Science topic

Proteins - Science topic

Explore the latest questions and answers in Proteins, and find Proteins experts.
Questions related to Proteins
  • asked a question related to Proteins
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 Proteins
Question
5 answers
Good afternoon,
I try to extract cell wall proteins from green macroalgae, I have tried different protocols and one of them included step of EGTA extraction, however this fraction is jelly like, so I expect polysaccharides from cell wall to be present. When loaded to SDS-PAGE, it was obvious that the gel pores were blocked and no bends were obtained, just light blue streak. According to Bradford, in this fraction I have quite a lot of proteins with which I would like to do Western Blot... I have already tried aceton precipitation, but still a lot of polysaccharides remained....So please do you have any recommendations how to get rid of these polysaccharides?
Thank you very much,
Tereza
Relevant answer
Answer
I met the same problem. Did you solve it?
  • asked a question related to Proteins
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 Proteins
Question
2 answers
I want to stain muscle cells for presence of glycoproteins. For this I am trying to do lectin staining. However, right now I do not have a working protocol and I have come across 2 lectins in literature that can bind N-glycans (glycoproteins i am interested in). I want to know which of these two could be a better choice? And if anyone has done lectin staining before and can suggest me a protocol or a product? I am ok to use a fluorescence conjugate lectin or a regular one with the use of secondary antibody biding.
Relevant answer
Answer
Thank You Ana.
  • asked a question related to Proteins
Question
5 answers
Hi
I'm purifying some mutants of the protein I study. The wild type protein exists as a monomer and is 28kDa.
I have two mutants (same protein, same number of amino acids but with 8 amino acid substitutions at defined positions), one of the mutants (mutant 1) analysed using size exclusion chromatography with multi-angle static light scattering (SEC-MALS) and its MW was shown to be 33kDa and has an oligomerization state of 1.2. The other mutant, mutant 2, also measured by SEC-MALS was 59kDa with an oligomerization state of 2.2.
For the wild type to measure the concentration I've just been using the MW (28kDa) and extinction coefficient (calculated by entering the sequence into online software ProtParam) and using a NanoDrop measuring absorbance at 280. This gives the concentration in mg/ml which I then convert to molar concentration.
For the mutants I want to measure their concentration the same way - measuring A280 on the NanoDrop using the mutants MW and extinction coefficient and calculating molarity from mg/ml. I'm not sure if this is an obvious/stupid question but what MW weight and extinction coefficient would you use for the mutants on the NanoDrop? E.g. For example mutant 2 molecular weight (MW) of the protein based on its amino acids (AA) composition is predicted to be 28kDa, but SEC-MALS shows it is 59kDa as the protein forms an oligomer.
My instant is to use 59kDa and the computed extinction coefficient predicted from the AA composition - is this correct?
Thanks in advance!
Relevant answer
Answer
Thanks for your reply very helpful - so if I’ve understood you properly - it is incorrect to use the SEC-MALS molecular weight when measuring UV 280 absorbance to determine protein concentration of the mutants as the extinction coefficient is based on the protein being denatured. Rather I should use the predicted Mw just based on the amino acid composition.
I agree the deviation from whole numbers points to an oligomer mixture. I want to add these recombinantly purified mutants to permeabilised cells to observe their localisation (they are fluorescent proteins) so knowing concentration accurately is important for these experiments.
So for example for mutant 2 whose Mw just based on amino acid composition is 28kDa - if A280 on the nanodrop gives a concentration of 10 mg/ml which would be 0.35mM, the concentration protein is 0.17mM? (as the oligomerizaiton state is 2 taken the closest integer value). I should say the ability of these mutants to form oligomers is because the mutations increase their aggregation propensities.
Am I correct in this? Is the above way accurate or would something like the Bradford assay be better?
  • asked a question related to Proteins
Question
4 answers
Hello, I hope analytical chemistry people or biochemistry major fellows could help me. Kindly advise on how to prepare the following denaturation buffer: [2% SDS, 1 M β-mercaptoethanol (β-ME)]?
The buffer is expected to be used with Endoglycosidase H enzyme, extracted from Streptomyces plicatus, CAS 52769-51-4 | 324717.
Relevant answer
Answer
2-mercaptoethanol (or β-mercaptoethanol) is a liquid with a concentration of 14.2 M. It is very smelly, so use it in a fume hood, if possible.
Sodium dodecyl sulfate is a solid. The dust from SDS can irritant the lungs, so it's a good idea to wear a filter mask when weighing it.
2% in this context means 2 grams/100 ml of solution.
For 100 ml of the solution, you need 2 grams of SDS and 7 ml of β-mercaptoethanol (1 M/14.2 M x 100 ml). To somewhat less than 90 ml of distilled water in a beaker, stirred with a magnetic stirrer, add the β-mercaptoethanol and dissolve the SDS. Bring the volume to 100 ml with distilled water.
  • asked a question related to Proteins
Question
5 answers
Earlier I had used Serva IPG Strips in which it was easy to decipher the gel side of the strips but recently we shifted to Bio Rad and here we are encountering this problem. Your valuable suggestions are required.
Relevant answer
Answer
Thanks a lot Abhishek Singh
  • asked a question related to Proteins
Question
6 answers
I have extensively searched google scholar but I am struggling to find any groups who have previously used Rosetta to conduct ab-initio structure modelling of single-pass or membrane anchored proteins and I'm specifically not talking about homology modelling just ab-initio.
Please let me know if you have read any papers or know anyone who has done this,
thanks.
2nd year PhD student at University of Liverpool.
Relevant answer
Answer
Waqas Abbasi, Hi- No Rosetta-Membrane did not work well at all for this task and I would not waste your time attempting to do so, unless you have preliminary signs indicating positively for your protein (or your membrane anchored protein has some specific other attributes meaning it may work better). I would suggest firstly you thoroughly reading Anne Marie Honegger's post above and the link/paper they kindly provided there though.
I would strongly suggest you instead just try and use the new generation methods RosettaFold and AlphaFold2 as they seem to be able to position single/double TMHs away from the membrane-associated globular regions/domains of the chains better, albeit still fa from perfect. However, there does seem to be some signs in the near-ish future similar methods may be released better for such instances of TMH-anchored membrane proteins, but it remains to be seen.
Best of luck,
David
  • asked a question related to Proteins
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 Proteins
Question
4 answers
Hi all,
Just wondering if anyone has experienced similiar issues when running SDS-PAGE gels? The ladder stops running straight toward to bottom of the gel? Ponceau staining of the nitrocellulose membrane showed some lanes had expanded while others had shrunk (don't have a picture of this unfortunately). I could see my target protein however the bands wern't uniform. Anyone have any advice?
Some helpful info -
I pour my own gels using standard recipe (16%)
Buffers are always made fresh (although not PHd)
Ladder is from ThermoFisher, catalogue #26616
Just let me know if any other information would be helpful!
Thankyou!
Relevant answer
Answer
"Smiling" on gels (upward curvature of the bands near the edge of the gel) is probably caused by uneven heating. Reeducing the voltage may help, as will avoiding using the edge lanes.
Narrowing and widening of lanes is caused by differences in ionic strength (and maybe pH) between adjacent wells. This can be avoided by making sure all the samples have similar ionic strengths, or by precipitating the protein in the samples to prepare the gel samples.
  • asked a question related to Proteins
Question
3 answers
My stock protein concentration is 2.13mg/ml. I have to have a final concentration of 0.83 nanomoles and my final volume for the protein would be 50 microlitres. The molecular weight of the protein is 55kDa. I did a few calculations based on this and I got a volume of protein to add to my experiment, but it is impossible to measure it using pipettes. Do I have to dilute the protein more to get to a measurable volume for my final concentration?
Relevant answer
Answer
Haimantika Dey just be sure that as Adam B Shapiro pointed out you are needing 0.83nM (and not 0.83 nanomoles in 50ul volume.
  • asked a question related to Proteins
Question
5 answers
Does anyone know an accurate technique to quantify intracellular protein concentration besides ELISA? I need a reliable and highly quantitative method to measure protein concentration to gather data from cell culture experiments and incorporate them into my model of protein-protein interactions.
Relevant answer
Answer
if the protein expression in blood monocytes were low,
but when I measured the levels of this protein in serum by ELISA were high?
what is the appropriate explanation?
  • asked a question related to Proteins
Question
6 answers
Hi, I need to check microbial growth on wastes rich in proteins that tends to ferment pretty quickly. To get an idea of what and how much it's growing, I thought about using Plate Count agar, YS or YM for screening for yeasts, maybe Violet Red Bile for gram negative. What can I use to check protease activity? Since it's a mixed colture, I cannot use super selective media.
Could this course of action work for a totally uknown mixed colture? The goal is to identify the microrganisms responsible of spoilage and fermentation.
Thanks in advance for the help.
Relevant answer
Answer
Screen? Screen for what attribute?
  • asked a question related to Proteins
Question
6 answers
Hi!
I need to prove that my protein is localized on the cell-surface but not inside the cell in soluble form.
How can I make it?
There is an opinion to wash cells, to label all cell-surface proteins with NHS-biotin, and to collect labelled fraction of cell-surface proteins with streptavidin magnetic beads. After that I can make western blot with specific Ab.
Maybe there is another way?
Protein and cell lines are human.
Relevant answer
Answer
Chad M. Petit Can you share protocol to detect cell surface protein without permeabilization?
  • asked a question related to Proteins
Question
5 answers
Does anyone have experience with Casein kinase 1, I am working with its homolog in yeast (Yck). Is it regulated or just a constitutively active protein? 
I checked its expression levels, they don't change in response to different carbon source. But I am trying to determine if its activity is modulated by phosphorylation or not.
Relevant answer
Answer
Hello
Would recommend this research article :
Kind regards
  • asked a question related to Proteins
Question
6 answers
I am trying to detect the secretion of certain tagged soluble protein following transfection in 293T cells. These proteins are epitope tagged and can be visualized via Western Blotting. Therefore, I plan on using the supernatant to run a western blot to determine whether these proteins are secreted.
I am torn between TCA precipitation, ultracentrifugation, or just taking the supernatant and running it on a protein gel.
For context, these transfections are done in 6 well plates and in Optimem.
Does anyone have any suggestions as what the best way to detect these secreted proteins is?
Thank you!
Relevant answer
Answer
Although you have already received good suggestions. But still, if you need a detailed explanation follow this link it has quite a good suggestion.
  • asked a question related to Proteins
Question
5 answers
Recently I run a stain-free gel (Bio-Rad) to separate my protein of interest and do total protein normalization. After separation, I imaged the gel on ChemiDoc. Unfortunately, I overexposed the gel. Please could you advise on how to do total protein normalization in the setting of overexposed gel? I have attached the image of the gel for your reference.
Relevant answer
Answer
I couldn't open your archive because I don't have or know any scn extension free software to use. Could you send the image as an ordinary image file?
Anyway, about your question, you may try a few filters at your ChemiDoc equipment, maybe there, you can find a better resolution to use and then try to normalize it. Otherwise, best otion would be to do another gel and acquire it correctly.
If you can, send the ordinary iamge for me to know how overexposed it is. Ok?
Hope it helps.
Best wishes!
  • asked a question related to Proteins
Question
3 answers
The ones we have tried are either against the signalling peptide or do not recognise small levels of expression. Or show only one band in Western analyses.
