or specific challenges we should anticipate.
Thank you in advance for your valuable input!
Relevant answer
Answer
Dear Chen Lior
I believe you can try to use the holdup method adapted to cellular context. You can gie a look to Zembo et al. Sci. Adv. 2022 "Native holdup to measure binding affinities from cell extracts". The title alone already evokes all the requests you have.
Best
  • asked a question related to Peptides
Question
1 answer
I have run MaxQuant in LFQ mode, and it seems that some of the variable modification specified in the specific-group parameters are displaying an intensity of 0 despite the protein/peptide has been listed in the "modification.txt" document. I have also tried running iBAQ mode and that didn't seem to solve the problem. When checking the proteingroup,txt the protein has been assigned with an intensity. Any idea why the protein could be listed in the modification.txt without having a value ?
Thanks
Relevant answer
Answer
Hi Ibtissam,
did you defined the mentioned variable modifications in global parameters/protein quantification tab (peptides for quantification)?
Best,
Murat
  • asked a question related to Peptides
Question
7 answers
It is a cationic peptide with a length of less than 20 amino acids.
Relevant answer
Answer
Zahra Hajihashemi The lipid composition of eukaryotic and procaryotic cells is quite different. Generally, there is an higher proportion of anionic lipids in bacterial membranes than in mammalian membranes. Therefore, to target the bacterial membrane, you can add cationic amino acids on your peptide (lysine, arginine or histidine). What we have seen from our experiments, there must be a right balance between the positive net charge and the hydrophobic moment of the peptide. Increasing the positive net charge increases the performance (up to the a certain point). If the hydrophobic moment is too high, you lose in selectivity towards bacterial cell membranes. In order to improve the stability, you can incorporate non-natural amino acids.
  • asked a question related to Peptides
Question
10 answers
Hello everyone, I'm working with predicted antimicrobial peptide using machine learning. Recently, we use an antimicrobial peptide from DRAMP which had been verified to have antimicrobial activity. We synthesized the peptide exactly as what provided in DRAMP. so we did test using disc diffusion assay but no activity. is there any possible issue like it must be tested in microdilution method or due to the synthesis parameter? Thanks in advance for the help and idea.
Relevant answer
Answer
Did you characterize the peptide, both with bioinformatics and experimental? I mean:
-Using for example Heliquest (https://heliquest.ipmc.cnrs.fr ) you get a proper idea about the peptide (its hydrophobicity, net charge, etc.). See for classification for example:
-Before checking in your assay, I think it is good to check the peptide in some way. If for example bioinformatics predicts membrane activity you can check quickly by using a monolayer technique or depending on what you have available in the lab check by circular dichroism (with and without lipids), see for example:
If this seems fine, then do realise that sometimes a pretty insoluble peptide (as described in the link above) that requires something like DMSO or TFE is all of a sudden reasonably soluble in aqua dest. as described here:
However, in your medium there will be a buffer containing salt (and a certain pH) and the ionic strength in the medium can cause aggregation (this can be checked with the peptide and the use of for example circular dichroism).
Obviously, something might have gone wrong with the synthesis, or the prediction is simply wrong, and the peptide has no antimicrobial activity but a lot can be checked before one have to draw this conclusion.
Best regards.
  • asked a question related to Peptides
Question
1 answer
"The reactivity of peptides was confirmed using western blotting and enzyme-linked immunosorbent assay (ELISA) assays"
Relevant answer
Answer
Assuming the peptide functions by binding. The target protein could be immobilized on a plate or transferred to a blot. Then the peptide (likely also with fluorescent / chemiluminescent tags) could be applied to bind to the protein for detection.
  • asked a question related to Peptides
Question
2 answers
Hello everyone,
I am trying to isolate antimicrobial peptides from plants. After precipitating the peptides with acetone, most of the pellet (peptides) did not dissolve. I used dH2O/PBS to dissolve the pellet. I do not want to use SDS or urea to dissolve the pellet because it will affect my cell culture experiment.
This is my peptides isolation protocol:
-The plant material was ground in a blender. For extraction, 10% CH3COOH (Acetic Acid) in the presence of a commercial proteinase inhibitor was added to the floured plant parts (added to the ground material at a ratio of 1:10 (w:v)).
- The extraction was carried out with continuous vigorous stirring for 1 h at room temperature. After 1 h, mixture was sieved; fine particles were separated by centrifugation at 4700 rpm for 10 min. The supernatant was filtered through a Whatman paper filter.
-Cold acetone (-70 °C) was poured into the filtrate at a ratio of 1:7 with gentle stirring; the mixture was then kept at +4 °C for overnight.
-After this time, the suspension was centrifuged at 4700 rpm for 10 min. The supernatant was removed and the resulting fraction was dried at room temperature. The dried precipitate was redissolved in dH2O or PBS.
Relevant answer
Answer
Try dissolving it in dimethylsulfoxide (DMSO). This is a water-miscible solvent that is generally very useful for this sort of thing, and it is compatible with subsequent biological experiments when diluted to a few % by volume.
  • asked a question related to Peptides
Question
3 answers
Hey all! I have a question about my proteomics data evaluated in Proteome Discoverer. I got three volcano plots of three biological groups in a ratio with control group. In one group I see a strange pattern, while other two look normally. Log2 ratio is somehow 100% -related to p-values with no exception (please see the graphs). Obviously it is not the issue of plotting itself, but in calculating the ratio or the p-value. Quantification was done using non-nested design, label-free quatification, pairwise-based ratio calculation, t-test, normalization of total peptide amount.
Does anyone knows the reason for that pattern?
Thanks a lot!
Relevant answer
Answer
I've also had some funny looking volcano plots from PD data. Discussion here: https://www.researchgate.net/post/Artefact_in_volcano_plot
I never got to the root cause, but the funny lines of points lining up were an artefact of normalisation - mind you, the underlying table of PSMs from PD was the original source.
Unfortunately I never got around to identifying why PD gave odd output.
Sorry I can't be more helpful!
Sam
  • asked a question related to Peptides
Question
6 answers
Hello, I am proteomics researcher.
we got stuck in problem detecting immunopeptidome HLA class peptides.
After, enriching peptides, we detect 150 ng/ul concentration of peptide using nanodrop (protein A 280 mode).
and about 750ng of peptides were injected to mass spectrometer. (our mass spectrometer is tims-tof, so if we inject 200ng of HeLa peptide, 40000 peptides can be detected.)
However, 100 peptides were detected in our HLA sample. Furthermore, intensity of peptides signal is low...
why is a large amount of peptides detected in the nanodrops although there seems to be a small amount of peptides in mass spectrometry data?
I heard that A 280 mode in nanodrop detect peptide concentration by measuring tryptophan or tyrosine.
is it possible there are many free tryptophan and tyrosine in the sample, so it make nanodrop conecntration high but not be detected in mass spectrometer?
If you have any idea, please let me know
thank you very much
Relevant answer
Answer
Thank you for your kind reply.
I will try searching HLA peptide atlas.
Best regards
Jaekwan.
  • asked a question related to Peptides
Question
4 answers
I am having a problem with getting primary structure of peptide. I have used an online tool, PepDraw but it does give option of downloading. How can I get high quality primary structure of peptide.
Relevant answer
Answer
Yes, this is a frequently asked matter. See for suggestions:
or for example https://www.softwaretestinghelp.com/increase-resolution-of-image/ (scroll down towards How to improve the resolution of a photo).
It is possible since I see papers with figures of PepDraw generated results, see for example
Article A short artificial antimicrobial peptide shows potential to ...
Best regards.
  • asked a question related to Peptides
Question
1 answer
Why choose PNA-HPLC methods for divalent siRNA detection over PCR?
Relevant answer
Answer
PNA have favorable binding conditions and -kinetics compared to oligonucleotides and are stable against nucleases and proteases.
  • asked a question related to Peptides
Question
6 answers
Dear ResearchGate Community,
I am currently facing a challenge in the LC-MS analysis of a highly hydrophobic peptide (already purified to >98% by a manufacturer) on our 1260 LC-MS System.
