Science method

Buffer - Science method

Explore the latest questions and answers in Buffer, and find Buffer experts.
Questions related to Buffer
  • asked a question related to Buffer
Question
2 answers
I want to prepare 0.05 M bicarbonate carbonate, pH 9.6 as coating buffer for ELISA.
Could anyone help me with right recipe, procedure, and or reference in the preparation?
Thank you in advance.
  • asked a question related to Buffer
Question
5 answers
I am purifying 1-deoxy-D-xylulose 5-phosphate synthase (DXS) enzyme in E. coli BL21 (DE3) cells. I used one-step nickel chromatography to purify it, but I analysed the sample in the SEC column, and it showed an aggregation peak that eluted in the void column. I followed the protocol of a student who have already graduated. I also analysed her sample, it also contained the aggregation peak but was much smaller than that of mine.
Now, I am confusing what this aggregation is, where it occurs, and how to prevent it.
Media: M9/H2O
Lysis buffer: 20 mM Tris, 250 mM NaCl, 2 mM MgCl2, 0.5 mM TPP, 5mM DTT, pH 8.0 containing Complete protease inhibitors, 250U/mL Benzonase. The cells were lysed by sonication (20s on, 40s off, 70%, 5min) after incubating on ice for 30min, then centrifuged at 19,000g, 40min at 4 °C, the supernatant was filtered and applied to Histrap HP 5ml column.
IMAC Equilibration buffer: 20 mM Tris, 250 mM NaCl, 2 mM MgCl2, 0.5 mM TPP, 5mM DTT pH 8.0. 60mM imidazole as the washing buffer and 200mM imidazole as the elution buffer.
DXS is a dimer, the WM of the monomer is ~68kD.
I analysed the fraction without any treatment after nickel purification, it also showed two peaks as shown in Figure A. I have collected these two peaks, the A260/A280 of the first peak (aggregation) is ~1.0 and that of the second one is ~0.7. Did this mean my protein associated with the nucleus? The SDS-PAGE results are in Figure B, and the Native-gel results are in Figure C. In the native gel, the aggregation couldn't run into it.
I have changed the sonication to cell disruption, but it didn't work. All I could think of was to improve the lysis step, and I was not quite sure if the expression step could affect the aggregation. The Bensonaze I used is >90%, and I'd like to use a higher-purity one. Will this help? Did anyone meet this before, how to improve it? Many thanks for your suggestions.
Relevant answer
Answer
Thanks, Md. Muzahid Ahmed Ezaj . Recently I have done a protein purification, I increased the concentration of Mg and Thiamine (cofactors to stabilise the dimer) and made sure the pH was exactly 8.0, it has improved a lot.
  • asked a question related to Buffer
Question
1 answer
I am performing western blot and recently i have been obtaining faint bands for the samples i had already run and had got darker bands. I wish to determine the concentration of protein in those samples, but they are now gel-ready (loaded in laemmlli buffer). can anyone please suggest a way.
Relevant answer
Answer
It's difficult because of the presence of substances that interfere with protein assays (detergent, reducing agent, dye). The approach I would take would be to run the samples on SDS-PAGE, transfer the gel to the Western membrane, stain it temporarily with Ponceau S, and photograph the stain in white light with a gel documentation instrument. Then you can integrate the density of all the bands in each lane as if they were a single band. Then you can destain the membrane and use it for immunoblotting.
  • asked a question related to Buffer
Question
2 answers
Hello, I am currently trying to optimise a bacterial expression system using BL2 1DE3 bacteria and IPTG. In order to just have a quick look, whether I can see any protein overexpression on a Coomassie stain, I was wondering: can I lyse my bacterial pellets in 4x Laemmli buffer (+5% beta mercaptoethanol) for a whole cell lysate and then directly run this on a gel?
Relevant answer
Answer
Yes. I routinely do this to check for over-expression. 60ul of culture + 20ul of 4x sample buffer + reducing agent. I heat 5 minutes at 80C then vortex for 30 seconds to break the DNA and reduce viscosity. Load 10-20ul on PAGE. Then detect with coomassie blue
  • asked a question related to Buffer
Question
1 answer
Problem 1: In my research, I encountered an issue with my wet transblot procedure: despite using 8-10ug of RNA sample and applying a voltage of 10 v for 2 hours in TBE transfer buffer, I observed incomplete transfer of the top band from the UREA gel (8M) to the nylon membrane upon examination.
Problem 2: for northern blot, I conducted prehybridization at temperatures ranging from 55°C, 60 °C, and 65°C, followed by membrane washing with SSC buffer and blocking with blocking solution. Subsequently, I proceeded wash the membrane in wash buffer and soak in detection buffer and applied CDP-Star on top of the membrane. The entire membrane exhibited fluorescence (is it normal?), later the resulting X-ray film exposure did not reveal the desired bands. Background noise quite bad. I would appreciate any professional guidance or suggestions to address this discrepancy."