Relevant answer
Answer
Hello,
I think I can recommend the CaSR antibody from CUSABIO, which was verified in ELISA, WB, IHC and tested in Human, Mouse, Rat. You can find more details on this page: https://www.cusabio.com/Polyclonal-Antibody/Calcium-Sensing-Receptor--Antibody-11083532.html
  • asked a question related to Proteins
Question
11 answers
I've conjugated a PEG4-maleimide (MW 613.66) onto an antibody fragment (52 kDa), but I'm trying to figure out how this affects A260/280 measurements on a Nanodrop afterwards. After conjugation and removal of unconjugated material on a PD-10 column, I measure the concentration of the eluted aliquots but the measurements seem really off, and the A260/A280 ratio is nowhere close to 0.5. I blanked the Nanodrop with the PD-10 elution buffer. Seems strange that such a small molecule could affect the readings that much. Does anyone have experience with PEG and if/how it could affect concentration measurements after conjugation?
Relevant answer
Answer
Cynthia,
Please contact me by email at
[email protected]. I will try to "talk you through" the method of using the refractive index and UV absorption to calculate the protein and PEG concentrations without attempting a BCA assay or other chemical assay of the protein concentration. Our method is based on the Kunitani reference that I cited in this string of discussions in February 2015.
Best wishes,
Merry
  • asked a question related to Proteins
Question
4 answers
Hello everyone!
So I have a peptide with ~14 kDa molecular weight.
It contains one Cysteine in the middle of the sequence.
I tried to desulfurize this Cysteine.
But how do I know if it worked or not? I use UPLC with ESI-MS, H2O/ ACN.
Since the mass diference is very small, I suspect that mass spectra of product will not be much different from the starting material.
Retention time should also be very similar, at least I can't see the second peak. Or perhaps conversion is 100% and there won't be a second peak, I don't know :)
Any suggestions?
Relevant answer
Answer
Is the purpose of the separation simply to check whether the desulfurization worked? If so, then you could try to measure the thiol before and after desulfurization treatment. For example, you could react the mixture with fluorescein-maleimide. The protein will become fluorescent if the thiol is still present. To quantify, you can measure the absorbance of the attached fluorescein and calculate a protein (A280):dye (A495) ratio.
  • asked a question related to Proteins
Question
2 answers
Hello,
I have a system with two proteins. I want to compute the energies of these proteins separately. So, I create two separate groups, say, Prot-1 and Prot-2, and add these to my rerun mdp file as energygrps. However, when I perform gmx energy on the rerun.edr file, any energy term associated with Prot-2 returns 0. Prot-1 returns energy values for the total protein system (i.e. for both proteins). I am not sure what is going wrong here. Any suggestion is highly appreciated.
Best,
Saumyak
Relevant answer
Answer
Hello, the most practical way to do this analysis is:
1) in index.ndp create the two groups separately
2) in *.mdp add the two groups to compute the energy
3) use rerum and you will get a new ener.edr
4) use gmx energy and select correct ener.edr together with index.ndp
  • asked a question related to Proteins
Question
5 answers
I have done SMD of protein at applying constant velocity using NAMD software and CHARMM ff. Since, this is my first time in performing Steered MD, I am not sure as to should I do umbrella sampling alongwith SMD? Are the results of SMD without any umbrella sampling significant ? It would be helpful if I could get some references as well.
Relevant answer
Answer
Sharmi Mazumder could you figure out how to pull and fix multiple atoms in smd ? i'm interested to know about the same.
  • asked a question related to Proteins
Question
4 answers
Andree Hubber (2004) described effector-coding genes with signal aa for both T3SS and T4SS.
Relevant answer
Answer
Some interesting results may be in support of this idea: Wang, Y., Yang, F., Zhu, PF. et al. Use of the rhizobial type III effector gene nopP to improve Agrobacterium rhizogenes-mediated transformation of Lotus japonicus. Plant Methods 17, 66 (2021). https://doi.org/10.1186/s13007-021-00764-z
  • asked a question related to Proteins
Question
8 answers
Kindly discuss your ideas and viewpoints on the origin of life and the RNA world hypothesis.
What are the contradictory views on why researchers are still unsure about the origin of life through RNA or such analogous molecular intermediate pre-cursors preceding its existence?
"The general notion of an “RNA World” is that, in the early development of life on the Earth, genetic continuity was assured by the replication of RNA and genetically encoded proteins were not involved as catalysts. There is now strong evidence indicating that an RNA World did indeed exist before DNA- and protein-based life. However, arguments regarding whether life on Earth began with RNA are more tenuous. It might be imagined that all of the components of RNA were available in some prebiotic pool and that these components assembled into replicating, evolving polynucleotides without the prior existence of any evolved macromolecules. A thorough consideration of this “RNA-first” view of the origin of life must reconcile concerns regarding the intractable mixtures that are obtained in experiments designed to simulate the chemistry of the primitive Earth. Perhaps these concerns will eventually be resolved, and recent experimental findings provide some reason for optimism. However, the problem of the origin of the RNA World is far from being solved, and it is fruitful to consider the alternative possibility that RNA was preceded by some other replicating, evolving molecule, just as DNA and proteins were preceded by RNA." - Robertson and Joyce
[This is as per the explanation by Michael P Robertson and Gerald F Joyce in the article: "The origins of the RNA world." published in the Cold Spring Harb. Perspect. Biol. 4, a003608 (2012).]
The scientific community must resolve this contradicting conjecture through rational discussion and debate backed by strong experimental evidence on what must be the pre-cursor molecule to the Origin of Life if it is not RNA!
Relevant answer
Answer
One of the issues that is holding the concept of an RNA world back from being more scientifically useful - irrespective of whether there ever was such a thing - is that we don't use the idea in the scientific way it was intended. Just like any other prebiotic scenario, it is not (nor has it ever been) a scientific hypothesis. In fact, scenarios are usually not intended as such. Scenario authors from all niches (including RNA world) have pointed out that scenarios themselves are untestable. However, they guide thinking and allow to conceive of hypotheses that are testable. If we go through the old literature we find very explicit passages to support this fact.
For the specific authors advanced in the question, G.F. Joyce and L.E. Orgel, we have a passage from 1999 in "prospects for understanding the origins of the RNA world". (The RNA World 2nd ed. 49-77).
"The presumed RNA World should be viewed as a milestone, a plateau in the early history of life on earth. So too, the concept of an RNA World has been a milestone in the scientific study of life's origins. Although this concept does not explain how life originated, it has helped to guide scientific thinking and has served to focus experimental efforts."
You can find this point of view expressed in foundational work for all niches related to the popular scenarios today. But you can also find it for scenarios most people in origins have never heard of. E.g. the idea that celllular life started with terpenoids found in G. Ourisson and Y. Nakatomi's "the terpenoid theory of the origin of cellular life: the evolution of terpenoids to cholesterol. (1994) Chem & Biol. 1 11-23".
"The hypothesis provides an attractive way of ordering the terpenoids: like all evolutionary theories, it cannot be tested directly. The ideas summarized here do, however, suggest a multitude of experiments having some bearing on the fundamental and fascinating question: how did the first cells appear? We hope to carry out some of them."
A related line of thought - but highly influential - is the exposition by Harold J. Morowitz from 1992 in his book "Beginnings of Cellular Life: Metabolism Recapitulates Biogenesis". If we go to the conclusion, we find this explicit clarification on the distinction between a genuine scientific theory and a scenario:
"at this stage of the thought process, it is important to focus on the hypothesis that intermediary metabolism recapitulates prebiotic chemical evolution. This hypothesis is not a strictly vulnerable theory in the Popperian sense, but it does provide us with a valuable heuristic method for using modern knowledge of biochemistry to search for events that have left their trace. If the intermediary metabolism of autotrophs does not recapitulate biogenesis, then the discontinuities will have to be explained."
More than 2 decades back, many authors made a clear distinction regarding this nuance. Scenarios are here to help: they guide thinking and design experiments. They only guide thinking in a scientifically meaningful direction as long as we can easily abondon scenarios and enthusiastically continue replacing them with new, more informed scenarios. A situation where a scenario gets entrenched and where researchers treat it as a scientific hypothesis is - by construction - hard to escape.
In fact, this is exactly the situation that many researchers have described around the 80s, when criticism mounted against the prebiotic broth scenario. The passage from Wächtershäuser's 1988 "Theory of a Surface Metabolism" is telling:
"The prebiotic broth theory has received devastating criticism for being logically paradoxical (11, 135), incompatible with thermodynamics (11, 144, 160), chemically and geochemically implausible (134, 136, 144), discontinuous with biology and biochemistry (160), and experimentally refuted (135, 160). The reason for the tenacity with which it is retained as accepted dogma has been forcefully and clearly stated by Scherer (126): "If this rejection is substantiated, there will remain no scientifically valid model of the selforganization of the first living cells on earth."
Clearly, the broth scenario had overstayed its welcome. One reason for this is that its 'claims' (which for a scenario can only be speculations) were too much in contradiction with claims from fields of science that do not suffer the same restrictions when it comes to testing and refuting their theories. One example of a very controversial idea that can be found in Haldane's formulation of a broth scenario, is the purported necessity of a long, highly functional protein randomly emerging from a soup, as an extremely rare event: we expect this to be prohibitively unlikely and hence a far from parsimonious explanation.
Quite a few of the critiques voiced against the prebiotic broth scenario are equally valid critiques of some scenarios we have today, including RNA world.
The RNA world is an old and multifaceted concept. There are contrasting formulations that make different claims (to be interpreted as speculations) about history. As with the prebiotic broth scenario (and any scenario), it has raised genuine scientific objections. These have remained largely unadressed, in spite of its long dominance.
It is instructive to bear in mind that scenarios don't come from nowhere. They're fairly detailed speculations about purported historical events. To make them, each author makes assumptions. Some of these concern speculations that later became testable, e.g. about chemistry and physics. You will find different scenario authors make different assumptions and different arguments (and flaws therein). There's an inevitable bias here with respect to the fields an author is trained in. Some of the foundational assumptions in popular scenarios like RNA world are at least 50 years old, but some unchallenged assumptions date back to a literature that is more than a century old. A time before IUPAC, modern quantum mechanics, genetics, and so forth.
That has been enough time to forget that scenarios like RNA world are by construction not testable hypotheses and that they were not intended as such. Scenarios are here to guide thinking, to inspire experiments. The best thing a scenario can do for us, is generate insights that spur us to change the way we think and thereby necessitate replacing our old scenarios with new ones, and repeat the cycle. The science coming out of the community today is a lot more conducive to doing that than previously.
The same cannot be said for the rather myopic RNA-centric framing of a question in the cited passage, which attempts to elevate RNA world to more than a scenario. Rather than forcing ourselves to think about the rather narrow and outdated proposal by Joyce and Robertson, ("consider the alternative possibility that RNA was preceded by some other replicating, evolving molecule"), it is more productive to critically revisit all the things that have been assumed and argued when the concept of an RNA world was conceived and how which of these premises are considered valid or plausible today, and which ones back then. Is there a formulation of RNA world for abiogenesis that is logically sufficient? And if so is it logically necessary that abiogenesis proceeded this way?
It is also instructive to check how much of the logic was sound. e.g. the rhetorical tricks employed in RNA world introduce all sorts of hidden assumptionsm.
As an example of the latter: some still justify an RNA world by the party trick 'chicken-and-egg' question 'protein or RNA, which came first?', only to conclude with 'RNA, it encodes proteins' and hastily conclude with an even stronger 'RNA-first' for abiogenesis. 'chicken-and-egg' fallacies are nothing new in origins. In fact, they were already identified as such long ago. E.g. in chapter 8 of "Seven Clues to the Origin of Life (1985)" by Cairns-Smith, there's an illustrated passage detailing that these types of paradoxes in origins frame the question in a manner that prevent us from considering scaffolds.