The experimental setup involved running a gradient of 5-80% ACN over 30 minutes on an analytical column (C18, 95Å, 4.6 x 50 mm, 5 µm). I injected 1, 2, 5, and 50 µl from a sample with a concentration of approximately 0.5 µg/µl (prepared in either 100% Methanol or 80% ACN+20%H2O+0.1%FA).
However, I observed multiple peaks at retention times between 21-24 minutes with my expected m/z in the Total Ion Current (TIC) MS. Interestingly, there were no UV peaks at either 220 or 280 nm (see pic).
Has anyone else observed such behavior (i.e., broad peaks in MS with no UV signal) with any highly hydrophobic peptide or compound?
I would greatly appreciate any suggestions or tips to resolve this issue and achieve a single peak in both TIC and UV (220nm) for this highly hydrophobic peptide.
P.S.: I can observe a single peak in both MS (TIC) and UV (220nm) with my water-soluble hydrophilic peptide, indicating that the LC-MS System is functioning properly with the chosen mobile phases (A: 0.1% TFA/water, B: 0.1% TFA).
Thank you in advance for your assistance.
Relevant answer
Answer
I would suggest an initial UV scan to figure out whether zhe peptide absorbs UV light, however I don’t believe that you see an obvious peak due to the missing aromatich aas. If it is almost pure, why do you need to load it on column? I think you should consider the infusion of the peptide directly to ms
  • asked a question related to Peptides
Question
1 answer
Hello everyone! Throughout my master's thesis and Ph.D. studies, I have encountered difficulties in achieving optimal clean-up of the UHPLC-ESI-MS/MS system.
My research focus lies in the analysis of peptides derived from plasma samples, and despite implementing an extensive pretreatment procedure aimed at eliminating proteins, particularly albumin, the chromatographic system consistently exhibits issues such as increased noise in the chromatogram and elevated column backpressure. Even the use of a pre-column has not provided a complete resolution to these challenges.
Notably, these problems seem to intensify when my colleagues, particularly those working in the metabolomic field, use the instrument following my experiments. Despite employing a thorough cleaning protocol involving a prolonged gradient elution (lasting approximately 1.5 hours) to ensure the removal of all potential samples residues from the system, the results remain the same.
I would like to know if you have come up with the same problems and if you ahve any insights on potential strategies to overcome these persistent issues. Whether it involves refining the pretreatment procedure, exploring alternative stationary phases, optimizing the cleaning protocol of the system, or considering additional precautions for shared instrument use.
Looking forward to hearing your experiences!
Relevant answer
Answer
Hello Mariano,
It is pretty common to have carry-over analyzing plasma/serum samples. Someone recommended me the use of Trifluoroethanol to wash out all retained peptides. Basically you just use it as a sample and inject 1 or 2ul direct onto the column with a relative short gradient. It does not damage the column in my experience, so maybe you can give it a try in the future.
Good luck!
  • asked a question related to Peptides
Question
3 answers
I have a peptide dissolved in DMSO and MES buffer, and I aim to determine its absorbance using UV-Vis spectroscopy. However, I'm encountering issues with obtaining meaningful results at 220 nm, the specified detection wavelength for peptides. I suspect the problem is related to the absorbance of my blank sample, which is in the range of 2 or 3, potentially due to matrix interference.
Could you please guide how to address and resolve this issue?
Relevant answer
Answer
Spectrophotometers have limits to how high an absorbance they can measure accurately. Depending on the instrument, this may be as low as 1 or as high as 3. If you are struggling to measure the absorbance of your peptide because of high background absorbance, one solution is to use a shorter pathlength. Remember that absorbance is directly proportional to the pathlength of light through the sample. If you are using a standard 1-cm-pathlength cuvette and get an absorbance of 2, if you switch to a 0.2-cm-pathlength cuvette, the absorbance will be 0.4, well within the useful range.
  • asked a question related to Peptides
Question
4 answers
Hi,
i synthesized cyclic peptide including disulfide bond from linear peptide.
However, I could not find a way to check whether the synthesis was successful.
In most cases, it is confirmed by mass spectroscopy(MS/LC), but I don't have access to that method.
How can we distinguish between cyclic and linear peptides except MS/LC?
thank you!
have a nice day !
Relevant answer
Answer
thank you for kind answers !
i will try paper chromatography, kaiser test, NMR/MS, all things in order.
  • asked a question related to Peptides
Question
4 answers
I want to find method or web tools to calculate the pka and helicity of peptide,please help me,thanks!The length of my peptides is 9 and 19.
Relevant answer
Answer
Thank you very much! I will read this paper in detail, thanks for your help! Rob Keller
  • asked a question related to Peptides
Question
2 answers
Hi, everybody?
I have some peptide which have two cystein.
so i want to make cyclized peptide using disulfide bond.
but, i have a concern that multimeric peptide by inter-disulfide bond formation.
how to avoid inter-disulfide bond and proceed only intra-disulfide bond?
thank you very much for your answers !!
Relevant answer
Answer
Thank you for your kind answer.
the article you recommend is great help to me.
have a nice day !!
  • asked a question related to Peptides
Question
1 answer
Hello, I am searching about the antibacterial activity of mersacidin antimicrobial peptide, the majority of papers used of recombinant form of this peptide for investigation of the biological activity of this peptide. Can any person reply to my question, do synthetic mersacidin peptides have biological activity?
thanks
Relevant answer
Answer
Depends if they undergo glycation or not.
  • asked a question related to Peptides
Question
2 answers
I would like to find out what the Italian scientists found in the Covid vaccines
Relevant answer
Answer
I am so glad to spend few time to enrich our knowledge about Covid vaccine
  • asked a question related to Peptides
Question
3 answers
Hello
I have a compound of small peptides(based on reference paper)and I can detect the peptide concentration.
I don't know about details of it.
I see the very good antimicrobial effect of it and based on several papers it depends of peptide.
We have any method to detect total peptide concentration?
  • asked a question related to Peptides
Question
5 answers
Hi every one
I have a peptide library (around 300 peptides) that I would like to dock peptides to its target while I want a high throughput molecular docking webserver or software to do this. Does anyone know any web server or software for this purpose?
Relevant answer
You may try the standalone version of HADDOCK then:
Also, you may try FlexPepDock from Rosseta. Maybe that one is easier for running a whole library
Bests,
  • asked a question related to Peptides
Question
2 answers
what is the best method to extract proteins from serum and tissue sample?
what is the best method to identify the signature protein/peptide between serum and tissue samples by using mass spectrometry?
Relevant answer
Answer
Hi Eef Dirksen,
Thanks for your comment.
I have some serum and tissue samples from patients with a certain disease, and I want to identify signature proteins or peptides that can differentiate them from healthy controls. I am wondering what is the best method to extract and analyze (top down or bottom up) these biomarkers.We have a Q-Exactive mass spectrometer for this experiment. how to handle the data analysis and interpretation? Is there any specific software or tools to process and visualize the results? 
  • asked a question related to Peptides
Question
8 answers
I've been trying to make a solution out of DMF, Fmoc-osu and Glycine. For protection of glycine. I have a hot plate with a magnet and an ultrasonic machine. But nothing is working! I have also been trying to slowly add water to the mixture but Fmoc-osu precipitates as soon as i mix it. And it turns into a big mess. Please help me!
Relevant answer
Answer
DCM is a less polar solvent than DMF. So, it could dissolve Fmoc-Osu.
To glycine, you could try to dissolve in DMF.
Remember that the solubility depends on the quantity of solute and solvent.
Note:
-Consult in the bibliography the solubility of Glycine, Fmoc-OSU, and Fmoc-Gly-OH.
-Do not use water to form Fmoc-Gly-OH.
-Usually, Fmoc-amino acids are soluble in DMF, DMF/DCM, and DCM (in some cases).
-You could use ACN as a reaction solvent.
-Do not use water to form Fmoc-Gly-O-. Later you could use HCl/H2O to protonate Fmoc-Gly-O- to Fmoc-Gly-OH.