Relevant answer
Answer
problem in northern blotting is often sample degradation by RNases (both endogenous to the sample and through environmental contamination), which can be avoided by proper sterilization of glassware and the use of RNase inhibitors such as DEPC (diethylpyrocarbonate)
Northern blots are used to detect the presence of specific mRNA molecules. To do a northern blot, RNA is loaded into the wells of a gel, and separated according to size by electrophoresis. The RNA is then transferred to a membrane filter in a process called blotting.
Northern Blots: 0.05 fmol detection limit with near-IR RNA Probes.
To disrupt the secondary structure of RNA, either formaldehyde or glyoxal/DMSO (dimethyl sulfoxide) is commonly used as a denaturing reagent. Formaldehyde is simply added to the samples and gels, so that the method using formaldehyde is easier to run the gels than that using glyoxal/DMSO.
  • asked a question related to Buffer
Question
1 answer
Dear researchers
How to prepare buffer solution with pH of 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 using NaH2PO4 and Na2HPO4?
Should we use NaOH and HCl?
Relevant answer
Answer
Buffers with pH from 5.8 to 8.0 can be prepared by mixing NaH2PO4 and Na2HPO4 solutions in various proportions, according to tables that can be found online.
You can use acid or base to adjust the pH outside of that range, but the solutions will not necessarily be effective as a buffer at all pHs, because buffering capacity is maximal at a pH close to the pKa of the buffering agent. For the NaH2PO4 and Na2HPO4-based buffers, the relevant pKa for the H+ + HPO42- <=> H2PO4- equilibrium is about 7.2.
The pKa for the HPO4- <=> PO43- + H+ equilibrium is about 12.4, so you could titrate Na2HPO4 with NaOH to make buffers in the range of about 11.4 to 13.4.
The pKa for the H2PO4- + H+ <=> H3PO4 equilibrium is about 2.2, so you could titrate phosphoric acid with NaOH to make buffers with pHs from the pH of phosphoric acid (~1.5 for a 0.1 M solution), up to about 3.2.
So, certain pH ranges are not accessible for buffering with phosphate, and other buffers should be used. Here is a buffer reference site:
  • asked a question related to Buffer
Question
3 answers
I'm a beginner in ITC, does anyone have an idea why when measuring the heat of dilution and in measuring the interaction of HSA with the drug or even with the buffer itself, the curve goes up? HSA measurement at HEPES pH 7.4 and pH 6 give similar results. Different buffer concentrations and different ionic strength do not solve the problem...
Relevant answer
Answer
Providing images from the assays, and some information on the experimental conditions and the experimental setup, illustrating whatever problems you are experiencing in your assays will help a lot to figure out the source of the problem.
  • asked a question related to Buffer
Question
1 answer
how to prepare 0.02 M sodium phosphate buffer (containing 6 mM NaCl, pH 6.9)
  • asked a question related to Buffer
Question
11 answers
I am running SDS-Page western blot using 10% acrylamide gels. However, my samples are not migrating more than 55 kDa. The bands are not defined. I am using 4x Laemmli buffer with LDS from Biorad. The cell lysates are human whole brain lysates. I am wondering if the LDS has something to do with this? I tried to boil the samples at 95 degrees for 5 min; heat at 70 degrees for 10 min, all did not work.
Relevant answer
Answer
The problem was with the Human Brain Whole Tissue Lysate (Adult Whole Normal), novus. When compared with colorectal cell lysate, this difference was obvious. Thank you all for your comments.
  • asked a question related to Buffer
Question
3 answers
We received a limited amount of an antiserum antibody as a gift from another lab and are trying to avoid having to purify it. Our initial titration blot with blocking in 5% milk successfully detected the target protein, but encountered significant non-specific binding likely from high albumin-IgG in the serum.
Would using BSA for blocking decrease the non-specific binding, or could it exacerbate the issue due to additional albumin from the BSA? We have never performed westerns with unpurified antiserum antibodies before so any help or tips would be appreciated!
Edit: This is NOT a phospho-specific antibody
Relevant answer
Answer
I think nonfat dry milk milk and BSA (1-5% each) work equally well as blocking agents for Western blots. Milk is much less expensive than BSA. Use the name brand (Carnation) if it's available.
I'm not sure what you mean by "additional albumin from the BSA." BSA is albumin. The blocking agent in milk is casein. You can also by protein-free blocking agents, which are expensive but useful when you are using strepatividin-based detection, since biological blocking agents can contain biotin.
A good way to remove unwanted immunoglobulins when you only have a little antiserum is to affinity purify on a micro scale. Preincubate a small amount of the antiserum with a sample containing the unwanted antigens but not the desired antigen, if such a sample is available or can be prepared.
  • asked a question related to Buffer
Question
2 answers
After treating my membrane with the primary antibody, the destaining buffer was accidentally used instead of tbst buffer. Immediately after use (about 3 seconds later) it was replaced with tbst buffer (10 minutes, 4 times wash), will it affect the antibody signal? The composition of the destaining buffer is acetic acid, methanol, and 3dw.
Relevant answer
Answer
I would expect exposure to destain solution (methanol/acetic acid), even for a short time, to negatively effect the interaction of the primary antibody with the antigen for 2 reasons: the organic solvent will have a denaturing effect on the antibody, and the acidity will interfere with antibody binding (low pH is used to elute antibodies from antigen affinity columns and vice versa). The brief duration of the exposure (3 seconds) may mitigate these effects to some extent, however.