"
The fact is that even the so-called simple organisms such as E. coli are very complex enterprises with all sorts of things going on together. There is plenty of scope for accidental discoveries of effective new combinations of subsystems. It seems inevitable that every so often an older way of doing things will be displaced by a newer way that depends on a new set of subsystems. It is then that seemingly paradoxical collaborations may come about.
To see how, consider this very simplified model - an arch of stones: This might seem to be a paradoxical structure if you had been told that it arose from a succession of small modifications, that it had been built one stone at a time.
scaffolds that starts like this:
This might seem to be a paradoxical structure if you had been told that it arose from a succession of small modifications, that it had been built one stone at a time. How can you build any kind of arch gradually? The answer is with a supporting scaffolding. In this case you might have used a scaffolding of stones. First you would build a wall, one stone at a time:
Then you would remove stones to leave the 'paradoxical' structure.
"
It should be noted that in 2022, even in RNA-world, very few scholars remain that find RNA-first a convincing idea. As a scenario, however, it is not useless: it is instructive to consider what the underlying ideas are that at some point in time made such a highly specific idea compelling to so many of us.
A fixed motif in scenario papers is to start explicitly and implicitly assuming a few things about what chemistry can and cannot do and some properties of abiogenesis. These sort of assumptions used to be spelled out routinely, also outside scenario papers. Let me give two examples.
The original 1953 paper for the "Frank Model" "on spontaneous asymmetric synthesis", has the passages
".. the defining property of a living entity the ability to reproduce its own kind ...
confining attention to chemical molecules, the complexity of any having this essential property of life is likely to be great enough to make it highly improbable that it has a centre of symmetry."
(*I should point out that Frank makes an important error here: the capacity for molecular reproduction is not a molecular property but a property of a reaction network. If we add an additional thermodynamic criterion this property is autocatalysis and we can then check this claim from the IUPAC definition: https://goldbook.iupac.org/terms/view/C00876. It turns out there are trivial ways to make small networks that have this property https://chemrxiv.org/engage/api-gateway/chemrxiv/assets/orp/resource/item/60c74d67469df42226f44295/original/emergent-autocat-animation.gif.)
The point to retain here is that Frank considers it to be generally accepted that one can assume this property to be prohibitively rare in chemistry. This belief was wideheld, and we can e.g. read in "the units of selection" (1970) by Lewontin a summary on scientific views on abiogenesis
"The present view ... Since there was no autocatalysis, there was no reproduction or heredity and so no possibility of natural selection."
The coacervates in Oparins scenario were notably invoked to adress this issue.
When it comes to assumptions in scenarios, this systematically involved conjecturing that chemistry 'in the wild' intrinsically and deterministically becomes a 'mess', undergoing no meaningful complexification, and for which no reproduction and evolution can reasonably be expected. From there, it appears that no process of abiogenesis should conceivably occur naturally, and thereafter some specific sequence of exceptional events is proposed as plausible, because it appears to be the sole contender.
Let us make more explicit why this is not an innocent procedure:
We still find our understanding of 'basic chemistry' to be plagued with limitations and long-lived misinterpretations (e.g. 2 days ago we learned that methyl substitution destabilizes radicals instead of the textbook knowledge that it stabilizes them ).
Moving beyond the basics, we by and large lack a lot of formal theory, experiment, or even a simple reference frame for the things that happen then. Joyce and Robertson honor the tradition of purporting from the outset that 'chemistry in the wild' becomes an intractable mess. The issue is that we don't know at all if that's the case. We cannot assume this from the outset, we need to extensively study it. We require extensive experiments and theory and a reference frame for all the phenomenology associatied with complex systems (e.g. multiple components, compartments, multiple forms of nonequilibrium driving, length scales, time scales).
In making the routine assumption of 'messy, intractable chemistry that can neither complexify nor multiply', we have decided in advance that, once we finally understand 'chemistry in the wild' with its 'so-called intractible mixtures', it cannot have any bearing on abiogenesis. Let alone explain it.
That is a disproportionately bold conjecture about fundamental science, and a very consequential one: all historical scenarios - RNA world being one out of many - have been justified by formulating conjectures of this sort (many authors also insist on other properties, e.g. chemistry being deterministic). Clearly, it should be the first priority of everyone in the field to test this conjecture, by extensively and rigorously studying complex chemical systems as an end in itself. If the conjecture is correct, it provides an important validation for historical scenario approaches. If the conjecture turn out wrong, we are in a much better position to conceive of more scientifically informed scenarios, but potentially the approach will change entirely.
In presenting it as such, I am making it appear as if it could be an open question whether the chemical conjectures underpinning our scenarios in origins may be true or not. In fact, we have learned quite a few things in the meantime. And some clumsy mistakes were made elsewhere.
- Determinism:
When it comes to chemistry being deterministic (a key tennet of e.g. Sutherlands scenario and Wächtershäusers surface metabolism): upon critical evaluation of what is known of basic chemistry this idea becomes unacceptable, especially when considering the chemical processes on the surface of a planet, as opposed to a quick reaction in pyrex.
1) insofar as it is reproducible, modern chemistry owes much of it to big strides of standardization in glassware, methodology, synthesis protocols (e.g. usage of stirring bars).
2) lab chemistry exhibits many forms of contingency. This is particularly the case when it comes to phase behavior, e.g. habit modification, polymorphism. Aspirin purportedly has 8 reported polymorphs, phenobarbitone 13.
3) glassware is cleaned between reactions, thereby making successive reactions in the same glassware independent. In nature, this property of independence is absent. In fact, effort to make an evolutionary classification of minerals are rooted in the opposite: that certain minerals start to form conditional on the presence of certain others. (https://pubs.geoscienceworld.org/msa/ammin/article/104/6/810/570840/An-evolutionary-system-of-mineralogy-Proposal-for)
- Autocatalysis:
A first issue to get out of the way is the misconception that autocatalysis is prohibitively rare. A prominent PI in origins (RNA world, not a chemist) told me that chemists throughout history have found exactly one example. Claims about the contents of a literature one cannot realistically have read in a lifetime is a common error we can find in the origins literature. Below are some reviews.
I should stress that these reviews discuss examples from a few niches in chemistry. These reviews do at least allow to have 100s of counterexamples to dubious claims about no autocatalysis in chemistry, but it's only a small fraction. Virtually all branches of chemistry have regular reports of autocatalysis, but very few focus on autocatalysis in its own right. And hence most branches do not review their reported examples.
By critically examining the IUPAC definitions, one can show that autocatalysis is dramatially more widespread than long thought. In part, this is because the definition applies to a wealth of situations where the term is not routinely employed. By examinging the requirements of autocatalysis as an emergent network property, one can demonstrate that this property emerges particularly readily in a heterogeneous / multicompartment context. With the disclaimer that I'm an author I refer to the following:
- Messy chemistry:
Refreshing counterexamples are afforded by the literature on systems chemistry and dynamic combinatorial libraries.
In the context of origins, a recent work that is greatly aiding in fixing our misconceptions is : https://www.nature.com/articles/s41557-022-00956-7
Where a reaction of purported immense complexity is found to exhibit highly reproducible and ordered behavior as function of environmental inputs. How chemistry exactly works on this level is still poorly understood. I think I do, but it'll have to await peer review. But we cannot in good scientific conscience take for granted anymore that chemistry becomes messy and intractable. When we do the experiments, we see something very different.
in conclusion, I want to come back to the final point of the question
"The scientific community must resolve this contradicting conjecture through rational discussion and debate backed by strong experimental evidence on what must be the pre-cursor molecule to the Origin of Life if it is not RNA!"
No. The sientific community should strive to do what it can justify scientifically. Those that find it fruitful to relegate the RNA world - which is not a hypothesis - are justified in doing so. Notably because it is is founded on scientifically refuted premises and logical errors.
Those that find ways to make it fruitful to keep it are justified in doing so: it's a scenario, one can draw inspiration from it. Perhaps a thoroughly altered version can be developed that fixes previous issues.
Above all else, RNA is an amazing molecule that has been used for fundamental research that concerns everyone in origins, and will continue to do so irrespective of how serious the RNA world scenario is still taken.
What the origins of life community needs, first and foremost, however, is concern itself with more important matters.
Complex chemistry needs to be studied thoroughly on an experimental and theoretical level.
New scenarios are needed. And these scenarios should no longer require chemistry to have properties it doesn't have, and vice versa. These scenarios should also explicitly be appraciated for what they are, an for what they're not. They're here to help, to guide thinking, inspire experiments, produce testable predictions, update our beliefs. They are not scientific hypotheses in and of themselves.
  • asked a question related to Proteins
Question
1 answer
I am just a new student doing research about carbonic anhydrase, I found so many papers classify their CA protein into alpha, beta, and gamma classes, but they didn't say the exact method they used to classify them. I guess when they once found a new CA, they did sequence alignment with different classes CA sequence. But I am not sure which CA sequence they aligned with, and what the signature of the alignment that makes the new CA just this class type. Could you give me some hints or papers that illustrate the classification method?
Relevant answer
Answer
"At least five distinct CA families are recognized: α, β, γ, δ and ζ. These families have no significant amino acid sequence similarity and in most cases are thought to be an example of convergent evolution."
This article and the literature cited therein will answer your question.
  • asked a question related to Proteins
Question
3 answers
Long story short, I need to degrade 30ug of RNA and i need to do it at 4C. I want to use only as much RNAse A as necessary.
So if performing the reaction at 4C, how long of incubation time and how much RNAse A would i need to degrade 30ug of RNA?
For example, would 1ug of RNAse A(20ug/ml conc.) for 15min at 4C be enough?
Relevant answer
Answer
You will probably have to determine this yourself.
Setup a few reactions (with and without RNase A and RNase A at different concentrations). Run RNA out on a TBE urea gel to monitor amount of hydrolysis.
  • asked a question related to Proteins
Question
5 answers
I need to perform a WB on such proteins
Relevant answer
Answer
Partha Nagchowdhuri, I know this post was long ago, but did you ever figure out how to extract the protein from those brains?
  • asked a question related to Proteins
Question
2 answers
I would like to perform, among other things, electrophoresis with preserved proteins (not denatured).
To do so, I need to first extract proteins with a "soft" buffer.
The best would have been M-TER, but I was wondering if B-TER would do the job (because that's what I have in the lab).
(In order to increase the yield of proteins extraction, I planned to perform griding through Precellys and sonication with a probe.)
Thank you for your help.
Jean
Relevant answer
Answer
Hi Noel,
I don't think that you will have the same extraction efficiency for your stratum corneum samples with B-PER buffer since this buffer is optimized to extract proteins from bacteria. I would recommend to use M-PER buffer since it is optimized to mildly extract proteins from mammalian samples.
Best,
Murat
  • asked a question related to Proteins
Question
5 answers
Can anyone suggest a method to quantify proteins in melanin containing samples?
Relevant answer
Answer
Please refer to the research article given below.
Highlight:
Using several assay methods, synthetic eumelanin prepared by autooxidation of L-beta-(3,4-dihydroxyphenyl) alanine and a natural melanin isolated from dog hair melanosomes were tested in model experiments to assess their possible interference in protein determination. The degree of interference was assessed by comparing the data obtained with the melanin samples with those derived from measurements of bovine serum albumin. In the common biuret and Lowry methods melanin interferes by falsely increasing the values obtained; the addition of Folin reagent only after melanin removal, as suggested by Doezema, decreased but did not eliminate melanin interference. Methods working at acid pH such as those according to Salo and Honkavaara with Ponceau S or Sedmak and Grossberg or Spector using Coomassie blue G-250 proved much better.
Although melanins adsorbed a small amount of dye from the reaction systems in these procedures, their sensitivity to proteins makes the melanin interference negligible. Such procedures can therefore be recommended for protein determination in the presence of melanin.