Follow the reaction by TLC of HPLC.
  • asked a question related to Peptides
Question
1 answer
Has anyone experienced peptide absorption in the blank plasma when using an ELISA kit? If so, what could be the possible reasons for this problem, and what strategies or techniques were employed to successfully address this issue?
Relevant answer
Answer
I think you're saying you're getting an unacceptably high background reading from your negative control. It's good that you're concerned about this problem. It is common, especially for human blood samples.
If you are using human blood, be sure to take proper bloodborne pathogen precautions, including your vaccinations and PPE.
Consider using serum rather than plasma. And for human samples you should heat the sera to kill viruses.
You should be diluting your samples in blocker solution. For ELISA you probably can start with a 1:1000 dilution and work toward 1:1000000.
You can reduce non-specific binding by adding things to your blocker solution. Starting with a base of PBS, blockers usually contain a protein like casein or albumin at 0.1% (1 mg/mL) and a nonionic detergent like Tween at 0.01%. You can try using a different protein. You can safely increase the protein 10-fold. Depending on the ELISA format, you may want to include 1% control serum in your diluent and blocker as a blocker protein. Increasing the detergent (up to 10x) may decrease your overall signal, but it may increase signal/noise ratio. And sometimes it helps to add 0.1M glycine.
  • asked a question related to Peptides
Question
2 answers
Greetings,
I am here to take an expert help from all the respected researchers and scientists here. I am working on antimicrobial peptide hydrogels. I have designed antimicrobial peptides library and predicted their antibiofilm activity through self-build ML prediction model. My aim was the formation of peptide gel. But I am failed to do so.
For your expert opinion, I would like to mention few important properties of my peptide. Well, it is designed in such a way that it has higher hydrophilicity, it's basic and positively charged. It contains 12 amino acid residues, and it is KLR rich peptide. Theoretical/predicted Isoelectric point is 12.45. So far, I have tried high as well as low pH stimulus. I have also tried freeze thaw cycles. But it is not getting gelatin structure. It is important to mention that I am trying on concentration of 5mg/500ul (also as 10mg/ml).
Now, I am going to try to give ionic pressure by the addition of salts such as MgCl2, CaCl3 etc.
I would like you scientific community to help me in this regard. Ask me questions so that we could have more clarity.
Relevant answer
Answer
Dmitriy Berillo Thank you
  • asked a question related to Peptides
Question
2 answers
How to determine the volume of buffer for unconjugated peptide removal from nanocomposite using a D-tube dialyzer, also How to test check whether the free peptide can be removed by this process or not?
Relevant answer
Answer
Thank you @ Dr. John Carter for the response nd suggestions.
  • asked a question related to Peptides
Question
3 answers
hr
Relevant answer
Answer
For large-volume applications, you may prefer batch-mode purification. Add the prequibilirated bulk resin into the sample and incubate for a while and shake continuously. AAs will adsorb on resin while Chlorides are not.
If the purpose is just analysis, you can derivate the AAs and LLE using organic solvents such as ıso-octane, chloroform, acetonitrile, MTBE, etc may give a chance of both chloride removal and enrichment of the AAs.
  • asked a question related to Peptides
Question
2 answers
Hi all,
I am currently conducting research that involves the use of PatchDock Unfortunately, I have been unable to obtain the necessary documentation and software from the PatchDock team.
If any of you have access to the PatchDock software or its documentation, or if you could provide guidance on where I might be able to obtain it, I would greatly appreciate your assistance. My goal is to establish the module in our HPC system for our ongoing research.
Best
Relevant answer
Answer
Hi Murat,
Yes I tried indeed.
Thank you
Best
Safa
  • asked a question related to Peptides
Question
3 answers
We need to accurately measure the protein/peptide concentration in a sample that is at least 80% DNA (primarily ssDNA less than 80 bases long with some dsDNA also). A majority of the methods I've looked at (Lowry, Bradford, NanoOrange, etc.) will not work with such a high DNA content. We also don't want to add any proteins (like DNAse) to get rid of the DNA because that will affect our measurement of protein contamination.
Any suggestions are welcome. Thanks!
Relevant answer
Answer
You could also consider mass spectrometry. Intact mass spectrometry can detect the mass of all proteins in a sample without fragmentation if the proteins ionize well. Most mass spec systems are setup to detect only positive charges and since DNA is heavily negatively charged, it will be invisible to the detection. Mass spec is very sensitive to low quantities of protein, but the more protein you have the better the detection as there is more to detect.
Bottom up Liquid chromatography with MS/MS and trypsinization can detect the mass of all peptide fragments after trypsinization of peptides and proteins, but deconvolution of what those peptides correspond to is kind of catch22 because you have to know the sequence of the proteins in your sample. Nevertheless, it will detect all peptide fragments.
  • asked a question related to Peptides
Question
2 answers
I have a collection (around 500) of peptide 3D structures (PDB file each 10 residues long and for a given sequence). I need to cluster it based on RMSD values among them. Is there any Python module or any other software which could do that and identify the distinct structural clusters among those 500 files?
Relevant answer
Answer
Vladan Nedelkovski thanks for the reply. However, MDAnalysis takes in only PSF files and not PDF files. I have already computed the distance matrix and based on which I wanted to do the clustering. I have computed an RMSD-based distance matrix between every pair of structures. Still, I couldn't cluster it properly.
  • asked a question related to Peptides
Question
3 answers
Has anyone ever encountered a peptide that binds the wrong way in the protease binding pocket (meaning from c-term to n-term instead of the reverse) and that is still hydrolyzed?
Just curious...
Relevant answer
Answer
The reason I asked is that by docking peptides to protease binding sites this situation often occurs. Without knowing exactly why, I discard these results but if anyone knows better...
  • asked a question related to Peptides
Question
1 answer
I'm researching platelet-derived growth factor signaling, and recently ran into a big problem. Specifically, I am determining whether a particular peptide can activate the PDGF receptor. My initial results using western blot and immunofluorescent microscopy were very promising; compared to negative controls, I saw significant receptor phosphorylation after treatment with recombinant PDGF-BB, a peptide previously shown to activate signaling, or my experimental peptide.
However, my results are suddenly no longer reproducible. I now see no change in receptor phosphorylation or downstream pathway activation between any of the treatment/control groups. I have been troubleshooting for months, and have tried different media, different cell types, fresh reagents, different harvesting methods, and different timepoints. For reference, most of my experiments have been conducted at a PDGF-BB concentration of 100 ng/mL, and peptide concentrations of 1 ug/mL. Cells utilized include THP-1 macrophages differentiated using PMA, and HeLa cells. Timepoints have ranged from 30 minutes post treatment to 24 hours post treatment.
Based on literature, even if my experimental peptide does not activate the PDGF receptor, the PDGF-BB treatment should be a reliable positive control. Has anyone experienced a similar issue in which cells no longer respond to positive controls? Does anyone have any suggestions for conditions to test?
Relevant answer
Answer
If your serum concentration and consistent passage of cells, and the conditions (pH, Temperature) are standard, check if the loss of receptor phosphorylation could be due to increased phosphatase activity. You can try adding phosphatase inhibitors to your lysis buffer to preserve phosphorylation. Hope it would help. Otherwise check carefully all the standard parameters. If still the case use another validation method to verify the quality ingredients and reagents.
All the best.
  • asked a question related to Peptides
Question
4 answers
I am working on peptide containing microsuspension (particle size: 20-30 micron) which has to be sterilised, since it is a peptide molecule, facing challenges with heat sterilisation method. Please recommend suitable method.