  • asked a question related to Buffer
Question
4 answers
I have problem for removing polysccharides in the dna extraction of bacteria. when I want to collect supernatant after adding extraction buffer, suppernatant isn't sufficient and DNA concentration is law and isn't sharp. But RNA qualifiction and quantification is better than DNA. Can anyone help me how can I improve the quantication of DNA?
Relevant answer
Answer
Polysaccharides can be a common contaminant in bacterial DNA extraction, particularly when working with certain types of bacterial cells or tissues. These polysaccharides can interfere with downstream applications such as PCR or sequencing.
  • asked a question related to Buffer
Question
1 answer
I have animal feed sample. I am using 1 g feed and 5 ml buffer. After protein extraction. I diluted my sample 20 and 40 times. but the concentration and Percentage of protein varies in both dilutions but sample is same. For example, for 20 times dilution, i got 12% and for 40 times dilution, i got 14 %. I need same values for every dilution of same sample.
Relevant answer
Answer
12% versus 14% may not be a statistically significant difference, depending on the experimental variation. If you made replicate measurements at each dilution, what were the means and standard deviations of the measurements?
If the difference is statistically significant, then you should consider possible technical reasons for it, such as in the way the samples were taken for dilution. A 200 mg/mL suspension may be difficult to accurately dispense. Was there any residue left in the pipette? Was the sample uniformly mixed?
  • asked a question related to Buffer
Question
2 answers
For separation of Human from bacterial DNA
Relevant answer
Answer
The MCLB-1 buffer, also known as Modified Chelat Buffer, is commonly used in molecular biology and biochemistry for various applications such as protein extraction, enzyme assays, and DNA isolation. The composition of MCLB-1 buffer typically includes the following ingredients:
  1. Tris base: A buffering agent used to maintain a stable pH. Tris base is usually added at a concentration of around 50-100 mM.
  2. Calcium chloride (CaCl2): Calcium ions are often included in MCLB-1 buffer to stabilize nucleic acids and proteins. The concentration of CaCl2 is typically in the range of 5-10 mM.
  3. Magnesium chloride (MgCl2): Magnesium ions are essential cofactors for many enzymatic reactions, including those involved in DNA and RNA processing. MgCl2 is usually added at a concentration of 1-5 mM.
  4. Lithium chloride (LiCl): Lithium ions can be included in MCLB-1 buffer to enhance the precipitation of nucleic acids. The concentration of LiCl is typically around 50-100 mM.
  5. Bromophenol blue: This dye is often added to MCLB-1 buffer as a tracking dye for agarose gel electrophoresis. The concentration of bromophenol blue is usually very low, around 0.01%.
  6. Water: The remaining volume of the buffer is made up of distilled water or nuclease-free water to achieve the desired final volume and concentration of the buffer components.
Here's a general recipe for preparing MCLB-1 buffer:
  • Tris base: 50-100 mM
  • Calcium chloride (CaCl2): 5-10 mM
  • Magnesium chloride (MgCl2): 1-5 mM
  • Lithium chloride (LiCl): 50-100 mM
  • Bromophenol blue: 0.01%
  • Water: To make up the final volume
To prepare the buffer, dissolve the appropriate amounts of each component in distilled water, adjust the pH if necessary (typically to around pH 7.5), and sterilize the solution by filtration or autoclaving if needed. Store the buffer at the appropriate temperature and protect it from contamination
  • asked a question related to Buffer
Question
1 answer
I made a buffer solution dissolving 79 g of NaH2PO4.2H2O in 1000 mL DI water. Then tried to adjust the solution pH to 6.0 using HCl or NaOH. Initial pH was below 6, so I added NaOH into the solution. It started increasing the pH gradually, and solution pH came to 5.3. Then I added more NaOH (drop by drop and stirred thoroughly for a good amount of time) but pH was constant at 5.3 for quite some time. Then suddenly it jumped to 7.02. After that, I added HCl (drop by drop and stirred thoroughly) to decrease the pH, but nothing happened. pH was constant at 7.02 for a long time, then suddenly dropped to 5.3. Tried several times but pH jumps between 5.3 to 7.02 and vice versa. I can't seem to find any pH level between these two values. What should I do?
Relevant answer
Answer
You can prepare a sodium phosphate buffer of the desired pH by mixing various volume ratios of equimolar solutions of Na2HPO4 and NaH2PO4. Here is a site to help you.
By the way, it sounds like there may be a problem with your pH meter if you are getting sudden jumps. You may need to replace or recondition the electrode, or there may be a bad electrical connection. Or maybe you are not mixing the solution sufficiently. You should use continuous strong stirring with a magnetic stirrer.
  • asked a question related to Buffer
Question
4 answers
I tried glycine buffer but the yeast cells acidify the buffer so the pH goes down to 7-6 overnight. I need something that will stay around 9 and that isn't toxic to the cells.
