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
4 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!
  • 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
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
Same thing happened to me recently. I thought it was kind of a glitch and I was the only one. Have you found a reasonable explanation so far? I would be very grateful if you could share it.
Thank you for sharing Peter G Hains
  • 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.
  • asked a question related to Peptides
Question
4 answers
Hello, I want to figure out the degraded product of a peptide (m.w.: >3500)). I did an LC-MS analysis and got a few degraded products (new mass). How can we get the formula or sequence of that degraded peptide from its mass (m/z)? Please share any online tools or materials that will be helpful.
Thank you.
Relevant answer
Answer
If the fragments are made from common amino acids, you can get the determine what amino acids are in a peptide by using their masses, and trying different combinations of amino acid masses until you get a match in your molecular ion peak. You won't get a sequence in this fashion. This is a task best done by computer.
  • asked a question related to Peptides
Question
5 answers
Hello all, first post on ResearchGate!
I have been performing immunoprecipitations with FLAG beads (abcam, ab270704), eluting using SDS loading buffer (with DTT) and boiling. The eluted products are then used for SDS-PAGE and downstream Western Blots. We're concerned our proteins of interest are not being fully eluted from the beads using our current method. Abcam suggests a competitive elution using the DYKDDDDK peptide, however I'm having trouble finding it for purchase from them. Has anyone used that peptide product from another company and had success?
Thanks!
Relevant answer
Answer
If you want to elute your protein from the beads under native conditions, then the FLAG peptide is probably a good way to do it. If you are going to run the protein on an SDS-PAGE gel, then eluting with SDS and DTT and heating should get the job done, since that should denature the anti-FLAG antibody. I doubt that adding the FLAG peptide will enhance the elution any further.
What makes you think the proteins of interest are not being fully eluted?
  • asked a question related to Peptides
Question
1 answer
Hi,
1 - In my last proteomics analysis I obtained peptides with PEP 0 ( zero). should I exclude them because it could not be calculated or it is almost zero and it is sure about the peptide sequences?
2 - I am using DDA to analyze my complex proteome. I am interested in a single protein. I run MQ using a single fasta file with my protein sequence. I obtain peptides with certain PEP. I am re-doing the same analysis but together with the fasta file of the single protein of interest I am adding the whole proteome of the specific organism. I obtain same peptides, with same intensities BUT with different PEP. Why? does anyone have an idea. PEP is not supposed to change and when I run with the whole proteome is actually lower then with a single fasta sequence.
Does anybody have similar issues?
Thanks
Relevant answer
Answer
Hi Andrea
to 1:
In proteomics analysis with MaxQuant, a PEP (posterior error probability) score of 0 indicates that the corresponding peptide has been identified with the highest level of confidence. PEP is a statistical measure that estimates the probability that a given identification is incorrect. A PEP score of 0 means that the probability of an incorrect identification is essentially zero, providing strong evidence for the presence of the identified peptide in the sample. Excluding the PEP zero peptides would be to exclude the best hits in your analysis, which might not be the best way to get accurate results.
to 2:
The size of a database can have a significant impact on the PEP values. Since Maxquant is used in general for shotgun experiments the statistics generating the PEP values are setted exactly for this purpose. I dont think that the calculation of the PEPs generated by a search against one protein only will be plausible.
Best,
Murat
  • asked a question related to Peptides
Question
2 answers
I possess 250 peptides and one target protein, prompting my search for tools/servers that allow docking of multiple peptides simultaneously. While I have already utilized HPPdock, I am interested in exploring alternative options for this purpose. Are there any other tools available?
Relevant answer
Answer
Sangita Dixit You can also try this "CABS-dock web server" i am also sharing the link "http://biocomp.chem.uw.edu.pl/CABSdock/"
  • asked a question related to Peptides
Question
3 answers
I am currently trying to characterize a small peptide that was designed computationally and later synthesized and purchased from a manufacturer. I want to perform Gas Chromatography-Mass Spectrometry in order to confirm its sequence first but unable to find any proper protocol of recent time has delayed my work. Can anyone help me with a protocol? Could be your own, used for a work and published. Thank you in advance.