Relevant answer
Answer
Since the compound is a peptide, terminal sterilization (autoclaving) at 121C for 15 mins resulted in gelling of the formulation although there is no significant change in purity. Himanshu Bhatt
  • asked a question related to Peptides
Question
2 answers
I have developed an peptide with machine learning that targets receptors vegfr1, vegfr2, and vegfr3. I plan to test its binding specificity on each receptor and measure the downstream effects in a cell line
Relevant answer
Answer
Identifying a cell line that expresses VEGFR3 exclusively or with very low expression of VEGFR2 and VEGFR1 can be challenging, as many cell lines express multiple VEGF receptors. However, here are some approaches to help you identify suitable cell lines:
  1. Cell surface protein expression analysis: Use flow cytometry or fluorescence microscopy to analyze the cell surface expression of VEGFR1, VEGFR2, and VEGFR3 in various cell lines. This will help you identify cell lines with high expression of VEGFR3 and low expression of VEGFR1 and VEGFR2.
  2. qRT-PCR analysis: Perform quantitative reverse transcription polymerase chain reaction (qRT-PCR) to evaluate the mRNA expression levels of VEGFR1, VEGFR2, and VEGFR3 in different cell lines. This will give you an idea about the relative expression levels of these receptors in each cell line.
  3. Database mining: Utilize online databases such as ArrayExpress, Gene Expression Omnibus (GEO), or Cancer Cell Line Encyclopedia (CCLE) to explore gene expression profiles of various cell lines. You can use tools like GEO2R or limma to compare the expression levels of VEGFR1, VEGFR2, and VEGFR3 across different cell lines.
  4. Knockdown or knockout experiments: Consider performing knockdown or knockout experiments using CRISPR-Cas9 or shRNAs to specifically silence VEGFR1 and VEGFR2 in cell lines that express these receptors. This will allow you to evaluate the effect of VEGFR3 expression in the absence of VEGFR1 and VEGFR2.
  5. Use cell lines with known VEGF-A/VEGFR interactions: Some cancer cell lines, such as HUVEC (human umbilical vein endothelial cells) and HMVEC (human microvascular endothelial cells), are known to express high levels of VEGFR3 and have been shown to interact with VEGF-A. You may also consider using cell lines that have been engineered to overexpress VEGFR3.
  6. Screening assays: Develop or utilize existing screening assays that measure the binding affinity of your peptide to VEGFR1, VEGFR2, and VEGFR3. This will enable you to identify cell lines with high binding affinity for your peptide, indicating strong expression of VEGFR3 and potentially low expression of VEGFR1 and VEGFR2.
  7. Combinatorial approaches: Combine the above approaches to increase the confidence in identifying cell lines that meet your criteria. For example, you could first narrow down your options by analyzing gene expression profiles from online databases, followed by validating the expression levels using qRT-PCR or flow cytometry.
  8. Validation with orthogonal methods: Once you have identified potential cell lines, validate their expression profiles using orthogonal methods such as western blotting or immunofluorescence staining. This will provide further confirmation of VEGFR expression levels and help rule out any discrepancies due to technical variations.
  9. Test your peptide: Finally, test the binding specificity of your peptide on the shortlisted cell lines using techniques such as flow cytometry, immunoprecipitation, or surface plasmon resonance. This will help you determine the most suitable cell line(s) for your studies.
Remember that it is crucial to carefully evaluate and validate the expression profiles of VEGFR1, VEGFR2, and VEGFR3 in the selected cell lines to ensure the accuracy and reliability of your results.
  • asked a question related to Peptides
Question
4 answers
Hi I am going to test some peptides as positive control for my ELISA but don't know what sort of concentration to start of with. Any suggestion will be very much appreciated.
Relevant answer
Answer
Thank you very much for all the useful information, very much appreciated.
  • asked a question related to Peptides
Question
3 answers
We synthesized a peptide made up of 7 amino acids. After cleaving the peptide with TFA, we tried to precipitate the peptide from cold ether. However, it seems like our peptide is somehow soluble in ether. Are there any other organic solvents to precipitate peptides?
Relevant answer
Answer
You could lyophilize the mixed solution TFA-ether that contains the peptide.
  • asked a question related to Peptides
Question
5 answers
Dear All,
I am testing out an europium labelled peptide for the ligan binding assay and was hoping to find the Kd through the saturation curve.
Can people explain to me why my saturation curve is a straight line rather than what it should look like.
Thank you
Relevant answer
Answer
Hello! I think there is a biacore T-100 or (3000) at your university. This is good technology and if what are binding is large then it should work quite well. If both of the species are small then maybe not so much. Good luck. Here is the potential lab that has the instrument. https://espace.library.uq.edu.au/view/UQ:722458/UQ722458_OA.pdf
  • asked a question related to Peptides
Question
2 answers
I plan to perform the Fluorescence polarization experiment. I have some FITC peptides ordered from the company with purity > 95%.
My peptide is 15-20 a.a long and very basic with pI ~12.
I read that most people dissolve it in DMSO.
Can I dissolve my FITC peptides in water?
Thank you!
Relevant answer
Answer
You can use a peptide solubility calculator to predict its solubility:
Since it is composed of several charged residues with a pI ~ 12. It should be soluble about 3 pH units from its pH since it is uncharged when pH = pI.
Should be soluble in water or Tris pH ~ 8. Use autoclaved sterile water and filter the peptide.
If the peptide was composed of a lot of nonpolar amino acids then it wouldn't be soluble in water.
  • asked a question related to Peptides
Question
1 answer
Hi everyone. So I have performed MD simulations on desmond of my protein bound to a mutant peptide? My objective is to elaborate how the mutant peptide is inducing conformational changes in the protein. How to answer the above question by looking at the RMSD? How can I explain the given RMSD in detail?
Relevant answer
Answer
Hello Alina
You can begin by looking at the change in structure over time and comparing g it with changes in your RMSD. Where ever there is a drastic jump or dip in the RMSD, you have see what are the changes especially where the mutation was introduced and compare with the wild type.
You can look at the bonding behavior of the mutant and the wild type also.
I hope this helps
  • asked a question related to Peptides
Question
2 answers
The role played by every peptide involved in neuroexocytosis
Relevant answer
Answer
Thanks @Ramirez
  • asked a question related to Peptides
Question
4 answers
I am working on peptide molecule for SC delivery for my thesis work. Which solvents (or co-solvent) shall I use other than PEG, PG, Glycerin to increase the solubility (drug loading) of peptide molecule. Will combination of solvent would be beneficial? Or which surfactant can I add to it?
Relevant answer
Answer
Tris buffer
  • asked a question related to Peptides
Question
8 answers
I am planning to use high pH reversed-phase peptide fractionation kit from pierce (Catalog no.84868) for fractionating labelled peptides. I have used off-gel fractionator (Agilent) to fractionate the samples in the past and the results have not been great. 
Any reviews for the pierce kit based on personal experience that could be of use to me ?
Relevant answer
Answer
Alain Mangé Ok thanks
  • asked a question related to Peptides
Question
1 answer
are there any chemical agents that can stop phase separation of peptide?
Relevant answer
Answer
Phase separation? You mean it goes from water to an organic phase solvent, or vice versa, and you want to stop it? A surfactant should do the trick. You might start with Tween.
  • asked a question related to Peptides
Question
1 answer
I am trying to synthesize the following peptide, on automated peptide synthesiser: dabcyl-RAGGYIFS--edans. The sequence cleaves at dabcyl-GGYIFS-edans. This is what I see from maldi-tof. the peak for the truncated sequence has higher intensity than the required one. I tried using HATU, DIPEA and also PyPOB but the yield did not improve. I use the resin which has edans attached to it already. the HPLC results in poor yields of the desired product. Can anyone help with this peptide synthesis using EDANS-resin?
Relevant answer
Answer
EDANS (via its sulfo-group) can interact with a free amino group of the growing peptide chain (after its Fmoc deprotection) and mask and prevent it from the interaction with an added Fmoc amino acid. We are preparing a publication that discloses this event and suggests an approach for preventing it.
  • asked a question related to Peptides
Question
8 answers
I am looking for companies to order a couple of custom peptides. From my search Biomatik appears to best the best.. value/service. Do you have any experience with this company? 
Relevant answer
Answer
My name is Michael and I work for Biomatik. I highly recommend our custom peptide service for your research needs. Biomatik has been trusted by 10,000+ Scientists Since 2002.