References:
Sedmak and Grossberg.
Spector
Best.
  • asked a question related to Proteins
Question
3 answers
Hi all,
I was wondering if anyone knows-
which statistical test I should use in order to find whether a sample is an outlier in my proteomic data? It's obvious when looking at the PCA, but how should one calculate this?
Many thanks for your help!
Relevant answer
Answer
The standard for excluding a single data point is typically 95% (or 98%) confidence that it is beyond the standard deviation of the other cohort samples. The problem with most LC-MS/MS studies is that the standard deviationis so wide (because n is so low) that almost everything is within 95% confidence. PCA analysis is merely suggestive of what has the most variation between the cohorts. It is rarely statistically-valid at a given confidence level and always requires focused experimental follow up with more single biomarker-focused validation effort. When this is done, all the PCA targets typically disappear as aberrations. The basic issue is that biomarker concentrations cover a range of values for each cohort, these ranges typically overlap and you ultimately need to perform an ROC analysis to identify a useful biomarker, that requires 100's of patient samples.
  • asked a question related to Proteins
Question
5 answers
I am looking for a specific method I read years ago,, but cant find it right now,,
it was basically incubating the protein with some concentration of non-polar molecules,, have them attach to the protein hydrophobic patches, and i think they got a crystal structure or something,,
Any chance anyone recalls something similar???
Relevant answer
Answer
In fluorescence spectroscopy studies, ANS is recommended as “hydrophobic probe” for examining the non polar character of proteins.For example, for non polar surface patches of proteins, you can see the paper from (Cardamone and Puri, 1992), and many more.
M. Cardamone, N. Puri
Spectrofluorimetric assessment of the surface hydrophobicity of proteins
Biochem. J., 282 (1992), pp. 589-593
  • asked a question related to Proteins
Question
3 answers
Hello,
I am studying proteins bound to large (~500 nm) nanoparticles. I just want to know if there are any special recommendations for sample prep, to get a good CD signal, and whether near UV-CD or far UV-CD is better suited for this, considering the scattering effects I have to overcome.
Thanks in advance!
Relevant answer
Answer
Far UV-CD needs lower concentration, optical path length than near UV-CD. Also far UV CD region for proteins have higher molar attenuation coefficient than near region. Far UV-CD region is appropriate for analysis of secondary structure.Near UV-CD is informative about tertiary structure. For proteins bound to large (~500 nm) nanoparticles. ATR-FTIR analysis of amide I spectrum is useful for evaluating secondary structure. Also in ATR- FTIR expriment, scattering effect is not limiting factor and this technique is appropriate for nanoparticle surface analysis.
  • asked a question related to Proteins
Question
4 answers
Hello,
Our old but not to old DynaPro Plate reader I does not work anymore and Wyatt does not want to investigate the problem as the instrument is 10 years old. We have the money to pay them but they really do not want to loose time on it...
We would like to know if some of you know DLS instruments that are compatible with the measurement of several conditions (at least 30 conditions) in parallel. Of course the goal is to find a company that is able to do a maintenance.
Best,
Sébastien
Relevant answer
Answer
Protein aggregation over a range of temperatures (protein denaturation) is a common application for the plate reader. The wells can take tiny amounts of fluid. Have you spoken to Malvern SA?
Bâtiment Le Phénix 1, 24 Rue Émile Baudot, Palaiseau, 91120 France
Tel: Sales: + 33 1 69 35 18 08
Tel: Support: + 33 1 6935 1806
Fax: + 33 1 6019 1326
  • asked a question related to Proteins
Question
3 answers
Hello!
The bacterium that I am working with is a motile bacteria. I constructed over-expression plasmid constructs into the bacterium. I want to check is there any effect of overexpression of the proteins on bacterial motility. Can anyone suggest some reference papers?
  • asked a question related to Proteins
Question
4 answers
the assay was done using TMBZ solution which was made by dissolving 0.01 g of TMBZ in 5 ml of methanol and 15 ml of 0.25 M sodium acetate buffer (mixing sodium acetate and glacial acetic acid) (pH 5.0) and finally adding PBS and 3% Hydrogen peroxide. 
Relevant answer
Answer
Can anyone send me the protocol for making the standard curve of Cytochrome C from horse heart? I have faced some problems in making the concentration of 0.0025nmol to 0.02nmol of Cytochrome C from horse heart.
  • asked a question related to Proteins
Question
6 answers
Hi everyone,
I am about to define an experiment where we want to investigate 10 - 20 de novo small proteins. We are mainly interested in affinity but also want to show that proteins are folding properly. For that we are thinking about using circular dichroism. I am having seconds thoughts though if this is the right method in the long run. When it comes to publishing, I have the gut feeling that reviewers might ask for a crystal structure of the protein or even the complex. I am working on getting an impression myself by reading nature and science papers but I would like to get to know your advice and experience concerning the matter. What methods are best suited to give our research credibility that might be expected in high impact journals?
cheers
Martin
Relevant answer
Answer
If your protein fulfills a measurable function (e.g., enzymatic activity), then you can take the presence of that activity as proof of folding. To measure the stability of that structure, I'd try differential scanning calorimetry (DSC), which measures the change in heat capacity during (un)folding. Integration then gives the average change of enthalpy ΔH between two temperatures. Proteins unfold (and ideally refold) cooperatively over a narrow temperature or [denaturant] range.
If you have several related proteins, you can use the protein engineering method (10.1351/pac199163020187) to associate ΔΔG with sequence changes.
It is also possible to plot the rates of (un)folding as function of temperature and/or [denaturant] (chevron-plot, 10.1016/j.ymeth.2004.03.013), stopped-flow CD would be nice for that.
A transverse [denaturant] gradient can be used to measure unfolding by electrophoresis (10.1016/0022-2836(79)90279-1), amide deuterium exchange is used to measure their accessibility by ESI-MS or NMR. Some bound fluorescent dyes (e.g., ANS) change their intensity during unfolding, sometimes this can be done also with intrinsic Trp residues.
  • asked a question related to Proteins
Question
4 answers
Dear all,
I'm looking for a commercial partner who is able to determine log P (partition coefficient) of the protein with the use of bioinformatics tools. The protein mass is 2,4 kDa, it includes 22 amino acids and three S-S bridges.
Thank you in advance.
Relevant answer
Answer
you can follow this article
Wang H, Feng L, Webb GI, Kurgan L, Song J, Lin D. Critical evaluation of bioinformatics tools for the prediction of protein crystallization propensity [published correction appears in Brief Bioinform. 2017 Nov 1;18(6):1092]. Brief Bioinform. 2018;19(5):838-852. doi:10.1093/bib/bbx018
  • asked a question related to Proteins
Question
2 answers
Hello everyone,
I have been working with TNBS(2,4,6-Trinitrobenzenesulfonic acid, proteins detection) for three years and I'm wondering if TNBS has any expiration date?
I couldn't find anything on the box or Gbioscience's website which I purchased from.
I have doubts my TNBS doesn't work well anymore.
Thank you
Relevant answer
Answer
If you are talking about the aqueous solution, one company states 16 months at -20°C.
  • asked a question related to Proteins
Question
4 answers
All extant protein alignment algorithms that I am aware of perform poorly for complex proteins that have many possible isoforms. There are better ways of making these assignments. Do you know of a new approach?
  • asked a question related to Proteins
Question
7 answers
Hi everyone,
I have the problem in endogenous nuclease contamination in my protein preparation purified from E. coli BL21(DE3). I would like to ask your experts in avoiding or removing such contamination. Can you suggest me the protocols or an alternative E. coli host strain with tagged-nucleases?
Thank you in advance.  
Relevant answer
Answer
Hi,
I also have same problem with Taq polymerase and reverse transcriptse. I need to remove endonuclease and I have tried UnoQ, NuviaQ and Heparin column chromatography. But trace quantity of endonuclease is still present..kindly also suggest for method of endonuclease detection..do we have any kit...
  • asked a question related to Proteins
Question
4 answers
Hello,
I would like to find whether two proteins have a common interaction path in the graph using the STRING database. There is software that is doing it?
Relevant answer
Answer
This can be done in STRING itself: go to "Search" → "Multiple Proteins" tab → enter the proteins and confirm → (optional but recommended) adjust the interaction sources and minimal required score in "Settings" tab → expand the network by clicking "+ More" button as many times as needed to find a connection.
  • asked a question related to Proteins
Question
4 answers
Hello everyone!
I'm new to protein analysis and I would like to know what kind of instruments and software can be used to determine protein molecular weights as well as the sequence of a protein. Do you have any literature suggestions?
Thank you!
Relevant answer
Answer
Hi Ioanna,
You can use PAGE for appx. molecular weight of a protein , but you need a mass spectrometry for the exact mass. Also, you need a high resolution mass spec (MS/MS) for the peptide sequencing and posttranslational modifications on your protein. You should also know the sequence of the protein if you are interested in a single one OR you can download proteome databases from Uniprot for multiple proteins. Otherwise, you can try de novo sequencing.
  • asked a question related to Proteins
Question
36 answers
Demographers estimate that by 2050, the number of people on Earth will reach 10 billion. With such a number of people, the agricultural economy, logistics of food supplies and people's eating habits will have to change. It is likely that economics will force these processes, which will result in the transition of the majority of humanity to nutrition mainly based on vegetable and vegetarian diets. Meat production is many times more expensive than the production of cereals, fruits and vegetables. In addition, according to scientific research and the theory of futurologists, the production of traditional meat, e.g. pork and beef, may be replaced by the production of protein from insect breeding. Research shows that there are more proteins in the bodies of insects than in traditional meat dishes. In addition, the logistics of food supplies, agri-food products will have to improve. Systems for matching agricultural and reptile production to the current needs of the industry and the nutritional needs of people will be improved so as to reduce the scale of food wastage. The biggest threat to the implementation of this plan may be unexpected atmospheric phenomena, natural disasters, droughts, hurricanes, tropical heat in the areas in which agricultural crops have been cultivated so far. In addition, industrial exploitation of arable land and climate change causes soil depletion and the disintegration of areas suitable for agricultural production. Therefore, it will be necessary to continue the technological progress in the production of crops, in biotechnology, in the creation of new plant varieties resistant to pests and adverse climatic changes.
Please, answer, comments. I invite you to the discussion.
Relevant answer
Answer
With rising income food consumption patterns also change. Calorie intakes of poor and rich people are surprisingly similar, but rich people consume more protein. This adds about a further 1 percent growth to food demand which means that the world will need to produce approximately two percent more food annually if today’s poor become rich. The growth of supply needed for the future about 2 percent annually has to come mainly from available farmland to avoid an overly negative impact on fragile ecosystems. This requires finance, investments, innovation, and knowledge to improve the yields at existing farmlands. The yield gap between what’s needed and what’s being produced is still very high. On the other hand, reducing food waste can have a significant impact on the availability of food. Reducing food waste can improve the efficiency of food value chains and help to distribute food more evenly to those in need.
  • asked a question related to Proteins
Question
3 answers
I have 3 mutant proteins I have designed, cloned, sequenced, expressed and purified. Now I sent the proteins off for mass spec analysis just a tryptic digest as this was predicted to generate fragments adequate to confirm mutations. I got my results back today and have found that 2 of the 3 have wild-type residues instead of mutant. I am confused as the DNA sequencing following cloning was correct how can my protein have reverted back? I use the term native loosely as the native in this case was just the mutant I started my project with. Any ideas are greatly appreciated!