Relevant answer
Answer
Thank you for the response #Zeyu_Sun. LCMS is definitely the best choice but given the institution I am in has GCMS but no LCMS, I am planning to anyhow carry out a GC analysis and try my best.
  • asked a question related to Peptides
Question
2 answers
Why is tetramer only used to test the interaction between T cell and peptide-MHC complex and not to test binding affinity between peptide and MHC?
Relevant answer
Answer
Major histocompatibility complex (MHC) antigens bind peptides of diverse sequences with high affinity. Both MHC class I and MHC class II molecules contain peptide-binding grooves formed by two α-helices and eight β-strands. In the peptide-binding groove, specific amino acids compose pockets that accommodate the corresponding side chains of the anchor residues of the presented peptides. Peptide-binding preferences exist among different alleles of both of MHC I and MHC II molecules, which are mainly dependent on amino acid polymorphisms in the peptide-binding grooves of MHC chains. They do this in order to generate maximal immunological protection by covering the spectrum of peptides that may be seen by a host over the course of its lifetime.
On the other hand, TCR (T cell receptor) has a low avidity and fast off-rates for MHC-peptide complexes. So, MHC-peptide tetrameric complexes (so-called MHC tetramers) have been introduced for the detection of antigen-specific T cells. MHC tetramers have increased avidity for their cognate TCRs and are successfully used to directly visualize antigen-specific T cells ex vivo. MHC tetramer technology is based on the ability of MHC-peptide complexes to recognize the antigen-specific T cells at a single cell level.
Hope this information is helpful!
Best.
  • asked a question related to Peptides
Question
3 answers
I am trying to separate a small peptide (4 amino acids) synthesized using solid-phase synthesis after cleavage. I have tried dissolving the peptide in diethyl ether, but it did not percipitate from the cleavage reagent. What other methods can I use to effectively percipitate the peptide from the cleavage reagent and resin? Any suggestions or protocols would be greatly appreciated.
Relevant answer
Answer
As I mentioned in my first answer, just evaporate cleavage reagents (TFA/TIS/Water) and your peptide will precipitate. Do not use diethyl ether. As Recarda mentioned, your peptide dissolves in diethyl ether. However, using derivating reactions to modify the peptide makes your work much harder.
To isolate the peptide, you will use chromatography techniques. In my paper, I have used HPLC to isolate my peptides. Please read my paper. I hope it helps.
  • asked a question related to Peptides
Question
2 answers
Hi
My experiment is to look at the intracellular location of a small peptide. I have difficulties when I try to do the IF staining for the small peptide that has been uptaken by cells, and the main problem is the loss of signal after permeabilization.
The peptide is around 6k Da and labeled with Alexa flour 647. It was added to the cell culture media for 10min, then, cells were washed and fixed with 4% PFA, then permeabilized.
I have compared the signals of the live cell, 4% PFA fix-only cells, and fix+permeabilized cells, they all were treated with same peptide the signal loss happened mainly after the permeabilization process, and especially at the cell peripheral part. In the fix and permeabilized cells, the signal of the peptide is only shown in the central part of the cells.
My first thought is, maybe due to low MW, the fixation is not strong or long enough for the peptide, so I have tried different PFA concentration(1%-4%) and time (5min-2h), but increasing PFA concentration and time did not help.
I also tried 0.1%, 0.5%, 1% TX100 and saponin for the permeabilization, it seems saponin is better than TX100, but still have the issue of loss of peptide signal.
I am just wondering if anyone knows the optimized or specific fixation and permeabilization protocol for this kind of application with small peptides. Thanks
Relevant answer
Answer
Hi Arup,
Thank you for your detailed response and valuable information. I have tried methanol fixation, it seems methanol alone didn't work well, I will try methanol/acetone mix again, and will also try digitonin and blocking.
  • asked a question related to Peptides
Question
2 answers
after peptide synthesis then need use enough tfa cooktail to cleave the peptide from the resin. and how to easy separate the peptide and tfa is really bored work. especially when use much tfa.
can someone introduce any adsorbent easily extract the peptide from tfa cooktail ?