  • asked a question related to Peptides
Question
4 answers
I have trypsin-digested peptides from FACS-sorted samples, but they are contaminated with PEG.
Relevant answer
Answer
Alternatively go back over your sample preparation protocol and determine the source of the PEG contamination.
  • asked a question related to Peptides
Question
3 answers
Hi Friends, could anybody help with this query please? While doing the Bradford Assay I mixed 10 microlitres of my peptide sample with 990 microlitres of Bradford Assay and got the absorbance. So while doing the final calculation I should take into account this dilution and multiply by 100 to get the correct quantity of my peptide sample.
Thanks
Relevant answer
Answer
No. You should have made a standard curve with several concentrations of a protein standard, such as BSA. The standards should also have been added as 10 µl samples. You can therefore compare the absorbance of your sample with the standard curve without calculating a dilution factor for the volume of Bradford reagent added..
  • asked a question related to Peptides
Question
2 answers
I am measuring PET-FCS of a peptide and after measuring for 90 min. I am noticing that there is no PET in the FCS figure. However, if I run the experiment for only 5 min, there is some PET. Also, the diffusion time decreases over time during the measurement. I am assuming that some kind of fragmentation is happening due to the excitation laser.
I will be glad if anyone can explain this fact or any possible theories are also welcomed.
Relevant answer
Answer
It seems your suggestion is right. Laser exposure even at red wavelength (620-670 nm) could induce permanent damage to peptides or other biomolecules, and rate of possible signal degradation should depend on laser power in focal volume and total volume of the sample.
There are 3 of possible effects: 1) peptide fragmentation, 2) permanent peptide conformation switching 3) peptide precipitation/condensation out from solution and seeing FCS signal from impurities in the solution.
  • asked a question related to Peptides
Question
2 answers
Hi, I have been trying to dissolve a very hydrophobic peptide (containing cysteine) with DMF, however, the solution looks very cloudy when I add DMF, and further dilutions with water doesn't help much. Does anyone have any tips on this issue? In addition, I'm use NanoDrop to measure concentration, but when I use DMF as blank, it shows "bad blank". By the way, I'm measuring the concentration at A280. Thanks!
Relevant answer
Answer
I also tried to dissolve the peptide in DMSO and it worked, however, cysteine containing peptides should be dissolved in DMF instead.
  • asked a question related to Peptides
Question
1 answer
Can anyone explain why a study's ELISpot assays might be designed wherein its cells are being plated at a concentration of 200K/well (the standard amount) and then 100K/well? The concentration of the peptide being tested does not change, just the cells. We are somewhat blinded to what the study sponors are looking for, so I don't have the full details, but I struggle to understand what the point of this might be. Would a lesser concentration of cells be informative of the efficacy of the peptides? I would think that playing with the test peptide concentration would be better than that.
Relevant answer
Answer
The number of cells must be adapted to the cell type as well as the frequency of secreting cells namely, the lower the expected frequency of responding cells, the higher the number of cells per well. For instance, if the expected frequency is 1 in 10,000 cells, then you will need approximately 200K cells per well. Conversely, when the expected frequency is high, say 1 in 100 cells, then it will be sufficient to seed 100K cells per well.
Another important factor I would like to mention is that antigen specific responses will require higher cell number (200K) while the use of mitogens may require lower cell number (100K). You also need to know that a minimum number of cells is required for proper cell-to-cell contact and thus for optimal stimulation. For example, if one is analyzing antigen-specific T cells in a PBMC sample, the cell number should be 200K cells per well.
So, cell number would play an important role in ELISpot assays in addition to the test peptide concentration.
Best.
  • asked a question related to Peptides
Question
6 answers
I am trying to dimerize a synthetic peptide (22 amino acids) with a N-terminal cysteine, that was added for this purpose. I use the BM-(PEG)3 crosslinker from Thermo Fisher, which is based on maleimide-thiol chemistry. I reduce the sulfhydryl-bonds using TCEP, add the linker and stop the reaction with DTT. All according to the instructions provided by Thermo Fisher. I check the results with an SDS PAGE, but so far the protein bands stay on the same height before and after the reaction. I tried to get a positive control with insulin, lysozyme and murine SAA, but only the SAA shows a very faint band that could be a dimer.
Has anyone used this linker successfully or has any tips on how to get the reaction working?
Relevant answer
Answer
Yes, your ratio is suboptimal on theoretical grounds. I don’t think there is much wrong with the pH at 7.4, but the reaction will still work at lower pH values. Whatever pH is used, you do need the peptide in molar excess over the maleimide functions to get the desired product in high yield. A better approach than manipulating pH to stop the N-terminal amine from triggering the side reaction is simply to block the amine.
  • asked a question related to Peptides
Question
1 answer
If anyone can speculate as to how AMPs promote inflammation in colitis or how AMPs raise the amount of inflammatory cytokines in colitis, I would appreciate it.
Relevant answer
Answer
  • asked a question related to Peptides
Question
1 answer
I am planning to do folding pattern analysis on the secondary structure of peptide
Relevant answer
Answer
The choice depends on available instrumentation and expertise, and some what less on the peptide structure. If your peptide is big enough, or if it includes some feature to limit structure, then you may observe characteristic alpha and/or beta absorption using CD. A short peptide, with only one beta strand or part of a helix, may not fold stably in the absence of a partner.
I recommend you minimize Cl- ion, as it exhibits high background absorption. I suggest you also predict secondary structure based on primary structure, using any of various computer tools.
  • asked a question related to Peptides
Question
2 answers
I am planning to do folding pattern analysis on the secondary structure of peptide.
Relevant answer
Answer
I agree with Yun-Tzai. Both Raman spectroscopy and circular dichroism are powerful techniques used for analyzing protein folding and secondary structure, but they offer different types of information and have distinct advantages and limitations. I recommend you scientific articles that can illuminate for you the range of possibilities of these techniques:
The choice between the two methods depends on the specific goals and requirements of your peptide folding analysis. Let's compare both techniques:
1. Raman Spectroscopy:
  • provides information about the vibrational and rotational modes of molecules, including peptides and proteins.
  • useful for investigating changes in the conformation and tertiary structure of the peptide backbone and side chains.
  • can be applied to both aqueous and non-aqueous environments, making it versatile for studying different conditions.
  • can be used for real-time monitoring of folding/unfolding processes, allowing for kinetic analysis.
  • requires a relatively simple sample preparation and is less sensitive to the concentration of the sample.
  • can be more complex to interpret compared to CD spectra, and it might require sophisticated data analysis.
2. Circular Dichroism (CD):
  • measures the differential absorption of left- and right-handed circularly polarized light by chiral molecules, providing information on the secondary structure (alpha-helix, beta-sheet, random coil) of peptides and proteins.
  • widely used for studying the folding and conformational changes of biomolecules in solution.
  • relatively easy to interpret, as distinct patterns in the CD spectra correspond to specific secondary structure elements.
  • can be performed under different conditions, including various solvents and temperatures.
  • provides qualitative and quantitative information on the secondary structure content of peptides.
However, CD might not provide as much detailed structural information as Raman spectroscopy. If your primary focus is on obtaining detailed information about the vibrational modes and tertiary structure of your peptide during folding, Raman spectroscopy might be a more suitable choice. On the other hand, if you want to primarily analyze the changes in secondary structure during folding, CD would be the preferred method. In some cases, combining both techniques can provide a more comprehensive view of the folding process.
Good luck
  • asked a question related to Peptides
Question
6 answers
I have an acidic peptide with 12aa (containing 5 hydrophobic amino acids), disulfide bridge, Methionine, and amidation at the C-terminal. I have tried dissolving using acetonitrile, NH4HCO3 0.1 M, and 2.5% ammonia solution separately. However, I can still see some tiny particles in the solution, which is a little turbid.
How can I dissolve the peptide easily?
How can I be sure about the non-dissolved particles? Are they peptides or can they be impurities?