Relevant answer
Answer
If the MS service can do intact protein mass spectrometry, you can supply them with the intact purified proteins for mass measurement. As long as the mutations change the mass of the protein measurably, you should be able to tell whether the mutation was made (although not where it is).
  • asked a question related to Proteins
Question
10 answers
I want to preface this that I am a PhD student working on a bacterial genetics project and am in no way an expert in protein structures - I'm just messing around with this to see if it can provide any interesting insights that I may have missed just looking at sequence and homologs! Anyway, I am working with a protein that has no defined crystal structure, but we know which ligand it primarily binds. I was able to generate some predicted structures using I-TASSER, but since the ligand I'm interested in is not in the ligand-protein binding database it didn't predict any binding sites for it (though it did find how related ligands would bind which is great!). Does anyone know of (free) software in which you can provide a protein sequence/structure and a ligand name/structure and it would predict how they would likely bind? It seems like something that should exist, but I can't find it! Thanks :)
Relevant answer
Answer
As Shin Murakami and Iqra Aroob said, you have to go step-by-step: First build one or more homology models for your protein, then dock your ligand to the model. For a very knowledge-based approach, I would use SwissModel https://swissmodel.expasy.org , doing the "search for templates" first and carefully examine the list of potential templates, preferring those that contain ligands related to yours to ensure that potential ligand binding pockets your model are in open conformation. By following the links to UniProt in the result list you can find out whether a potential template has the same function as your protein, increasing the probability that the binding site is conserved between the two.
When overlaying the model structure with the original templates, you can check whether your ligand would be able to bind to the same pocket in a similar way by fitting your ligand to the ligand in the template.
You can when try docking into this pocket using SwissDock http://www.swissdock.ch
Swiss-model and Swiss-dock are associated with the originators of SwissProt/UniProt and therefore working more directly with what is known about the individual potential templates than more ab-initio focussed methods
If you are working with an enzyme and know the active site (e.g. from sequence conservation, mutational analysis or published data on orthologous proteins), use this information. if you have no prior knowledge of the binding site, Various binding-site prediction programs exist that you can use to identify potential binding pockets.
You can try 3DLigandSite http://www.sbg.bio.ic.ac.uk/3dligandsite/ for ligand site prediction with integrated Phyre modelling, submitting just the sequence, or submitting your own model (let me know how well it works, since I have not tried it)
As Iqra Aroob mentioned, AutoDock Vina http://vina.scripps.edu is a good docking tool. See this tutorial: http://vina.scripps.edu/tutorial.html to get started
If these knowledge-based methods fail to give good results, you go to more generic methods, iTasser https://zhanggroup.org/I-TASSER/ or even Quark https://zhanggroup.org/QUARK/ for modelling the protein
  • asked a question related to Proteins
Question
6 answers
Hello!
Right now, I am working with protein, which we obtained by lysating cells in a protease inhibitor mix with RIPA-Buffer. In the beginning we lysated 10^6 cells in a 0.5ml vessel, but because of slow growth we reduced the amount to 5*10^5 cells without changing any of the protocol. I determined the amount of protein with the Pierce 660nm Assay. Afterwards I performed Western-Blots with 30 µg protein per lane.
Here you see my SDS-Page and in Lanes 3, 7, 8 and 9 I used protein obtained out of 5*10^5 cells, while in Lanes 2, 4, 5, 6 and 10 I used protein obtained out of 10^6 cells. The dye runs at a different speed and even in Total-Protein-Stain you see, that the lanes with 5*10^5 cell-protein contain significantly less protein than the other lanes. The same goes for staining with a antibody.
My question is: Why is there less total protein in the lanes with 5*10^5 cells-protein than in the others, even though there should be the same amount everywhere? Is the Pierce-Assay at fault? Could there be problems with too much protease inhibitor for 5*10^5 cells? And most importantly, can you think of a way to still use the protein samples and quantify it with the 10^6 samples? Because we have 40 reaction vessels full of protein obtained by this method.
Your help would be greatly appreciated!
  • asked a question related to Proteins
Question
3 answers
I may have a luxury problem, but i can see my protein bands before i add the developer. Why is tha?. I follow a protocol that i got from my PI which in essential is the protocol published by Wray et al 1981. I followed it because i wanted to try it and see if i can use it to visualize my DNA protein crosslink.
I followed the protocol to the letter though when i add the pre-stain solution ( mix to the gel i see my protein bands within 5 min. 21ml of 0.36% sodium hydroxide to 2.5 ml of 30% ammonium hydroxide and dropwise added 4 ml of 20% silver nitrate solution to the mix while vortexing and raised volume to 100 ml using distilled water).
Can anyone explain this to me? i have tried searching for answers but the usual problem is that bands are not showing up or background his high.
Thank you so much for helping me.
cheers
Johannes
Relevant answer
Answer
Hello, you can try other silver dyeing methods, like a commercial kit, that will save you a lot of time and energy.
  • asked a question related to Proteins
Question
2 answers
I know that there are libraries for immune antibody libraries in scFv, Fab, or VHH format, but was looking for a way to buy comercial libraries for affibodies. Please let me know if you know a way.
Relevant answer
Answer
Affibody is a trademark of Affibody AB, Sweden. Hence, the only source might be this company: www.affibody.se
  • asked a question related to Proteins
Question
7 answers
Hi!
I have a protein system where a single water molecule can play a role in a ligand stabilization. To check whether this single water molecule is important in binding, I wanted to perform TI (thermodynamics integration) calculations, starting from system with water and annihilate it. My plan was to use AMBER software (PMEMD) for this purpose. I would replace water molecule with dummy atoms during TI. After many trials I finally got my tleap output (parmtop & prmcrd), but while running the script I am getting an error.
My question is: am I doing it right or there is a simpler way than TI to calculate an impact on energy after cutting our water molecule from the system?
I will be grateful for any tips!
Relevant answer
Answer
  • The total amount of energy released when a positron and an electron annihilate is 1.022 MeV, corresponding to the combined rest mass energies of the positron and electron. The energy is released in the form of photons.
  • The amount of energy (E) produced by annihilation is equal to the mass (m) that disappears multiplied by the square of the speed of light in a vacuum (c)—i.e., E = mc2.
  • asked a question related to Proteins
Question
9 answers
I came across papers using only transblot turbo transfer for TGX stain free. We dont have transblot turbo transfer, so am trying to optimize TGX stain free using wet transfer. I activated stain free gels for 45 sec, 2.5 min, 5 min (exposure for Intense) in Chemi Doc imager and transfered in Biorad LF PVDF with Towbin buffer at different transfer condition: 30 volt overnight, 75 volt for 3 hrs. Though transfer looks complete in both the transfer condition as I didnt see total protein bands in gel post transfer but I m getting a very faint total protein bands in membrane after transfer, which I think I cant use for normalization. I am concerned if the wet transfer conditions reduces total protein fluorescence on the membrane. Chemiluminescence image for specific protein in the same blot, however, is very good. So it would be very helpful if anyone could suggest wet transfer conditions for TGX stain free. My proteins of interest are within the range of 15 to 150kD. 
If the trans blot turbo is much better than wet transfer (with regard to transfer efficiency and post transfer total protein intensity on blot), we might have to install one in our lab.
Relevant answer
Answer
Hi Manita,
I am curious about using the TransBlot Turbo with the TGX stain-free gels. I am getting ready to use the TransBlot Turbo for the first time, and wondered if you had any advice that you could give me!
Thanks!
  • asked a question related to Proteins
Question
9 answers
I did ITC experiments with BSA and Its difficult to get same results with same concentration of protein as well as binding ligand.
Relevant answer
Answer
Dr. Ruby Bansal Sushil Kumar Tripathi How to calibrate the pippets, please?
  • asked a question related to Proteins
Question
1 answer
For an experiment i need to stop the initiation of the protein synthesis without stopping the already ongoing synthesis (so cycloheximid is not an option).
Relevant answer
Answer
In yeast!
I would suggest to make/use a yeast knock out strain having prt1 gene deleted along with your 'gene of interest'. The PRT1 is the part of eIF3b subunit of the eukaryotic translation initiation factor 3. It is well worked out and reported in the literature. This may give you a handle to study gene function in translational ON/OFF condition. The Prt1 can be supplied either by putting Prt1 in URA based vector so that it can be expelled out using FOA when required OR under some repressible system where Prt1 expression could be repressed.
  • asked a question related to Proteins
Question
3 answers
I am looking for a physical explanation on the fact that if prokaryotes had some kind of endomembranes it would not have been feasible for them to perform their functions. Any suggestions on this fact.
Relevant answer
Answer
Mmm....
But, if prokaryotes would have endomembrane system, by definition, the will no longer be prokaryotes.
But, leaving semantics apart, I think that may be a waste of energy and resources. The endomembrane system is useful for bigger cells because it increases surface/volume ratio (in order to accelerate material transport and chemical reactions). Prokaryotes solve that being smalls (as you mentioned).
I am thinking loud, je.
  • asked a question related to Proteins
Question
1 answer
I would like to do an EMSA on a known promoter binding site (biotinylated oligo) and we suspect that we need 3 proteins to be present in the complex to bind DNA. We will use transfected HEK-293 extracts overxpressing our proteins of interest. What strategy is the best :
1) Transfect HEK cells with one gene each time and have separate nuclear extracts with one of the 3 proteins and then combine them with the oligos before loading on the gel ?
2) Transfect HEK cells with all 3 genes combined and have just one nuclear extract to add to oligos ?
My main concerns are the stability of the complex during nuclear extraction and the capacity of the proteins to interact with each other and with DNA in the binding buffer before gel loading (we will use the LightShift® Chemiluminescent EMSA Kit from ThermoFisher. Thanks for your advice !
Relevant answer
Answer
If the proteins are present in nuclear extract already, just order the antibodies to get supershifts.
Or you can purchase pure proteins and run EMSAs with these.
Alternatively order the antibodies and do ChIP expt. to know which protein binds in that region.
  • asked a question related to Proteins
Question
6 answers
I have a Kyte-Doolittle Hydropathy Plot result of my protein. I'm trying to understand the result. I am particularly confused about the circled region of the plot. What does this mean?
Relevant answer
Answer
It all depends very much on what you’re aiming for, but using some bioinformatics might add some understanding about your protein and especially some of the regions within the protein. There are methods to:
-Predict secondary structure, using for example SOPMA: https://npsa-prabi.ibcp.fr/cgi-bin/npsa_automat.pl?page=/NPSA/npsa_sopma.html
-Predict hydrophobicity, net charge etc. using Heliquest: https://heliquest.ipmc.cnrs.fr/
-Predict the tendency of the peptide to form a transmembrane helix using ∆G predictor: https://dgpred.cbr.su.se/index.php?p=TMpred
-Predict possible lipid binding potential using Heliquest based Eisenberg plot:
Article New User-Friendly Approach to Obtain an Eisenberg Plot and I...
For an example using Heliquest etc. see:
Article Identification and in silico analysis of helical lipid bindi...
Best regards.
  • asked a question related to Proteins
Question
4 answers
Hello, so i am basically doing a time course and seeing the optimal time to solubilise my protein in DDM + CHS. Not really thinking at the time I solubilised my proteins for x hours then when they were done put them in a freezer until all samples were ready to be examined. Would they continue to solubilise at -20C, thus meaning I should restart the experiment?
Thank you
Relevant answer
Answer
It's highly unlikely that a protein will continue to dissolve at -20, but there could be other artefacts introduced from the freeze/thaw process. I'd want to be very sure the results are not being skewed by this before committing to this part of the protocol.
  • asked a question related to Proteins
Question
4 answers
These concerns MD simulations using Gromacs. Suggestions involving Amber/LAMMPS/etc., aren't going to be of any help.