Relevant answer
Answer
To maintain a high pH (pH 9) for yeast cell suspensions, you can use a buffer that is effective in this pH range. Tris buffer is commonly used for maintaining alkaline pH and may be suitable for your needs. Here's how you can prepare Tris buffer at pH 9:
  1. Tris Base (Tris(hydroxymethyl)aminomethane): Dissolve Tris base in distilled water to make a 1 M stock solution. The molecular weight of Tris base is 121.14 g/mol, so to make a 1 M solution, dissolve 121.14 g of Tris base in 1 liter of water.
  2. Adjust pH: Adjust the pH of the Tris solution to 9 using concentrated hydrochloric acid (HCl) or concentrated sodium hydroxide (NaOH). Use a pH meter or pH strips to monitor and adjust the pH as necessary.
  3. Final Dilution: Once the desired pH is achieved, dilute the Tris solution to the desired concentration for your experiment. Common working concentrations for Tris buffer range from 10 mM to 100 mM.
  4. Sterilization: Filter-sterilize the Tris buffer using a 0.22 μm membrane filter to remove any particulate matter and microorganisms.
  5. Storage: Store the Tris buffer at room temperature (if using within a few weeks) or at 4°C for longer-term storage. Avoid repeated freeze-thaw cycles.
Tris buffer is widely used in biological research and is generally compatible with yeast cell suspensions.
  • asked a question related to Buffer
Question
2 answers
Hello, everybody.
I am looking to add a DNAse treatment step to my bacterial lysis (gram-negative) procedure for crude enzyme extraction because my lysate always ends up being too viscous. We only have Zymo Research DNAse I in the lab and its information sheet says not to "avoid phosphate buffer and calcium chelators". However, I am using a chemical lysis procedure using Promega Cell Culture Lysis Reagent (following their bacterial lysis protocol) since we do not have a sonicator available. According to their information sheet, CCLR has 25mM Tris-phosphate (pH 7.8) and 2mM 1,2-diaminocyclohexane-N,N,N´,N´-tetraacetic acid. Does anyone know if adding DNAse I to my lysate will still work?
I am not sure of the composition of Zymo's DNA digestion buffer, but I was thinking of supplementing divalent ions to the lysate to counter the 2mM chelating agent in the lysis buffer. However, I am not too sure how to circumvent the phosphate problem. I am not sure how phosphate affects DNAse activity exactly. On the other hand, Thermofisher has a protocol for removing DNA from protein extracts (extracted using lysis reagent in phosphate buffer) using DNAse I. Will the phosphate component of my buffer significantly affect the activity of the DNAse?
Any insight on this matter will be greatly appreciated. Tips on how to solve the viscosity problem altogether are also greatly appreciated.
Thanks
Relevant answer
Answer
The following are the conditions when DNase I activity can be affected.
1. DNase I activity is influenced by divalent ions. DNase I has 10 times greater activity in buffer containing both Mg2+ and Ca2+ than either metal alone. Calcium is required to maintain structure and activity of DNase I. Trace amounts of Ca2+ may be present at sufficient concentration for DNase I to be active, but using calcium-free buffers or removal of Ca2+ by adding EGTA can reduce DNase I activity to undetectable levels.
2. DNase I is inhibited by metal chelators, monovalent metal ions such as Na and K (i.e., ≥ 100mM NaCl), SDS even at concentrations below 0.1%, reducing agents and ionic strength above 50-100mM.
3. DNase I is inactivated by heating to 65°C for 10 minutes in the presence of EGTA or EDTA.
4. DNase I is sensitive to physical denaturation. So, mix gently by inverting tube. Do not vortex.
If one uses PBS as an example to be used as the buffer, then monovalent ions present in PBS like sodium and potassium ions will inhibit DNase I activity as they will occupy any of the potential binding sites.
The contents of (1X) PBS are as follows.
137mM NaCl,
2.7mM KCl,
10mM Na2HPO4, and
1.8mM KH2PO4
However, the inhibition of DNase I can be reversed by adding divalent ions. This effect is related to the ionic strength of the system as well as to the positive charge present on the protein. I have not come across anything about phosphate on DNase I activity.
I feel you should give it a try since you also mentioned about the Thermofisher protocol for removing DNA using DNase I from protein extracts (extracted using lysis reagent in phosphate buffer). But do supplement divalent ions to the lysate.
Best.
  • asked a question related to Buffer
Question
3 answers
I have synthesized a sensor, which has sulphonic acid grp (-SO3H) and Boronic acid as well in it. And has a Molar mass of around 899 and also has an amide bond in it.. I am using Reverse phase HPLC to purify using ACN and Water(0.1%) as mobile phase. As I separate the compounds of one peak, the next very day it is splitting into two peaks in the chromatogram. Is TFA creating a problem? I could not figure it out. Should I need to use a buffer as an additive instead of TFA? Which buffer and how much is good for use? Could you please suggest?
Relevant answer
Answer
A molecule that contains sulphonic acid (-SO3H) Boronic acid and amide bonds may not be a good candidate for RP chromatography. Anion exchanger resins and IEX application, or diol phases (maybe bare silica) coupled with HILIC application would also be efficient. Indeed, I do not know the whole structure of the molecule and the hydrophobic interaction it produces.