Relevant answer
Answer
Comparing peptide solubility using different solvents the result was as follows:
Ammounia and DMF>DMSO>NH4HCO3
Ammonia 0.1 %
NH4HCO3 0.1 M
  • asked a question related to Peptides
Question
4 answers
I have two synthetic peptides (20-25 AA length) with terminal cysteines that I have been trying to conjugate to maleimide dyes. The maleimide-conjugation protocol has seemed easy enough, but I'm struggling to prove that my peptides have successfully tagged.
I had tried gel electrophoresis with Coomassie Blue, but later found out there was not sufficient peptide in my sample for the bound-tag to show. I then moved to HPLC but my lack of expertise and a lack of appropriate UV detector on the machine I used meant that was unsuccessful. Most recently I had submitted my tagged sample to an analytical service at my university for LCMS analysis, but the results have been inconclusive because there appears to be excessive fragmentation (and no obvious cysteine-tag fragments, or any fragments that differ by an m/z equal to the tag/tag+cysteine etc.).
I feel like this really shouldn't be that difficult, but it's absorbed months of my PhD project and I'm still no closer to proving the success of the maleimide tagging. Any advice?
Thank you.
Relevant answer
Answer
Edward Michelini It's a fluorophore, but I can't seem to find any HPLC machines available to me with an appropriate detector, or anybody with the right expertise in my school. I may have to have another think about doing preparative HPLC, I was getting quite good at the set up!!
Thank you :)
  • asked a question related to Peptides
Question
1 answer
I want to synthesize the peptides in liquid medium by using amino acids, but I don't have idea of synthesis of Pepetide in liquid medium. Please suggest if anyone has any idea about this.
Relevant answer
Answer
Well, of course you can do the usual syntheses also with having your half-built peptide scaffold swimming around instead of having it pinned to a substrate, but it is much harder to do it in a precise way, so probably your yield will decay while a larger fraction will end up as a side product and also you will probably have more in-process losses. In principle the usual coupling agents and protecting groups will still do their job.
  • asked a question related to Peptides
Question
3 answers
I'm running CHARMM36 simulations of a peptide on GROMACS and I need its N-terminus to be capped with an acetyl group. I checked the “aminoacids.n.tdb” file, but there is no entry for acetylated N-terminus. How can I find/make a terminus?
Relevant answer
Answer
Dear all, have you solved the problem? I have the same problem as Harry, could you please clarify how you solve this problem? Thanks much!
  • asked a question related to Peptides
Question
2 answers
Hello colleagues,
I am looking for help from those of you who work in peptide synthesis. I am preparing a peptide containing the following amino acids: HO-Cys(SAcm)- Asp(OtBu)-Pro-Gly-Lys(Boc)-NH2, by HRMS (mobile phase H2O/ACN + 0.1% FA) I observe a signal M=690.3, which corresponds to the desired peptide that lost a tBu protecting group from Asp. However, there is another signal with M=585.3, do you have any ideas of what it can be?
Relevant answer
Answer
Are you 100% sure your sampling peak is homogeneous ?
  • asked a question related to Peptides
Question
2 answers
I need to produce a custom antibody and I need the company to also synthesize the peptide. Does anybody have experience with a company that they liked?
Relevant answer
Answer
Hi Bonnie,
We got you! Send us the peptide or protein sequences and we will make suggestions for your antigen candidate. Once the project is set, we will synthesize the peptide and then use it for immunization.
Our deliverable will include the purified antibody AND the peptide that we use in the antibody production!
Send us a request with your project details to initiate our collaboration: https://www.biomatik.com/services/custom-antibody-service/polyclonal-antibody-production.html
We look forward to hearing from you soon.
Best,
Biomatik Team
  • asked a question related to Peptides
Question
1 answer
Hi all,
I was recently trying to calculate the concentration of amyloid beta concentration from the UV-Vis result. I found that it can be calculated based on absorbance at ~280nm with an extinction coefficient of 1490 M-1 cm-1. However, I did not observe such a peak in my sample at 280nm. Instead, there is a peak at 230nm (which should be the absorbance of the peptide?) I am wondering if I still could do the calculation based on this figure.
Any reply would be appreciated. Many thanks.
Relevant answer
Answer
If your matrix is complex (you have more things other than beta-amyloid in water, as it seems from the spectra) you would need to do a proper calibration first. You can not assume that the only thing that is absorbing at 280 nm is beta-amyloid even if the extinction coefficient is correct. So the short answer is no. Otherwise, if you have available the matrix without the beta-amyloid (this is, all the things in your sample before having the beta-amyloid) you could do a blank spectrum with that and assume you can use the coefficient. As a hard-work answer: you could measure several samples by UV-Vis and by a reference method in order to build a univariate (or multivariate) regression model and use it for then on.
  • asked a question related to Peptides
Question
1 answer
Anyone working on machine learning model or any other approach regarding this?
Relevant answer
Answer
I used a software program called DryLab from Snyder. But I am not sure if this is available any longer.
  • asked a question related to Peptides
Question
1 answer
Hi all,
May I know how to calculate the persistent length of a peptide using MD simulation trajectory ?
Thank you in advance !
Relevant answer
  • asked a question related to Peptides
Question
14 answers
My peptide is Cholecystokinin (CCK8), MW=1142.35 (COOH-D-Y-M-G-W-M-D-F-NH2).
Stock solution in NH4OH 0.05M and working solution in acetonitrile.
I do MS infusion at conc. 500 ng/ml in acetonitrile.
I use two LC/MS machines: Micromass - Quattro Premier XE of Waters (Tamdem Quadrupole) and Applied Biosystems - API 3200 LC/MS/MS (triple quadrupole)
I run ES + but I can not see the peak at 1+, 2+, 3+,4+,...for [M+H], [M+Na], [M+K]
I wonder whether I have missed some other adduct ions that could be created during the ionization?
Or maybe my peptide is being degraded during preparing the sample?
Please give me some advice! Thank you!
Relevant answer
Answer
Here are the other points, you may lean on;
If the purpose is quantification or purity check, LC-UV would be nice to use since the octapeptide you have several aromatic rings and would be highly responsive,
secondly, 500 ppb may be a low conc. to conduct a full scan..especially when the ionization efficiency is low.
Third, I would prefer combined flow scanning in place of infusion...In this mode, you are not taking benefits of the mobile phases which present donors to improve ionization...You should combine the lc flow and infusion (acid and/or DMSO additives in phases for pos ESI in this case) and retest the response...
By solving the peptide in an alkaline condition you are directing the peptide to deprotonation and this makes the peptide more amenable to neg ESI...If it is soluble in ACN directly..prepare your stock in ACN and dilute it with the same solvent, I prefer not to use aggressive pH which is not convenient for most of the peptides to the unintended H exchanges...
Last but not least, If the peptide is hydrophobic and dissolves only in organic solvents this is susceptible to be efficiently ionized in APCI, APPI rather than ESI...You may look for these alternative ionization techniques if MS analysis is the bottleneck and the abovementioned suggestions are useless...
Good Luck...
  • asked a question related to Peptides
Question
6 answers
I have been reading the following article on peptide structure prediction. https://www.biorxiv.org/content/10.1101/2022.02.17.480937v1.abstract I wanted to know whether alpha fold 2 can be used to generate distributions of structure of a given peptide sequence. In other words, Can one generate distributions of backbone Ramachandran angles of a given peptide sequence using structures predicted from various conformations generated by alpha fold2?
  • asked a question related to Peptides
Question
1 answer
I have read protocols for development of antibodies from rabbits post immunization with peptide. However, I shall be the first to do this in our lab. Can anyone suggest how to perform (video/tutorial) the rabbit immunization step by step with precautions?
Relevant answer
Answer
Hi Shubhita,
check the guide from University of South Florida:
I'm keeping my fingers crossed for your success of rabbit immunization.
  • asked a question related to Peptides
Question
1 answer
Hi everyone, I'm carrying out a peptide coupling reaction, with the coupling reagents being N,N'-Diisopropylcarbodiimide (DIC) & hydroxybenzotriazole (HOBt). I'm making glycine to couple with 3-chloro-2-methylaniline in the peptide coupling reaction. I have tried dimethylformamide (DMF), ethyl acetate, dichloromethane and acetronitrile, but glycine wouldn't dissolve in them. Kindly advise suitable solvents for the peptide coupling reaction or any chemicals that I should add to help dissolve the glycine. Thank you!