Relevant answer
Answer
thank you very much.I now just use Et2O to precipitate the peptide .but now I synthesize much quantity peptide in lab and need use more and more Et2O,and Et2O is very dangerous in lab so Iwant to find some adsorbent to see if can directly adsorb peptide from TFA ? can have some ideal?
  • asked a question related to Peptides
Question
2 answers
We are facing problem while doing cleavage. As we get final crude after using many combination of TIS Water & TFA and other was TFA TIS Phenol & DTT still not getting good results as impurity besides main peak was showing 7-13% and increases with increase in scale (determined by analytical hplc). Mass determination indicating it as +56 or 57 impurity determined tbu either not getting cleaved properly or getting reattached somewhere on amino acid sequence. But impurity fraction (the varying peak just beside main peak). The assumption of getting reattachment of tbu is correct or its some other impurity? And if yes then which it is.
Relevant answer
Answer
Could you give us some more information on your synthesis strategy? Which amino acids are protected with OtBu?
  • asked a question related to Peptides
Question
1 answer
I am trying to solubilize a peptide:oligonucleotide conjugate for intra-peritoneal injection into a mouse. At low concentrations, the mixture dissolves nicely in either H2O or saline, however at higher concentrations, everything crashes out. The peptide is hydrophobic but both the peptide and oligonucleotide are currently dissolved at very high (stock) concentrations in HBSS or TE.
I just tried Ethanol (10 and 50%), DMSO (10 and 50%), PEG-400 (10 and 50%) and Glycerol (50%). The saline and glycerol generally keep most of the protein:olgo conjugate in solution, but the others generate large solid pellets in the bottom of the tube after 30 minutes at room temperature.
Can anyone advise on other possible solvents to try?
Relevant answer
Answer
I'm a bit surprised that DMSO does not work. Have you tried adding DMSO to the water?
You also could try adding polyamines (e.g. spermidine or shorter poly-lysines/poly-arginines/poly-ornithines) to compensate for the negative charges of the DNA. For more sophisticated approaches, and some background information, please check this brochure: https://media.iris-biotech.de/flyers/IF20_2_Polymer_Therapeutics.pdf
  • asked a question related to Peptides
Question
7 answers
Dear all,
I need help now. I run HPLC with one of the peptide, on RP with 0.1% TFA/ACN as mobi phases. There is an impurity peak came out. The weird thing about this peak is 1) did not show in my blank and system suitability standard. 2) peak area/height is not changing when sample is diluted. 3)did not show up in the dilution buffer inject (10x more), 4) this peak give the same area if I double the injection, though my peptide peak is doubled.
what could be the cause of this peak? Anybody have experience of this lipid problem in HPLC? I only have VWD detection.
thanks
Relevant answer
Answer
Based on the information provided, it is possible that the impurity peak is caused by a secondary reaction that occurred during peptide synthesis or due to post-translational modifications of the peptide. The impurity peak may be structurally similar to the peptide and could be co-eluting with the peptide peak.
It is also possible that the impurity peak is caused by a minor component in the sample matrix that is not detected in the blank or dilution buffer.
To further investigate the cause of the impurity peak, you could try the following:
1. Change the mobile phase: Try changing the mobile phase or adjusting the gradient to see if the impurity peak disappears or shifts. This could help determine if the impurity peak is related to the mobile phase composition.
2. Try a different column: If the impurity peak persists, try a different column to see if it is specific to the column you are currently using.
3. Analyze the impurity: Collect the impurity peak and try to isolate and identify the impurity using techniques such as mass spectrometry or NMR spectroscopy.
4. Try a different detection method: If possible, try using a different detection method such as UV-Vis or fluorescence to see if the impurity peak is detected by other methods.
Regarding the possibility of the impurity peak being caused by lipids, it is possible, but it would be unlikely to cause a peak that is not affected by dilution or changes in injection volume. Lipid contamination typically causes peak tailing, and adding an organic modifier to the mobile phase, as previously mentioned, can help to address this issue.
  • asked a question related to Peptides
Question
1 answer
Can a boiled peptide serve as a negative control in biological assays? or does it necessarily have to be a scrambled peptide of the same length?