I'm looking for strategies that might save me on lines of code.
As I am sure you are aware there are several strategies for relaxing a protein in a bilayer. For example, a population approach is placing position restraints on the backbone which are decreased between simulations whilst preserving velocities. Finally, all position restraints are removed and the simulation is left to run unrestrained. This is useful for a sequential simulation->simulation->....->simulation, where the velocities are preserved.
My concern is when I want to run e.g., 4 identical simulations, each with different starting velocities. That way I can cover more phase space and I can calculate a metric of uncertainty when I'm measuring a property. This is very easy to do by hand. Each of the four production runs will use preserved velocities for a separate set of equilibration runs i.e.,
Sim1: eq_run -> eq_run -> eq_run -> production_run
Sim2: eq_run -> eq_run -> eq_run -> production_run
Sim3: eq_run -> eq_run -> eq_run -> production_run
Sim4: eq_run -> eq_run -> eq_run -> production_run
But this becomes very difficult when I have e.g., 100 models to build and run. That's 4*100 + 4*100 + 4*100 + 4*100 simulations with all corresponding files. Ultimately, I am looking for a strategy where I can introduce a bias or short external influence so that each model shares the same equilibration runs before spawning 4 unique production runs but without destroying the velocities preserved from the equilibration simulations i.e.,
Sim1,2,3,4: eq_run -> eq_run -> eq_run -> production_run1/2/3/4
I have a feeling, something like replica-exchange might work? Introducing 4 different temperature that gradually relaxes back to what they should be over a very short period of time just so that the preserved velocities diverge.
Thoughts are most welcome.
Relevant answer
Answer
The strategy you're trying to find is self contradicting. You try to "cover more phase space" by introducing some kind of perturbation from the same point in phase space (end of equ runs). But if you try to preserve the exact state at end of equ runs, it's limiting your exploration of the phase space.
If the only reason of asking this kind of strategy is file management, I'd suggest you to use scripts to do that. Trying to explore more phase space by starting from the same state point doesn't make sense, considering after the "perturbation", you still have to wait for your system to reach equilibrium again. It's equivalent to starting from, say different initial velocities.
Having said that, simulated annealing seems a good candidate of the "perturbation". Just raise/lower the temperature to some random temperature. Again, you have to wait the system to reach equilibrium after restoring your target temperature.
  • asked a question related to Proteins
Question
4 answers
I have tried a few times to couple proteins to my MagPlex beads. Each time I reach at the coupling step (in MES pH 5), I loose most of my beads (70-90%), since they do not stick to the magnet anymore, only diping a tip in the supernatant is enough to remove the beads from the magnet.
Does anyone encountered this problem?
Thanks for your answers
Relevant answer
Answer
Hi Pierre - did you finally reach a good solution? I know this is quite difficult to manage these beads during activation
  • asked a question related to Proteins
Question
3 answers
Hi researchers,
I want to test whether one protein has pro-tumorgenesis effect in pancreatic ductal adenocarcinomas (PDACs). I want to over express the protein in PDAC cell line first but I don't know which cell line should I do.
I checked ATCC website, there are many different cell lines. Anyone can share your experience in choosing cell line?
Thanks!
Relevant answer
Answer
How to get list of normal pancreatic cell lines available ?
  • asked a question related to Proteins
Question
5 answers
I'm currently using Bradford method for a protein assay. And I am measuring the proteolytic activity in roots of certain plants, as many studies found plants have in situ proteases to aid the nitrogen uptake. To determine the proteolytic activity, I will incubate the roots in a known ovalbumin standard solution. Then, after a period of time, the Bradford Assay will be conducted on the incubated solution to look for a decrease in the concentration. I am stilldetermining what concentration to use. In case you don't know, the dye itself without proteins has a maximum absorbance at 495nm, with proteins, it will have a maximum absorbance at 595nm. The dye also has a protein detecting range of 200 - 1500 μg/mL, which is the case for albumins (often BSA is used, but I was restricted to use ovalbumin). When I tested the ovalbumin solution with a concentration of 1500 μg/mL, no color change could be seen, the peak from spectrophotometric analysis also remained at 495nm. I then tested with a range of ovalbumin solutions, then realising with a concentration at or above 50mg/mL, a color change can be seen. The expected results are, there should be a color change & a peak at 495nm when the ovalbumin concentration is at 1500 μg/mL, but there wasn't, and it also requires a high concentration to detect protein concentration, which is odd for a sensitive assay like the Bradford method. Do you have any insights or ideas of what is causing this problem? My current idea is, there is something abnormal about the dye (this is the product I'm using from Bio-rad), or the ovalbumin powder that I used to make the solution has impurities. The impurities idea might be incorrect though, as the powder I'm using is lab grade, and the impurities shouldn't be too significant to this extent.
Relevant answer
Answer
So your sample assay contains 150µg of protein in 5,1mL . According to the BioRad calibration curve, you should expect an OD at 595nm above 0.8... Did you check your reagent with fresh calibrated BSA and/or IgG solutions to get sure it is OK? If you don't run the latter you can't state if the problem is coming from your ovalbumin solution or from your Bradford reagent (or from both)? Further, you have to check your spec and read against a blank sample (0.1mL of buffer + 5mL of reagent and also reading at 595nm is enough. More generally if you don't run any control it is more than difficult to state on the results you get...
  • asked a question related to Proteins
Question
5 answers
I am performing western blots on some old brain tissue and I did a BCA and I basically would need to add 200ul of each protein sample to achieve the right concentration which obviously too much for gel electrophoresis. I think the person who isolated this tissue a few years ago added too much lysis buffer because the sample is clear and usually it is at least a little cloudy. I am trying to figure out the best way to concentrate these protein samples and I am not sure if the lysis buffer will play a role. It looks like the simplest method is TCA or acetone precipitation... Does anyone have any input?
Thanks!
Relevant answer
Answer
Does anyone have a protocol for doing ammonium sulfate precipitation?
  • asked a question related to Proteins
Question
6 answers
Hey everyone,
In my sample preparation of protein sample I am using simultaneous TCEP and CAA for a reduction and alkylation in one step.
Using DTT and CAA a simultaneous reduction and alkylation is not possible - the DTT and the CAA will react with each other (right?) and the disulfide bridges are still there.
Now my question - Why DTT and CAA react with each other, but TCEP and CAA not? Why is this simultaneous reduction and alkylation possible? What are the differences?
I know..
- TCEP reacts with the free electron pair of the phosphorus
- thiol of DTT gets rid of a proton and the sulfur anion reacts
- both reactions are nucleophillic attacks
Thanks!
Relevant answer
Answer
Dear Julia Rauch thank you for your interesting technical question. Actually, I'm far from being an expert in this field as we are inorganic chemists. However, you might find the following articles useful with respect to your question:
1. Systematic Evaluation of Protein Reduction and Alkylation Reveals Massive Unspecific Side Effects by Iodine-containing Reagents
This article is freely available as public full text on RG.
2. Evaluation and optimization of reduction and alkylation methods to maximize peptide identification with MS-based proteomics
(please see attached pdf file)
3. Fast and Accurate Determination of Cysteine Reduction and Alkylation Efficacy in Proteomics Workflows
  • asked a question related to Proteins
Question
4 answers
Hello,
I am trying to find kinetic information (such as rate of enzyme production etc.) on the expression of taq polymerase in E-Coli after inserting a recombinant plasmid.
Any help would be appreciated
Relevant answer
Answer
You need precision and technique
  • asked a question related to Proteins
Question
9 answers
Hey
I'm trying to measure the concentration of a protein I've purified and then labeled with Atto 647 flourescent dye.
The protein has no tryptophans or tyrosines (although does have phenylalanines) so the absorbance is very low at 280nm. I can get around this using the Bradford assay to work out protein concentration when the protein is unlabeled. However I want to label the protein with an Atto 647 flourescent dye, but can not think of a way to calculate the dye to protein ratio as there is no absorbance at 280nm as mentioned. I'm pretty sure Bradford or BCA assays will not be useful either as the solution is blue in colour because of the 647 dye.
Anybody got any tips?
Cheers!
Relevant answer
Answer
Hi there,
UV absorbance at 205nm! As it quantifies peptide bond content of the protein. Therefore the signal is very strong and sample has to be diluted prior measurement (around 50µg/mL).
In the joint leaflet you'll find how to proceed:
  • asked a question related to Proteins
Question
3 answers
I am trying to perform homology modeling using the swiss-model but I have found that the swiss-model gives the same number of residues as that of the template. Whereas my sequence has more residues than that of the model predicted by the swiss-model. What should I do? I am confused as it does not give me an exact number of residues in the 3D structure.
Relevant answer
Answer
Dear Zeeshan
i'm agree with Annemarie Honegger that you have to consider the real protein fragment, and therefore, if present remove the predicted signal peptide from your sequence, because it will not be present in the mature protein.
However as a answer:
Swiss model (similarly to MODELLER) is an "homology modelling" based tool and he can build model only if a similar template is available and in case that the avaialble template cover only a portion of your protein you can built a model only for this part.
There are other software that are ab-initio (as Rosetta) or that are based on fragmented modelling (i-tasser)
in the video
uploaded on my blog ProteoCool
you can find some basic information about those.
ciao
Manuele
  • asked a question related to Proteins
Question
7 answers
For protease inhibitor assay with casein as a substrate what shall I use for hammarsten casein to dissolve completely?
Relevant answer
Answer
a little bit of heating the buffer casein suspension helps dissolve casein; get the suspension in the microwave oven for some seconds
  • asked a question related to Proteins
Question
4 answers
Hi Everyone,
I am trying to characterize the microstructure (localization of carbohydrates, fats, and proteins) of coconut milk powder using confocal microscopy. From the review of the available literature, I have found that fats and proteins can be non covalently labeled using fluorescent dyes. However, I am having trouble deciding the labeling protocol for the carbohydrate. Two common methods that I have seen in the papers are as follows:
1. Use antibodies or lectins conjugated with fluorophores to label carbohydrates
2. Covalent labeling of carbohydrates with FITC
If I use the first method, the signal from antibodies or lectins may interfere with the signal from protein present in the sample.
In the second method, the FITC (probe used for covalent labeling) also shows an affinity for proteins. So, I am afraid that the use of such a probe may also interfere with the protein signal (from the food sample). Is there any other probe available that I can use?
What would be the best way to label carbohydrates such that I can simultaneously image three components in the food mixture?
Thanks.
Relevant answer
Answer
Thank you all for your help :) I will read the articles that you have shared.
  • asked a question related to Proteins
Question
6 answers
Can I dissolve freeze-dried catalase or lysozyme directly into de-ionised water or do I need to use a buffer (separate solutions, not both proteins in one)? I will not be storing the solutions and will be using them straight away.
If I need a buffer what should I use?
What would be the max concentration solution I can prepare?
Thank you
Relevant answer
Answer
Enzymes show maximum activity at a particular p H and stable at molecular level at this p H. So ideal medium to dissolve enzyme protein is buffer of appropriate pH. Working concentration must be calculated from data provided by supplier which varies with substrates.
  • asked a question related to Proteins
Question
1 answer
It is well known that heating can denature proteins. However, what does happen to proteins in the case of short and ultrashort (microsecond or nanosecond scale) heating to extreme temperatures (100-1000 degrees of Celsius) ? Such heating occurs for example when applying ultra/short laser pulses or pulsed intense electric fields.
Relevant answer
Answer
At these short times proteins can withstand much higher temperatures. For example, gelatin gel (5-10 %w/v) remains solid when heated to 500-700 degrees C by IR radiation of microsecond duration. Instead, explosive evaporation of water will occur with mechanical damage to the tissue, possibly breakdown plasma cascades. The paper below LITERALLY has it all. Also you may read about Arrhenius integral which is a measure of thermal damage to macromolecules.