Sharing a chromatogram could be beneficial.
In RP, TFA acts as a weak ion pairing agent and it improves the resolution. Remain the TFA at both phases at 0.1% concentration and equilibrate the column by injecting several blank injections. Then test your sample. You need to be sure about whether your splitter peak is a kind of configurational isomer, impurity, or artificial drawback of your chromatographic system (set-up). Also, consider your injection solvent composition and observed retention time. Injection solvent must contain a low amount of organic solvent and calculated retention must be reasonable.
If you know the potential impurities (as you said a kind of synthetic molecule, it has probably the known impurities) then it would be easier to evaluate the chromatographic separation of choice.
Good luck,
  • asked a question related to Buffer
Question
3 answers
My western blot had a problem with the protein band. I just checked it and observed my loading buffer is expired. does anyone explain it is due to expired loading buffer or any other problem i have with it?
Relevant answer
Answer
I'm truly thrilled to hear about your results! Keep up the excellent work, and may you achieve even greater success in the future!
  • asked a question related to Buffer
Question
3 answers
Hello, I want to do EMSA with native PAGE to check protein-dna interactions.
The PIs of my proteins are between 8.1-8.5. I know that the pH of my buffer must be higher, so that the net charge is negative and the protein goes "downwards" to the anode. But do I have to adjust the pH (e.g. let's say 9.5) of everything? So separating gel, running buffer and loading dye? Or is the gel enough? I cannot find anything about running buffer and loading dye.
I my group we only did discontinous native gels so far, but in all recipes the pH of the stacking gel is around 6.8. Then my protein would run out of the gel, wouldn't it? Can I also change the pH of the stacking gel without changing the purpose of the stacking gel? I also found continuous native gels on the internet. Does that really work without getting a big smear?
Relevant answer
Answer
In Native PAGE (Polyacrylamide Gel Electrophoresis), the pH value of each component plays a crucial role in ensuring the proper separation and migration of proteins based on their native charge and size. maintaining appropriate pH values for each component in Native PAGE is essential for preserving the native structure and charge of proteins, ensuring accurate separation and analysis. Any deviation from the optimal pH range can lead to protein denaturation, aggregation, or altered migration patterns, affecting the reliability and reproducibility of the results.
  • asked a question related to Buffer
Question
4 answers
Dear Research Community,
I am encountering a significant hurdle in my research involving enzyme inhibition testing. The inhibitor I am investigating exhibits solubility exclusively in DMSO, rendering it insoluble in aqueous environments such as the 100mM phosphate buffer I am utilizing for enzyme kinetics studies. Upon attempting to incorporate the DMSO-dissolved inhibitor into the reaction mix, it precipitates out, leading to haze formation in the solution and hindering accurate data collection.
I am seeking insights and suggestions on how to effectively address this challenge. Specifically, I am interested in methodologies , or alternative solvents that could facilitate the integration of the inhibitor into the reaction mix without inducing precipitation. Additionally, any advice on modifying experimental conditions or buffer compositions to mitigate this issue would be greatly appreciated.
Thank you in advance for your expertise and assistance.
Relevant answer
Answer
In addition, the suggestion of Adam Shapiro, to lower the phosphate buffer concentration (e.g. from 100 mM to 50 mM ) can be helpful. So, revise your assay conditions.
  • asked a question related to Buffer
Question
4 answers
I regularly have low DNA concentrations on the vetigastropod tissues I extract. I have tried fresh tissue and older/museum tissues. We use the Thermo Scientific GeneJET Genomic DNA Purification Kit.
Heating the elution buffer and using less buffer to elute does help, but concentrations are still quite low. Downstream applications are PCR and sequencing.
Super desperate and would appreciate any suggestions on how to increase the yield from extractions!!
Relevant answer
Answer
Thank you!!
  • asked a question related to Buffer
Question
4 answers
We are interested in detecting aggrecan (250 KDa) by WB. No luck so far, are there any recommendations, such as running buffer, sample loading buffer? Thank you in advance!
Relevant answer
Answer
Detecting aggrecan in Western blotting of mouse brain homogenates can be challenging due to its large molecular weight (~250 kDa). However, with careful optimization of experimental conditions and antibody selection, it is feasible. Here are key points:
  1. Sample Preparation: Ensure effective homogenization of mouse brain tissue and use a suitable lysis buffer to extract proteins.
  2. Gel Electrophoresis: Utilize agarose or gradient polyacrylamide gels for resolving high molecular weight proteins. Run the gel long enough to allow sufficient migration of large proteins.
  3. Transfer: Optimize transfer conditions, considering longer transfer times or semi-dry transfer systems for efficient transfer of high molecular weight proteins.
  4. Blocking: Block the membrane effectively to minimize non-specific binding, using blocking buffers containing BSA or non-fat dry milk.
  5. Antibody Selection: Choose a specific and sensitive antibody validated for Western blotting with a demonstrated ability to detect aggrecan in brain tissue samples.
  6. Positive Control: Include a positive control sample with known aggrecan content to validate experimental procedures.
  7. Detection: Utilize sensitive detection methods such as chemiluminescence or fluorescence, ensuring appropriate exposure times to avoid signal saturation.