Relevant answer
Answer
A boiled peptide can serve as a negative control in some biological assays, but it may not be appropriate for all assays.
Boiling a peptide can denature its structure and disrupt its function, rendering it inactive. This can be useful as a negative control in some assays where the activity of the peptide is being specifically tested, such as in enzyme assays or receptor binding assays.
However, in other assays, such as assays testing for cellular uptake or transport, a boiled peptide may not be an appropriate negative control. In these cases, a scrambled peptide of the same length is often used as a negative control. The scrambled peptide should have a similar amino acid composition to the active peptide but with a randomized sequence to ensure that any observed effects are not due to a specific sequence motif.
In general, the choice of negative control will depend on the specific assay and the nature of the peptide being tested. It is important to carefully consider the appropriate negative control for each assay to ensure that any observed effects are not due to artifacts or nonspecific interactions.
  • asked a question related to Peptides
Question
3 answers
I am expressing a fusion protein with a 6 HIS tagged eGFP SUMO followed by a small peptide. Because of the small size (~ 560 Da) of the peptide after cleavage from fusion, it is hard to visualize on any gel. I doubt that there is any cleavage happening at all. I wanted to know if expressing the fusion tag without any linkers between two proteins (eGFP and SUMO) makes it unrecognizable to the SUMO protease to cleave?
Relevant answer
Answer
If there is a lab in your area that can perform intact protein mass spectrometry, you can send them a sample with and without SUMO protease cleavage to see if the mass of the protein changed. The difference of 560 kDa should be easily resolved by that method.
  • asked a question related to Peptides
Question
7 answers
I am conducting a molecular docking study using peptides as ligands. I successfully ran the docking process using autodock vina, and when I viewed it using Discovery Studio, I realized that the ligand have missing bonds. I retracked my process and found out that the pdbqt file of my ligand is the problem. Here is a screenshot of my pdbqt file of my peptide, IF in DS. Attached also is the pdbqt file of my peptide.
Please help me on what to do.
Relevant answer
Answer
Jack Dela Cruz Great.... Happy to hear..
Regards
  • asked a question related to Peptides
Question
4 answers
I am developing a UHPLC method for content and purity for a peptide based on 100mM KPF6 and 100mM NH4PF6 pH 3.5. TFA as an ion pair reagent does not give the required separation performance. My question is less about method development HPLC for peptides, but more about the negative effects of KPF6/NH4PF6 in the UHPLC systems used (Waters IClass binary system and Thermo Vanquish Horizon binary system).
The negative effects on 3 instruments were already noticeable after 1 to 2 sequences (extreme pressure fluctuations):
The repairs carried out showed more or less the same damage:
- strongly porous seals in the pump heads
- damage to the pistons
- defective inlet/outlet check valves
One system was used over 2-3 months and showed the most damage.
I will ask my questions right at the beginning:
- What experience do you have in using KPF6, NH4PF6 as additive/buffer in mobile phases for UHPLC analysis or for method development for peptides?
- How can these reagents be used "safely" and robustly without causing damage to the pumps?
These chaotropic reagents seem to be highly corrosive. I would be glad to get some useful advice. Many thanks in advance for your support and advice.
Kind regards
Ronald
Mobile phases are:
Mobile phase A 0.1M KPF6 pH 3.5 / ACN 8:2 (V/V)
Mobile phase B 0.1M KPF6 pH3.5 / ACN 35:65 (V/V)
and
Mobile phase A 0.1M NH4PF6 pH 3.5 / ACN 8:2 (V/V)
Mobile Phase B 0.1M NH4PF6 pH 3.5 / ACN 35:65 (V/V)
Preparation:
0.3M Buffer KPF6 pH 3.5
  • 55.2 g KPF6, were weighed into a 2000 mL beaker. 1000 mL water was added and stirred until complete dissolution for 15 min. Sonification for approx. 2 min. It was adjusted to pH 3.5 with Orthophosphoric acid 85% (+/- 255 µL) and stirred well. The buffer was filtered with a Steritop-GP 1000 mL Express Plus PES 0.22 µm into a 1000 mL bottle.