  • asked a question related to Proteins
Question
1 answer
I am using DESMOND for molecular dynamics on protein LRP6 and its ligand. At stage 4 I am having a problem, "job failed due to backend error". The job has failed multiple times. We took the original pdb id 3s8v that is complex of dkk1 and lrp6, and from that we removed dkk1 and extracted the lrp6 and docked to get the appropriate ligand. But while using DESMOND it shows a backend error. Why is this happening?
Relevant answer
Answer
@ Ahila Natarajan Can you please guide me about Desmond (Schrodinger Maestro). I am the new user. As I run the simulation it give the error (attached) at stage 1. How to resolve it?
Regards,
Noor Ul ain
  • asked a question related to Proteins
Question
6 answers
Or is the concept inapplicable?
If it were applicable, could statistical mechanical methods apply? Does entropy?
Relevant answer
Answer
Amino acids in proteins are, of course, not free to move independently like a molecule in solution. First, they are connected to 2 other amino acids (or one other if they are the first or last in the chain, or up to 3 others for a cysteine in a disulfide bond). Second, they are subject to a variety of forces exerted by their surroundings, such as charge-charge interactions, hydrogen bonding, van der Waals interactions, and pi stacking (in the case of aromatic amino acids). Computational methods, particularly molecular dynamics, can be used to model the movement of amino acids in proteins over short time scales.
  • asked a question related to Proteins
Question
8 answers
I tried two antibodies and one show stain in the cytoplasm and the other one in the nucleus. Why?
(Blue - dapi; red - mitochondria; green - protein of interest)
And why is hard to find a good antibody for some proteins?
Relevant answer
Answer
Wolfgang Schechinger hello! both are polyclonal. One is raised against a synthetic peptide to a sequence at the N-terminal and the other one is raised against a recombinant protein corresponding to amino acids localized at the N-terminal.
  • asked a question related to Proteins
Question
3 answers
I am fabricating alphalactalbumin nanotubes via the self assembly process and using the serine protease enzyme. First, I want to assure if after formation the internal cavity has a hydrophobic nature and the outer surface presents hydrophilic properties and second I have prepared some SEM images but they do not demonstrate nanotubes, so I wonder if anyone has worked on this subject that I can talk to him.
Relevant answer
Answer
Dear Iman Katouzian
You can take help from the following article. In the Introduction section you can find all the necessary information you needed for your work:
“Structural characterization of α-lactalbumin nanotubes”
According to this article, “Nanotubes are formed by self-assembly of the milk protein α-lactalbumin, after partial hydrolysis by a protease from Bacillus licheniformis. These unique nanotubes are formed only in the presence of an appropriate cation at neutral pH. The α-lactalbumin nanotube is a heterogeneous self-assembled structure comprising diverse hydrolysis products of α-lactalbumin with molar masses around 11 kDa. “.
  • asked a question related to Proteins
Question
15 answers
Hello,
I am doing a hCBS purification on an AKTA device. The hCBS is produced by E. Coli. The AKTA gives me peaks for my sample application, flowthrough, wash and elution. The wash peak is always lower than the elution peak, however, the wash peak always gives a higher OD280 than the elution peak. I wonder how this is possible?
My own guess is that the wash peak measures the E. Coli proteins that are still bound to the column. With the wash, they are washed off and this are probably more proteins than my hCBS protein, that is washed off the column during elution. I do not know exactly though. I am a student doing my final internship on this and this is all new to me. So please forgive me if what I just said does not make sense at all.
Thank you in advance!
Relevant answer
Answer
The Unicorn software has a function to average the UV signal over the fractions (Fraction histogram). That should give you an indication of what to expect.
It looks like your elution buffer has some UV absorbance. What do you use as blank for the spectrophotometer? If you want it to be comparable to the chromatogram you should use the same buffer that the ÄKTA was zeroed against. If you want accurate determination of your protein concentration you should blank with the elution buffer.
  • asked a question related to Proteins
Question
14 answers
Is there any influence of heat denaturation on outer membrane proteins (bbarrel)? Except defolding of course, something like permanent structure change?
If protein will be expressed as inclusion bodies and then solution will be exposed to heat (15min ~60-70°C), can I still refold it without any problems and the fold will be all right?
Also, already folded protein, exposed to boiling heat and folded again will have any changes in the structure or functionality?
Relevant answer
Answer
The most common cause of protein folding is the loss of disulfide bonds
Due to oxidation, reduction, and buffer concentration
As for the temperature, I have no known yet
  • asked a question related to Proteins
Question
2 answers
Hello everyone :) I have a question. Does anyone know which is the best antibody to use against dCas9-VP64 fusion protein or just VP64?Thank you a lot in advance :)
Relevant answer
Answer
Marsela Hakani it's been a while since your post but did you ever find a suitable antibody? I have been searching and have yet to find a Cas9m4 or VP64 antibody and would be interested to know if you did locate one.
  • asked a question related to Proteins
Question
3 answers
Is it possible to use reverse-phase chromatography (HPLC) for purification.
Relevant answer
Answer
Hi Sangamesh,
Can you please provide me a little more detail?
Which resin did you use? what is ratio of monophosphate and resin?
I have been doing synthesizing guanosine monophosphate(GMP) using POCl3 and trimethyl phosphate, but when I pour my reaction mixture to DEAE sephadex A25 column, it seems like it did not bind to resin and elute with only water. But when I compare my TLC with commercially available GMP, it seems I have the right compound.
I would appreciate if you suggest me. thank you!
  • asked a question related to Proteins
Question
3 answers
Hello colleagues,
is there currently any prediction tool (ideally user-friendly web interface), that can identify protein(s) which will preferentially bind my DNA (or RNA) sequence motif (e.g. ATCGAATTCG). Many thanks for your suggestion/experiences :)
Martin
Relevant answer
Answer
Hi Martin,
I think you'll be interested by the tools proposed on this website:
fred
  • asked a question related to Proteins
Question
4 answers
Difference and advantages and disadvantages of using recombinant proteins versus chimeric proteins as antigens in serological tests
Relevant answer
Answer
Thank you Manuele Martinelli .! I got the idea, Thank you very much for the article as well.
Cheers,
Tharaka
  • asked a question related to Proteins
Question
4 answers
Hi, I am working on a practical involving protein digestion and I am confused as to how there can be a -19 Da difference. I believe it has something to do with the trypsin digestion and its accuracy, but I would like some clarification on the mechanisms of this. I am interested to hear people's thoughts on this.
The following three peptides are detected in the mass spectrometer with the masses listed.
Note that in each case the detected mass is 1 Da higher than the mass of the uncharged peptide because the form detected in the mass spectrometer is the protonated form (M+H+ ) and the mass of a hydrogen atom is 1 Da.
Peptide X: VVFAGAK: (M+H+ ) = 691.4 Da
Peptide Y: SQTTYAYR: (M+H+ ) = 989.5 Da
Peptide Z: VVFAGAKSQTTYAYR: (M+H+ ) = 1661.9 Da
The mass of peptide Z is the sum of the masses of peptides X and Y minus 19 Da.
What causes this -19 Da change.?
Relevant answer
Answer
Dear Leon Matt ,
The m/z value of a peptide [M+H]+ is the sum of the residue masses plus 18.015 for H2O plus 1.008 because of a charge.
Sounds right what Michitoshi Watanabe wrote.
  • asked a question related to Proteins
Question
10 answers
I'm working on magnetic molecular imprinting of proteins with chitosan and sodium alginate. At the first step, I coat the fe3o4 nanoparticles with sodium alginate and then coat the result with chitosan and protein. Then, I remove the protein to make MIPs. The problem is that the MIPs absorb both specific imprinted protein (albumin) and nonspecific protein (lysozyme). Does anyone know the reason or a certain method for this kind of imprinting?
Relevant answer
Answer
Hi,
Dear Postforosh
Call me at 6 to 8 in the evening, please
Good luck
  • asked a question related to Proteins
Question
10 answers
Anyone have any insight? It's becoming a recurring issue. I make fresh transfer buffer each time.
Relevant answer
Answer
Hi everyone, thank you so much for your input. Looks like it was some issue with the apparatus. I added more filter paper/extra sponges and the transfer went better. Even though it was a wet transfer, a tight sandwich is definitely important. My sponges were worn from use. We ordered new equipment and the issue isn't there anymore.
  • asked a question related to Proteins
Question
1 answer
I have seeded keratinocytes in 6 well plate. 120 ul of RIPA buffer(with Pro. K) was used to break cells on ice. However, Bradford reagent (Biorad, diluted with water , 1:4) did not develop any blue color upon addition (that is usual case when protein is present). cells were 100% confluent at time of lysis.
What could be possible reason of RIPA buffer failure. thanks
Relevant answer
Answer
Hi Khuram,
Could you explain your process a bit more in detail? And what you are using for your RIPA buffer? Depending on cell type, addition of RIPA won't be enough and agitation/scraping of the cellular material off the plate may be necessary (with sticky cells). As well, highly confluent cells do not lyse with RIPA as well as just sub-confluent cells (95%).
Considering above, the detection limit of the Bradford assay in conjunction with your lysis condition might not have been ideal.
Try following the lysis protocol below (see BioID methods section) and seeing if there is a difference in what you're doing and here. For on plate lysis, add the (mod)RIPA directly to the wells, agitate, wait 5 mins, and "top-up" the SDS concentration to 0.4% - 1% (to whatever SDS conc. the Bradford Assay kit you are using will tolerate). This should be more than sufficient (with pipetting off the lysate thoroughly from the wells) to lyse your cells properly.
  • asked a question related to Proteins
Question
9 answers
I am working with a florescent compound that is biotinylated. When strepdavidin is present in the system, the binding of the protein with the compound quenches the florescence. I need to perform an experiment in presence if streptavidin, however I need to measure the florescence of the compound. The problem I am facing is because of the florescence quenching when streptavidin is present in the same system. How can I effectively disrupt the biotin-streptavidin interaction so that I would be able to quantify the compound by measuring the florescence. I found few methods like heating and pH alterations, however mild conditions are reversible and harsh condition might affect my compound too. Is there any way to effectively break the biotin-streptavidin interaction and separate them? Both are in soluble state in the system. Any help would be highly appreciated. Thank you.
Relevant answer
Answer
Dear Ankit Pandeya,
1.A short incubation in nonionic aqueous solutions at temperatures above 70 degrees C can efficiently break the interaction without denaturing the streptavidin tetramer. Both biotin and the streptavidin remain active after dissociation and both molecules can therefore be re-used and your compound of interest might be not harmed.
2.We use methanol to elute the biotin/Streptavidin compounds but this might affect your experiment too much.
3.You can also try to add an excess of biotin so that the biotin competes with the biotin bound to streptavidin.
4.If all this does not work, it might worth to try the TAPS label from BioTeZ. The TAPAS label is a chemical tag which is recognized by an antibody and can be used as alternative to Biotin/Streptavidin.
Best, Janko
  • asked a question related to Proteins
Question
3 answers
I have dissolved SF(Silk Fibroin) in Ajisawa's solution and tried to dialyze it against water at room temparature. During dialysis, I have observed the precipitation of SF. I continued the process for 3 days and in the final solution, I have a thick cloudy white colored precipitation.
Is there any way to avoid the precipitation?
I would like to get the amino acid profile for the SF I am working with. 