  8. Quantification: Analyze Western blot bands using suitable software for accurate quantification of aggrecan expression levels.
By meticulously optimizing each step and addressing specific challenges associated with detecting large proteins like aggrecan, successful detection of the 250 kDa band in mouse brain homogenates can be achieved.
  • asked a question related to Buffer
Question
6 answers
I was wondering if the routinely used NativePAGE sample buffer with a pH of 6.8 causes protein precipitation if my protein has a pI of 7? Could I increase the buffer pH without affecting the electrophoresis? Thanks a lot for your help.
Relevant answer
Answer
To Adron Ung
"Just switch the leads of the electrodes: black to black and red to red going to the electrophoresis gel assembly with ... red to black and black to red going towards the battery. Now the cathode(-) is at the bottom of the gel and the anode is at the top. Science!"
What positive charged dye can be used for such case?
  • asked a question related to Buffer
Question
7 answers
i am trying to stain different proteins of interest in human paraffinized section.
my signal should be the vessel only, but regardless of the antibody I get circle shaped spots , I tried antigen retrieval with trypsin, and I tried different dilution of antibodies (1:100-1:1000) and blocking in 5% and 10% donkey serum
why I have those spots? how can I reduce them? I have the same issue at different wave lengths (regardless of secondary antibodies tag)
this is my protocol:
•Thickness of sections : 10µ
•Deparaffinization by heat 55degrees for 20 min then (xylene - xylene: ethanol - ethanol) 10min each*twice
•Hydration (ethanol 95% - 70% - 50% - tab water) 5min each*twice
•Antigen retrieval : citrate buffer 10min microwave then leave in buffer to cool
•Peroxide treatment: 3%H2O2 10min at room temp
•Blocking: 5% Donkey serum + 0.3% Triton-PBS 1 hr RT
•Antibodies:
•Primary in 1%BSA: VWF (1:200)
•Secondary 1:500 of Rabbit 594
Relevant answer
Answer
hello all,
Just an update, and for future researchers. I ended up doing multiple steps to decrease the background
1- sodium borohydride (1mg/ml in PBS over ice) for 30 min, refreshing it every 10 min
2- 3% H2O2 for 20 min
3- Sudan black for 30 min in dark
4- increase blocking to 10%Donkey serum + 0.3M glycine for 1 hr
5- increase washing to 15 min * 3 times after primary and secondary
thank you for your help
  • asked a question related to Buffer
Question
3 answers
Hi,
im looking at PAL activity in blueberry samples. I use L-Phenylalanine as a substrate and absorbance at 290nm. The absorbance values of my blanks are higher than 1, even my milli pore water is above 1. I diluted buffers, substrate... even remade everything. i am out of options.
my protocol is as follow:
acetone powdered tissue extracted with borate buffer pH 8.8 and PVPP.
75ul sample extract
150ul (l-phenylalanine 30uM and borate buffer 30mM)
after 30mins rxn stopped with 4N HCl.
absorbance at 290nm
my latest results are very close to each other (0.8-0.999) my blanks are above 1.
any ideas to what might be the problem?
Relevant answer
Answer
Standard polystyrene microplates are opaque at 290 nm. You require UV-transparent microplates.
  • asked a question related to Buffer
Question
2 answers
I purified a protein containing His tag via Ni-NTA chromatography wherein the protein was eluted in elution buffer containing 500mM imidazole, 50 mM Tris, 500mM Nacl. Since my next step of purification was Ion exchange chromatography, I dialyzed the protein in dialysis buffer containing 50mM Tris and 5mM beta mercapthoethanol. However, in the third round of dialysis, I observed extreme precipitation of the protein.
Kindly let me know what can be done to reduce the precipitation.
Relevant answer
Answer
Why did you added 5mM beta mercapthoethanol?
since in case you have S-S bond the addiction of reducing agent can induce protein unfolding.
which is the pI of your protein?
Did your protein contain free cysteines?
If not suggest to you to do not add reducing agent to preserve S-s bonds if presents and use desalting instead dialysis (which is long process)
you can find some more information about desalting on the following links
avaialble on my blog (ProteoCool)
best
Manueie
  • asked a question related to Buffer
Question
5 answers
According to Bradford assay, the concentrations are pretty low. Apparently I added way too much extraction buffer to my lysates. Is there a cheap and reliable way to concentrate my protein extracts?
Relevant answer
Answer
One way would be to use a centrifugal ultrafiltration device, such as a Microcon.
A less expensive way would be to precipitate the protein using trichloroacetic acid (TCA). In 1.5-ml microcentrifuge tubes, add 1/10 volume of 72% (w/v) TCA to each sample. Incubate on ice for 10 minutes. Centrifuge at the maximum speed of the microcentrifuge for 10 minutes. Carefully remove the entire supernatant without disturbing the precipitate. Dissolve the precipitate in SDS-PAGE sample buffer.
  • asked a question related to Buffer
Question
1 answer
Dear all,
I am looking for guidance relating to a formula to calculate mass of reagents based on the desired pH and molarity. Is there a formula for this?