Mobile phase A1: 0.1M KPF6 pH 3.5 / ACN 8:2 (V/V)
  • 330 mL 0.3M_ KPF6 buffer pH 3.5, 470 mL water and 200 mL acetonitrile was transferred into a 1000 mL bottle and mixed well. Sonnification for approx.5. min.
Mobile phase B1: 0.1M KPF6 pH 3.5 / ACN 35:65 (V/V)
  • 330 mL 0.3M_ KPF6 buffer pH 3.5, 20 mL water and 650 mL acetonitrile was transferred into a 1000 mL bottle and mixed well. Sonnification for approx.5. min.
0.3M Buffer NH4PF6 pH 3.5
  • 48.9 g NH4PF6, were weighed into a 2000 mL beaker. 1000 mL water were added and stirred until complete dissolution for 5 min. It was adjusted to pH 3.5 with 25 % NH4OH (+/- 340 µL) and stirred well. The buffer was filtered with a Steritop-GP 1000 mL Express Plus PES 0.22 µm into a 1000 mL bottle.
Mobile phase A1: 0.1M NH4PF6 pH 3.5 / ACN 8:2 (V/V)
  • 330 mL 0.3M_ NH4PF6 buffer pH 3.5, 470 mL water and 200 mL acetonitrile was transferred into a 1000 mL bottle and mixed well. Sonnification for approx.5. min.
Mobile phase B1: 0.1M NH4PF6 pH 3.5 / ACN 35:65 (V/V)
  • 330 mL 0.3M_ NH4PF6 buffer pH 3.5, 20 mL water and 650 mL acetonitrile was transferred into a 1000 mL bottle and mixed well. Sonnification for approx.5. min.
After sequence, the systems were flushed with
  • 85min with ACN/H2O 15/85; 0.4 mL/min
  • 15 min with ACN/H2O 75/25; 0.4 mL/min
  • 15 min with ACN/H2O 50/50; 0.4 mL/min
  • following a low flow ACN/H2O 50/50; 0.1 mL/min
Relevant answer
Answer
Dear William Letter Thanks a lot for bringing this up. The instruments were serviced and repaired by their vendors. As thong as the degasser work fine, I think they are not being considered. I will keep this point in mind. Thanks.
  • asked a question related to Peptides
Question
5 answers
If we have Peptide sequence, can we develop correct DNA sequence from that? If Yes, what can be most suitable process? What can be the pros and cons?
Your answer will highly be appreciated.
Many thanks!
Relevant answer
Answer
If it's an organism that has had at least some of its genome sequenced, try using BLAST. a "tBLASTn" search will compare the protein sequence to translated nucleotides.
Good luck!
  • asked a question related to Peptides
Question
4 answers
I am testing different combinations of peptides grafted to alginate hydrogels. Inside these hydrogels I have culture human mesenchymal stem cells and now I want to see how the different combinations of peptide influence the cell behavior. For that, I would like to understand whether cell volume changes as well as cell major length, sphericity or other measurement that can describe cell geometry. I've been trying to use Amira-avizo to measure diameter/length (approximated from a 2D disc) of cells that have been 3D reconstructed. I already managed to get all these values but they do not correlate very well with the 3D reconstructions that I've obtained from my different conditions. Is there anyone ho know how to use this Amira for this purpose for this purpose or have suggestions for other software ?
Relevant answer
Answer
This video explain how can measure in amira-avizo the diameter of pores
  • asked a question related to Peptides
Question
1 answer
we have synthesized 27Mer peptide which contains 8 glycine amino acid. and due to inefficient coupling one of the glycine coupling was partially completed giving us the peak of desglycine peptide.
The retention time in HPLC for the both is same. we have tried different methods , buffers and organic modifiers.
if anyone has encountered a same problem and solved it please let me know.
Relevant answer
Answer
If you're using microwave assisted peptide synthesizer then I must recommend to increase the coupling reaction time after each successive glycine coupling or you can just let the sps resin aggitated under nitrogen bubbling condition with the reactant amino acid (glycine) solution for 10 to 15 minutes before applying microwave power and temperature of your actual coupling step so that you can ensure the maximum coupling extent...I have found this one quite useful during long peptide synthesis....