Relevant answer
Answer
Based on my interpretation of your inquiry, I'd like to know if you employed a base (such as DIPEA or TEA) in the reaction. The reaction would not take place if the base was not there. It's possible that's why your reaction mixture stayed insoluble.
Simply follow the steps below to trigger your reaction.
BOP reagent, TEA & DMF at 70 oC.
  • asked a question related to Peptides
Question
1 answer
Dear all,
I am wondering if there is an easy data format to share the composition of a biochemical solution, for example a buffer consisting of certain concentrations of different substances, as well as some biologically active molecules (peptides, oligonucleotides, proteins, etc). Preferably the latter would have links to their accession numbers in different databases, or if that is not available, a FASTA sequence. I am seeking to improve the re-usability of data within the single molecule field.
Thank you!
Relevant answer
Answer
This question may be partly answered by reference to the STRENDA guidelines (see link below), which strictly apply specifically to enzymology experiments, but the guidelines could be applied to other types of biochemical experiments to make sure they are adequately described.
  • asked a question related to Peptides
Question
1 answer
Dear Colleagues,
How can I synthesize a peptide with N-terminal protection by the benzyloxycarbonyl group (Z-group) using Fmoc/tBu Solid-Phase Peptide Synthesis?
Can Z-Leu-OH be coupled during SPPS as the last N-ter amino acid? The peptide is 5-mer with the sequence: Cbz-Leu-Asp-Lys-Ala-Leu-OH.
Can we protect the N-terminal-CBZ group during peptide cleavage?
I´m unsure if the N-terminal-CBZ group is stable to 95% TFA treatment during peptide cleavage (& side-chain PGs removal) from the Wang resin.
Any reference would be helpful.
Thank you very much for your kind input.
Relevant answer
Answer
You can take two routes:
1) Follow standard Fmoc-SPPS procedure -> remove the N-terminal Fmoc -> protect the free N-terminus with Cbz-Cl (CAS 501-53-1) -> cleave the peptide with TFA -> purify on HPLC
Note: If you take this route, be sure that none of the side chains in the peptide are Fmoc-protected, or else you will get a peptide with Cbz-protected side chains.
2) Follow standard Fmoc-SPPS until the last-but-one residue -> perform the last coupling using a Cbz-protected amino acid (ex. Z-Leu-OH, CAS 2018-66-8) -> cleave with TFA -> purify on HPLC
  • asked a question related to Peptides
Question
1 answer
I like to do SPPS by Fmoc/tBu chemistry to make a peptide (Z-Leu-Lys-Glu-Ala-OH) with an N-terminal-CBZ/Z group at Leu. I like to use Z- Leu-OH for the last coupling, but after the synthesis, I want to keep the Cbz/Z protection group at the N-terminus.
How to preserve the benzyloxycarbonyl (Cbz/Z) group at the N-terminus of the peptide during the removal of side-chain protecting groups with anhydrous TFA?
Relevant answer
Answer
Hi Rajesh, the Cbz group should be stable during your deprotection/cleavage reactions. So you will preserve the Cbz group on the N terminus when you cleave it with TFA (no additional procedures required)
  • asked a question related to Peptides
Question
1 answer
Covid-19 vaccination can induce multiple sclerosis via cross-reactive CD4+ T cells recognizing SARS-CoV-2 spike protein and myelin peptides
Qiu, Y.; Batruch, M.; Naghavian, R.; Jelcic, I.; Vlad, B; Hilty, M.; Ineichen, B.; Wang, J.; Sospedra, M.; Martin, R.. Multiple Sclerosis Journal; 28(3 Supplement):776, 2022. Article in English | EMBASE | ID: covidwho-2138820
Relevant answer
Answer
Here It is the thing you were looking for
  • asked a question related to Peptides
Question
1 answer
I need help with O-Phthaladehyde (OPA) Fluorescent Peptide Assay. I am looking for the proocole or the procedure .
Relevant answer
Answer
Using keywords for your specific peptide and the technique you are considering, try a keyword search using a search engine (e.g. GOOGLE, Bing).
Depending on the sample format you wish to use (i.e. well plates), many kits with all of the reagents are available from many suppliers including Thermo Scientific (Pierce™ Kit) too. Protocols vary depending on which kit and which derivatization agents are used, but the protocols can be found by searching as noted above.
  • asked a question related to Peptides
Question
2 answers
I want to use a RALA peptide vector, and I have found that some studies centrifuge the plasmid/RALA complexes and resuspend the pellet before use, while others seem not to do so. It seems like the studies that don't centrifuge end up with smaller particle sizes than the studies that do centrifuge, but I haven't been able to find any definitive confirmation of this trend. Intuitively, it seems like spinning them at 10k RPM would mash them together to some extent, possibly causing aggregation/melding of the particles and lead to larger particle sizes after resuspension. The only advantage to centrifuging and resuspending I can see is that it would eliminate any toxic effects of free floating/non-encapsulated plasmid, but this wouldn't even really be a concern in vitro, right? Does anybody know of a study that has investigated this? Thanks.
Relevant answer
Answer
Yes, there are a few studies that have investigated the effects of centrifugation on RALA peptide vector complexes. In general, these studies have found that centrifugation can lead to aggregation of the complexes, which can result in larger particle sizes. This is likely because the centrifugal force causes the complexes to collide with each other, which can damage the complexes and cause them to aggregate.
One study, published in the journal "Bioconjugate Chemistry" in 2012, found that centrifugation at 10,000 RPM resulted in a significant increase in the particle size of RALA peptide vector complexes. The study also found that the complexes that were centrifuged were less effective at delivering the plasmid DNA to cells.
Another study, published in the journal "Molecular Pharmaceutics" in 2013, found that centrifugation at 10,000 RPM resulted in a decrease in the transfection efficiency of RALA peptide vector complexes. The study also found that the complexes that were centrifuged were more likely to aggregate.
These studies suggest that centrifugation can have negative effects on the properties of RALA peptide vector complexes. Therefore, it is generally recommended to avoid centrifuging these complexes unless absolutely necessary.
If you do need to centrifuge RALA peptide vector complexes, it is important to use a low centrifugation speed (e.g., 5,000 RPM) and a short centrifugation time (e.g., 5 minutes). You should also avoid resuspending the pellet after centrifugation.
It is also important to note that the effects of centrifugation on RALA peptide vector complexes may vary depending on the specific protocol that is used. Therefore, it is important to experiment with different centrifugation conditions to determine the optimal conditions for your specific application.
  • asked a question related to Peptides
Question
3 answers
Hello all,
I recently started to use peptides for cellular studies. Unfortunately, when I was designing the peptides I did not have a FITC tag to them (the peptide was synthesized by an external company). The peptides still have an -NH2 terminal that I can react with FITC-NHS pretty easily. But due to the small size I would not be able to isolate the peptide from unreacted/hydrolyzed FITC-NHS by MW cutoff as I would for proteins.
The peptides are not very cheap so I would seek alternatives than purchasing more tagged peptides. While I know I could purify it by HPLC followed by concentration / lyophilization, it's adding a lot of procedures to the pipeline and I'm considering if there could be alternatives. And so, I'm wondering if peptides could be precipitated by TCA/acetone precipitation similar to proteins?
While I believe the precipitation is dependent on the peptide structure, I used one of our peptides and tried it out - the precipitation worked (I saw visual white fluffs) and the peptide absorbance curve showed peaks at 214/280. The curve looked identical to the peptide solution (unprecipitated) but with a lower absorbance unit (22 AU compared to 5 AU post-precipitation).
My question is, is this way of isolating peptides legit? And if I used FITC-NHS, would FITC also be precipitated by any chance? Will the peptide lose stability / increase cytotoxicity during TCA exposure? I am not using the peptide for functional studies, just trying to visualize cellular internalization.