I am looking to create, and justify with calculations, a phosphate buffer. I am intending to create phosphate buffers 0.1 M pH 6.0 and 0.2 M pH 6.8.
Thank you in advance for any guidance :)
Kieran
  • asked a question related to Buffer
Question
3 answers
Cannot separate between 10~25kD, always have a line around 25kD...
(Resolving gel buffer: 30% Acrylamide/Bis, 1.5M Tris-Cl, pH 8.8, 10% SDS, 10% APS, TEMED)
Relevant answer
Answer
I suggest using a tricine-page modification to get a higher resolution for lower MW especially for the below 20kda...
  • asked a question related to Buffer
Question
1 answer
We prepared alginate beads 2% in CaCl2 200 mM, after stirring for 1h the beads were washed with water, and incubated with the substrate in the presence of 100 mM tartrate buffer pH 5.5 after 1h incubation we noticed huge weight loss of the beads
Relevant answer
Answer
I think it has something to do with CaCl2 concentration.
High concentration of CaCl2 might cause breakage and weight loss of alginate beads.
I recommend to check this article (open access)
I hope you find it helpful.
Best regards.
  • asked a question related to Buffer
Question
3 answers
I am working with leishmania threonine synthase (LdTS) enzyme of aspartic acid pathway which is ~75kDa protein. I purified the protein and confirmed by western blot too. The condition used for induction is -
0.1mM IPTG concentration at 20°C for 12hours.
For purification I am using Tris buffer and PBS buffer both with Imidazole concentration as by Ni2+ NTA chromatography-
Wash I 5mM
Wash II 10mM
Elution 250mM
The problem which I am facing is that I am non specific bands along with my band of interest. How get rid of this? As well as how I can improve my yield? Currently my yield is 4.146mg/L with Tris and 3.886mg/L with PBS buffer.
I have attached the gel images and blot image below.
Relevant answer
Answer
You can increase the purity by increasing length of the 6XHis tag allowing for stronger imidazole wash and greater concentration of imidazole to elute your polyhistidine tagged protein . How many Histidines do you want? You can add up to an additional 6 histidines (If you already have 6 then you can have 12. If you want another 3 you can have 9) using the following method:
1. Design Forward and Reverse Primers Inverse Primers that amplify the plasmid backbone facing away from each other. The primers contain a gap and overlap distance of zero bases for no frameshift errors.
The forward primer contains near the 5'-overhang or in the middle at (CAC)x6 insertion to add 6 histidines. The reverse primer contains no mutations.
Phosphorylate the primers with T4 polynucleotide kinase + ATP + Mg2+
Perform the PCR with the phosphorylated primers on your wildtype plasmid DNA. See if you get PCR product of the size of your plasmid on a gel. If so then DPNI digestion and then perform gel purification or PCR product purification.
Blunt end ligate the phosphorylated PCR product together with T4 DNA ligase + ATP + Mg2+.
Transform the ligation reaction to E.coli cells, plate on appropriate antibiotic agar plates, select colonies to purify the plasmids from and DNA sequence the plasmids from those colonies.
If the PCR worked and the ligation worked then one of the colonies will have the intended mutagenic insertion of however many extra histidines you wanted. Just like with site directed mutagenesis it is still possible to get primer insertions so make sure you sequence the clones and select the right clones.
I have done this so I know it works.
  • asked a question related to Buffer
Question
1 answer
As an alternative to looking at the conservation of cysteine residues amongst homologs of a protein of interest, is anyone aware of a server that can predict whether disulphide bond formation within a protein is likely to be required for correct folding/oligomerisation? That way one could add a reducing agent to the buffer to reduce the chances of unwanted aggregates forming. Essentially it might be useful to reduce the need for an optimisation step i.e. detection of aggregation after a gel filtration run.
Relevant answer
Answer
It is a difficult question, and I doubt there is a reliable tool for that. You could enter a known protein sequence into various tools (disulfind, etc') and see if they can confirm the formation of a disulfide bond. If a crystalized version of the protein (or a homolog of it) show disulfide bonds, then the answer may be yes. The following page may be of some use:
Alternatively, you could look up evidence of dimerization in the literature - native gels before and after reduction, size estimation via gel filtration columns, and so on.
  • asked a question related to Buffer
Question
1 answer
What are the differences between 50 mM glycine-hydrochloric acid buffer (pH 3) and 100 mM glycine-hydrochloric acid buffer (pH 3), especially regarding osmotic pressure? Why do they have the same pH value but different glycine concentrations.If I need to use it to wash cell , which concentration of glycine buffer is closer to isotonic, thereby ensuring cell survival?
Relevant answer
Answer
The difference in glycine concentration results in a difference in buffering capacity, the ability of the solution to resist a change in pH when acid or base is added. The higher concentration solution has greater buffering capacity. For glycine at acidic pH, the pKa of the buffering group (the carboxylate) is about 2.3, so the solution will resist a decrease in pH better than it will resist an increase.