Relevant answer
Answer
you need to be really careful in how much water you will be dialyzed in when you dissolve silk fibroin (SF) in ajisawa solution. the coagulation of silk is very common when you dissolve in Ajisawa. Some methods suggest adding urea in the first dialysis water. Try Prof Kaplan's papers when doing anything with SF.
  • asked a question related to Proteins
Question
6 answers
Dear all,
I am wondering if someone uses a single point or double point method to determine the intrinsic viscosity of their proteins.
These methods are usually developed for polymers and does not fit for proteins.
Could you share with me your experimence on this.
Best,
Sébastien
  • asked a question related to Proteins
Question
4 answers
I am trying to isolate membrane bound protein from a number of different mouse tissues and I wanted to know if high salt (400mM) RIPA buffer would work on fat.
Relevant answer
Answer
RIPA buffer is very inefficient for extract protein from adipose tissue. Following link gives pretty good answers.
  • asked a question related to Proteins
Question
3 answers
Hi, can anyone guide me with this query of; In order to obtain apo-form of α-LA, is by adding 3-5 mM EGTA to the solution of holo-α-LA (Ca2+bound). This solution was dialysed against the several changes of 0.1 M KCl solution at pH 7.0 and 4 °C”
1.How can we check the real content of calcium in the protein samples? 2. Do 0.1 M KCl solution contains enough calcium to saturate α-LA during the dialysis procedure. 3. At pH 4.5 (ie, the isoelectric pI of α-LA) part of the calcium will be displaced by protons but exact calcium content in the α-LA sample is unknown.
Relevant answer
Answer
EGTA binds Ca2+ very tightly. If the Ca2+ is in equilibrium with the solution to any degree, it will eventually be chelated and will be effectively unavailable for binding to anything else.
As for KCl, it depends on how pure it is. Some of the more expensive grades of common reagents supply an elemental analysis. (example: https://www.sigmaaldrich.com/catalog/product/mm/104936?lang=en&region=US)
You can also remove any minor contaminating divalent metals by passing the solution through Chelex resin.
  • asked a question related to Proteins
Question
1 answer
Hi everyone,
I've been decellularizing 2D cell cultures for immunos and LC-MS/MS using 20 mM NH4OH. Basically, I incubate my cells with this solution for 5 min and keep the ECM that is attached to the dishes. For immunos is quite simple, as I basically fix it in PFA and it's ready to go. For LC-MS/MS I used an SDS-PAGE sample buffer and scratched the samples with it to recover as much as possible. For these assays, no problem at all.
Now I want to make hydrogels out of it and for that I need to solubilize them first. Here is where my problems start. I've tried incubating the plates with 2M urea buffer in a 0,15 M NaCl 0,05 M Tris HCl pH 7,4 buffer with 1% protease inhibitors at 4 ºC in the dark for 1h (scratching the plates before and after incubation) but haven't been able to get any protein when quantifying it (BCA, I've checked and this kit is compatible). Is there any tip/suggestion you could share with me? Is there something I'm doing wrong? Any help is deeply appreciated :)
Note: I'm now going to try and leave the samples at 4 ºC for 2 days to see if that helps.
Thanks a lot in advance!
Relevant answer
Answer
ECM is mainly sugars. I'm not sure how to make hydrogels out of them, but solubilising them will require breaking the long chain sugars (hyaluronic acid, chondroitin, etc.)
  • asked a question related to Proteins
Question
15 answers
Anyone knows what may have gone wrong with those western blots? I initially thought it might be due to air bubbles, but I'm not sure. Someone can help me? Thanks in advance
Relevant answer
Answer
As Irene Reimche and Pierre Béguin mentioned, air bubbles is a likely candidate. It also looks like there was some movement during the blotting process where the membrane partially slide relative to the gel. Lastly it might be the membrane was not thoroughly wetted before using. Be sure to read the instructions on how to properly wet the membrane as it varies a lot depending upon which membrane type you are using.
  • asked a question related to Proteins
Question
10 answers
I'm doing a Bradfod assay but currently I'm still confused as to what blank i should use for my sample, which is the supernatant of PAO1 culture grown in AB minimal medium. For the standard, i use serially diluted BSA in dH2O, so the blanks should be a mixture of Bradford reagent and dH2O. But for the sample's blank, what should i use? Should i use the dH2O blank as well, but also add another sample of fresh AB medium, then subtract the absorption of fresh AB medium from the absorption of PAO1 culture's supernatant??
Relevant answer
Answer
for blank reading. use all reagents which used for measure bradford assay without sample
  • asked a question related to Proteins
Question
4 answers
I am interested in potentially using commercially available customized HPC molecular dynamics simulations. However, I cannot find pricing information on the websites of the vendors. Could someone who has had experience with such services please let me know what kind of price range I might expect for a customized molecular dynamics service? To give context, here are some of the vendors that I am considering:
Relevant answer
Answer
Hi Logan,
Since you already have interested suppliers, why not contact their customer service for quotations? Different companies have different technical support services and final delivery quality, so it’s difficult to compare, depending on whether you care more about data quality or cost.
  • asked a question related to Proteins
Question
12 answers
I have installed the AutoDock Vina and MGLTools 1.5.6. whenever I'm running through CMD. It's showing, --config' is not recognised as an internal or external command, operable program or batch file.
I'm typing this command in CMD as I saw in the video tutorial.
Program Files (x86)\The Scripps Research Institute\Vina> --config conf.txt  --log log.txt
And I kept protein.pdbqt ligand.pdbqt and conf.txt in same folder The Scripps Research Institute\Vina
please help me. 
'
Relevant answer
Answer
Deeti try running the command
"C:\Program Files (x86)\The Scripps Research Institute\Vina\vina.exe" --config config.txt --log log.txt
  • asked a question related to Proteins
Question
10 answers
In general, in a ligand-protein binding study, is there a limit to what percentage of a ligand can be used relative to the final reaction volume?
For instance, a ligand is dissolved in buffer A or water or any organic solvent and the reaction has to be set with a protein in a buffer B at some pH. Is there some established information regarding what the maximum percentage of that ligand can be employed such that the buffer properties of the final solution doesn't change?
Please tell me what to look into. Thank you.
Relevant answer
Answer
There are several different problems here:
  1. Pipetting error is lowest when the volumes of all solutions are near identical. So if you mix an enzyme and a substrate solution, both should be near 50% of the final volume.
  2. Changes in composition are minimised either by making all solutions similar in composition (pH, ions...) or by minimising the volume of a deviating solution. For example, if your substrate is dissolved in an organic solvent like ethanol or DMSO, you want to limit the solvent content of the final mixture to a few percent. In this case the effect of solvent is minimised by having the same concentration in all assays. For example, if you use 1, 2,...,10 µL of substrate, you have to add 9, 8, ..., 0 µL of neat solvent. Of course, in such a case you would also do a compatibility study: What is the highest solvent concentration you can get away with, without significantly changing Vmax and Km.
  3. If your substrate is most stable at a pH different from the pH-optimum of the enzyme, you can minimise the effect of substrate addition by keeping the buffering capacity of the substrate solution as low as possible, and that of the reaction buffer sufficient so that changes of pH upon substrate addition are minimised.
  4. Aside: Some reactions change the pH, for example by producing protons. In such a case it is worthwhile to select a buffering agent with a pKa above the desired pH. Thus, the protons produced would initially increase the buffering capacity of the reaction mix. In case of proton consumption, you would, for the same reason, choose a buffer with pKa below the desired pH.
  • asked a question related to Proteins
Question
4 answers
Does anyone know whether proteins of the eukaryotic translation machinery (e.g. translation initiation/elongation factors) appear frequently in pulldown-analysis of overexpressed proteins?
Relevant answer
Answer
Following
  • asked a question related to Proteins
Question
5 answers
Fungal effector proteins, Secretome.
Relevant answer
Answer
Hi, I'm also looking for it, any news?
  • asked a question related to Proteins
Question
5 answers
Actually, i would like to predict the transmembrane region in several membrane proteins. I have tried several online software's including TMpred, MEMSAT, PROTTER, HMMTOP, TMHMM and DAS. I have got different results from each software. For example i have predicted TM helix for protein WelM and got the results as follows
TMpred - 2 helices,
MEMSAT - 2 helices (Figure attached)
PROTTER - no helices (Figure attached),
HMMTOP- no helices,
TMHMM- no helices,
DAS- 3 helices
Im so confused about the results, anyone can suggest me the trustworthy software.
Relevant answer
Answer
What if I have a GPCR (7TM) and in Protter it always shows 18 TM?
  • asked a question related to Proteins
Question
17 answers
We have dataset containing large numbers of proteins belonging to PDB, SwissProt, TargetDB and unknown sources. We have no idea about their nucleotide sequences, but we are interested in understanding codon preference in these proteins. I would highly appreciate it if you please advise me on how to extract original nucleotide sequences of these proteins.
Relevant answer
Answer
Hi,
Just in case you guys are still interested, this may be the answer:
1. Use your amino acid sequence to blastP in NCBI by selecting the appropriate organism or database;
2. Retain a list of the protein id for the 100% match hit for each of your query sequence, save it in a text file;
3. upload the text list file to NCBI Batch Entrez, to retrieve record for your protein sequences;
4. On the results page, there is an option "Send to", click it, then there is an option to download the "FASTA CDS" for all your protein queries.
I found this option by accident, Hope it helps :)
  • asked a question related to Proteins
Question
4 answers
I want to measure different ions (i.g. Ca2+, Mg2+, Zn2+) in a complex proteins mixture with the MS. However, in the protein mixture some ions will most likely be bound to proteins. I want to free the ions from the proteins, so I can measure them. What is the best way to do this? I was thinking about some kind of method that destroys the proteins but does not precipitate them. Does such a method exist? If so, what is the protocol for this method? Since I'm only interested in the ions, it does not matter what the effect of the method on the rest of the sample is.
Relevant answer
Answer
The answer is: Do not use MS. Use, as suggested AAS, or ICP, after acid digestion of your sample. There are plenty of recipes available in literature like microwave-assisted nitric acid digestion, followed by an element spectroscopy approach.
Good luck with your research
Detlef.
  • asked a question related to Proteins
Question
21 answers
I want to extract membrane+cytosolic proteins for western blots.
I'm working with hardy bacteria, so I have to bead beat for full lysis.
During bead beating, the manufacturer protocol recommends I use detergent-free lysis buffer because the foaming prevents efficient lysis by the beads.
My question: Should I add detergent -- after lysis -- to separate the insoluble/membrane proteins from the cell debris?
Or, can I simply take ~100 uL of lysate (free of detergent), add some SDS loading buffer, boil+centrifuge that, and get good membrane proteins on my western blot?
Relevant answer
Answer
Okay, first you lyse your bacteria.
After that you carry on a low-speed centrifugation as previous suggestions said, you discard the pellet (debris) and you centrifuge the supernatant (1h, 100000 x g)
The supernatant of the ultracentrifugation will contain the cytosolic proteins, the pellet will contain membranes (including plasma membranes and thus membrane proteins)
Resuspend the pellet in a buffer, try it to homogeneize it as much as possible and keep it on ice.
Perform some assays, BCA to quantify proteins, SDS PAGE and Western blot. For these, you may take the volume you need from the sample and mix it with the Laemmli buffer, just do not overload it with proteins. Laemmli buffer has already SDS, so it will solubilize the membranes (keep in solution your proteins)
If you want to solubilize all of the membranes, adjust them to the needed concentration and add the desired detergent. Not all detergents will work, carry on small scale trials before deciding which is better to use.
After solubilization, centrifuge again at 100000 x g, 1H. The supernatant will contain membrane proteins that are being kept in solution by the detergents, the pellet contains insoluble membranes and aggregates.Work with the supernatant to carry on purifications etc.