- If this way makes no sense I will refer to other methods. Thanks for any suggestions.
Relevant answer
Answer
Success depends a little on the hydrophobicity of your peptide.
I'd try dry Et2O first, esp. if the volume of reaction is small, so all water gets can dissolve in the ether. If it should not work, remove the ether and try isopropanol or ethanol: Begin with small amounts, cool and centrifuge, then increase the alcohol concentration until your labeled peptide precipitates. Fluorescein will precipitate sooner or later, too (pH dependent).
If you try Adam's suggestion, keep all fractions until you know that the operation was successful. For concentrating the peptide, use precipitation as above. There is a high chance that the fluorescein gets trapped on the column and is moving *very* slowly (which is in your favor).
  • asked a question related to Peptides
Question
3 answers
Hello,
I need an advise, I’m using C7C PhD peptide library from NEB and after three rounds of selection and ELISA assay I sent my selected clones to Sanger sequencing. When I obtained the results I found out that the majority of sequenced samples (clones) had the sequence of M13KE vector without the insert of peptide library.
 I wanted to know whether it is a common problem? Are there any steps to overcome this situation? Does it will affect the results of my selection? Why I got so many sequence of M13KE vector?
Sorry for so many questions, but I didn’t find anything about this in the literature.
Thank you in advance for advise.
Relevant answer
Answer
Hi Magdalena, I know this is old, but just wondering how you were able to resolve this issue. I encountered the same problem with the PhD-C7C phage library; majority of my clones were insertless after the third panning, even on the fourth panning. I have increased the stringency and made several modifications on the panning procedures but I still have insertless clones (60-90%). Meanwhile I bought a different library from a different vendor and all the clones have inserts, meaning that the problem is actually from the PhD-C7C library itself.
  • asked a question related to Peptides
Question
2 answers
I am thinking of inserting a small marker peptide into HDR fragments to detect and visualize CRISPR recombinant colonies.
Is this possible?
please guide me.
Relevant answer
Answer
Thank you for your guidance. In fact, my goal is a marker to isolate mutant bacteria from the wild strain after CRISPR recombination.
  • asked a question related to Peptides
Question
1 answer
I wanted to have a rough estimate on time it takes to get the complete thermal distributions of backbone Ramchandran angles of a given peptide size of 18. Is it possible to estimate the time before doing the Monte carlo simulations. In the attached paper they have mentioned it took 108 CPU hours with 30 cores for 200 milliion CPU steps for TRP cage 20 residue protein. With this info is it possible to estimate the wall time of the MC simulations of a given biomolecule ?
Relevant answer
Answer
In my opinion, that time will be sequence dependent, since not all the residues have the same regions available in the Ramachandran angles space. After all, Monte Carlo is a sampling methodology, and the "correct size" of a statistical sample is always system dependent.
When I use MC, I always run several independent samplings (using a different seed number for the random number generator). If the results are statistically equivalent (according to you accuracy criteria, system dependent again), I assume that the individual samplings are correct. In addition, you can average over the results of the independent samplings. I prefer this strategy, with several medium size independent samplings, to one individual very large sampling, where you may have become trapped in metastable states.
  • asked a question related to Peptides
Question
2 answers
We are developing a peptide ELISA for IgM and IgG detection.
We infected mice with parasite 1; for cross-reactivity analyses, we infected mice with two parasites (groups 2 and 3) and included a negative control group. Peptides were synthesized by solid phase peptide synthesis (Fmoc), with ~80% purity, from a native antigenic protein from parasite 1. Sera from different periods of infection were collected and frozen.
We coated the plates with BSA (40μg/mL) for 1 hour, and treated with glutaraldehyde 5% for 1 hour. Then, we added the peptide and incubated it overnight. Finally, we added glycine for 30min. We blocked the plate with PBS-BSA 5% for 1 hour, added the diluted samples and incubated for 1 hour under mild agitation. We added anti-biotinylated IgM or anti-IgG-HRP for 1 hour. For IgM, we added avidin-HRP for 30 minutes. For both, we added TMB for 10min, stopped with sulfuric acid and read at 450nm.
The results are not consistent inter-assays (low reproducibility). The ODs from the negative control group are higher than blank wells and similar to those from parasite 1. Sera from groups 2 and 3 resulted in higher ODs than those of the parasite 1 group. We did not expect this cross-reactivity, given the peptides appeared specific to parasite 1 in certain periods of infection and showed no homology to other parasites or microorganisms on Protein BLAST.
Does anyone have any experience with peptide ELISA that could help us?
We washed the plates after every step, as shown below:
•BSA: washed once with carbonate-bicarbonate buffer (pH 9,6)
•Glutaraldehyde: washed twice with carbonate-bicarbonate buffer (pH 9,6)
•Peptides: washed twice with carbonate-bicarbonate buffer (pH 9,6)
Glycine: washed once with PBS
•IgM
Blocking and sera: washed 3 times with PBS-Tween 20 0,05%
Anti-biotinylated IgM: washed 5 times with PBS-Tween 20 0,05%
Avidin-HRP: washed 7 times, 5 minutes each, with PBS-Tween 20 0,05%
•IgG
Blocking: washed 3 times with PBS-Tween 20 0,05%
Sera: washed 3 times with PBS-Tween 20 0,05%
Secondary antibody: washed 3 times, 5 min each, with PBS-Tween 20 0,05%
Relevant answer
Answer
With glutaraldehyde, you'll crosslink N-termini and lysines with the substrate (BSA). Depending on your peptides' sequences, you might generate adducts which are not recognized by the antibodies you're fishing for, by masking the epitope(s) or generating crosslinking artifacts (crosslinked peptides).
Do you have the option to obtain peptides with only an N- or C-terminal cysteine and use SMCC (maleimide/NHS ester chemistry) for immobilization?
If there are internal cysteines or lysines present, it will be a bit tricky, and you need protection groups during synthesis which are only removed after coupling the peptide to the carrier protein.
If possible, couple the peptides to BSA first and then use this as a stock to coat your plates, to reduce variation between tests.
  • asked a question related to Peptides
Question
4 answers
I was using fragbuilder module in python to generate peptides of sizes 4, 6, and 10. However, the issue with fragbuilder module is that some of the bond angles are deviating from the standard values. For instance, C_alpha--C--N bond angle standard value is 121 degrees but fragbuilder assigns 111 degrees. This angle deviation causes a deviation in the distance between the nearest neighbor C_alpha---C_alpha and its value is 3.721 angstrom and the typical standard value is 3.8 A. Also another bond angle is a deviation from the standard value by 6 degrees which is the C_alpha---C---N whose value is 111.4 degrees and typical standard values are 117 degrees. My doubt is how much deviation is allowed for MD simulations of peptides (or proteins) while fixing the bond lengths and bonds angles ?
Relevant answer
Answer
Gary James Hunter Thanks for you reply.
  • asked a question related to Peptides
Question
3 answers
Hello. For energy minimization of my peptide ligand, I am using Chimera software. Is it necessary to add hydrogen to the ligand prior to docking?
Relevant answer
Answer
It is necessary to add polar hydrogens for the molecular docking process. Because these hydrogens can participate in hydrogen bonding. Some programs automatically add polar hydrogens before docking, but to be sure, enter the polar hydrogens manually.
  • asked a question related to Peptides
Question
3 answers
Example : I have the following sequence, of the penetratin peptide, conjugate with Cf:
RQIKIWFQNRRKWKKNH2.
The synthesis is done m=150mg resin, the resin substitution 0.65mmol/g.
MW=2471.301g/ mol.
I have got as crude after the synthesis m=134 mg. and I purify 20mg, I have got 4.88 mg.
Thank you
Relevant answer
Answer
The crude material contains salts and byproducts (TFA salts, ...).
Calculate the theoretical yield based on resin capacity (0.15g* 0.65mmol/g), peptide molecular weight (add reasonable counter-ions, if necessary).
Then express real yield as a percentage of the theoretical yield. Done.