The osmotic pressure is proportional to the concentration of molecules or ions. Isotonic saline has a concentration of 0.9 weight % of NaCl, which is 154 mM NaCl. NaCl is fully dissociated into ions in solution, and therefore 154 mM NaCl has an osmolarity of 308 mOsm, due to the presence of 154 mM each Na+ and Cl-.
The osmolarity calculation for the glycine-HCl pH 3 buffer is tricky. Simplistically, the osmolarity of glycine-HCl is 3 times the molarity, since it is composed of 3 species (glycine, H+ and Cl-). However, this isn't exactly true, since some of the H+ is consumed to form protonated glycine. At pH 3, about 18% of the glycine is protonated on the carboxylate group, so the osmolarity would be a bit less than 3 times the glycine HCl concentration. Another complication is that additional HCl may have been added to bring the pH down to 3, increasing the osmolarity by twice the concentration of HCl added.
Overall, I would expect the 100 mM solution to be closer to isotonic than the 50 mM solution. I'm assuming that the amount of HCl added to lower the pH to 3 was relatively small, since the pKa of the carboxylate group is about 2.3. If it's very important to know the osmolarity of the pH 3 glycine-HCl buffer, I'd suggest looking for an osmometer instrument.
  • asked a question related to Buffer
Question
4 answers
I am trying to purify a his tagged protein using IMAC. The homologs of this protein purify very well. However, this protein precipitates on elution.
Buffer composition- 20mM Tris-HCl pH7.4, 500mM NaCl, 0.5M Imidazole-HCl pH7.4
Since the homologs are soluble I am not expecting any drastic changes in buffer compositions to be required - this might be a bit naive.
One theory I have is that the protein is eluting too concentrated - causing precipitation. Might it be worth eluting the protein into buffer to dilute the fractions and reduce the likelihood of precipitation?
This protein has a signal sequence which was cleaved during cloning. It might be that I have taken away some of the surface charge that makes the protein soluble. Something I will also check.
Any advice is much appreciated!
Relevant answer
Answer
I notice there is no glycerol in your buffer. 10% - 20% v/v glycerol does wonders.
  • asked a question related to Buffer
Question
8 answers
I have been trying to subclone a gene into the pEGFPC1 vector, and chose BspEI and SalI as my restriction sites. As a control, I tried to perform a single digestion (2hrs, 37 degrees) of the empty vector separately using the two enzymes (BspEI and SalI HF) in NEB Buffer 3.1 (both enzymes show 100% activity as per NEB). However, only BspEI worked, and SalIHF didn't. Could anyone point out why SalI HF was not able to digest the vector in NEB Buffer 3.1?
PS:
  • I want both of the enzymes to work in buffer 3.1 as I want to set up a double restriction digestion. I tried sequential digestion but got a very faint DNA band after a gel run.
  • I can't choose different cloning sites, because all the remaining are present in my gene of interest.
Relevant answer
Answer
Michael J. Benedik I have seen RE cut dna get bigger when the enzyme binds to the target dna but has not cut , Then it looks large ( runs slowly) but addition of 0.05%sds denatures the mixture and the dna runs more like its actual linear size
  • asked a question related to Buffer
Question
2 answers
i use 1.5% agarose gel and 5ul ethidium bromide for 300ml TAE buffer
Relevant answer
Answer
Thank a lot john.
  • asked a question related to Buffer
Question
1 answer
I am preparing the NAP Buffer described in Camacho-Sánchez et al., 2013. The protocol calls for dissolution of 700 grams of Ammonium Sulfate in 1L of water. This step is described as "taking hours" and should be done at "low to moderate heat", but I've had the buffer on a hotplate stirrer for more than 16 hours at 55°C and it still doesn't dissolve completely. Has anyone else had this problem? What temperature range do you use? How long does it usually take you?
Thanks!
Relevant answer
Answer
Dissolving 700 grams of Ammonium Sulfate in 1L of water for NAP Buffer can be challenging, and your experience aligns with what Camacho-Sanchez et al., 2013 describes - it can take hours at low to moderate heat. Here are some suggestions that might help:
  • Double-check Temperature: While 55°C isn't excessively high, ensure your hotplate stirrer temperature is accurate. You can use a separate thermometer to verify the temperature of the solution.
  • Adjust Mixing: Try a more vigorous stirring rate. This can help improve contact between the ammonium sulfate and the water, accelerating dissolution.
  • Try Gradual Addition: Instead of adding all 700 grams at once, consider adding it in smaller portions. This can help reduce saturation and make the dissolving process more efficient.
  • Extended Heating: If the above suggestions don't work, you might need to extend the heating time. However, be cautious of exceeding moderate heat as it can lead to decomposition of some buffer components.
  • asked a question related to Buffer
Question
1 answer
Hi, This is E. Park. I'm trying to purify a protein which was produced in E. coli as inclusion bodies. I already solubilized the inclusion bodies using a 6M guanidine-HCl buffer. Here is my question: I want to store this solubilized inclusion body for 12 hours at 4 degrees Celsius. Should I store this at a lower temperature (freeze) or is it okay to proceed with this method?
Relevant answer
Answer
It should be OK to store the solubilized inclusion bodies as you planned. If you included a reducing agent, there might be some loss of reducing power over time due to oxidation. If that is a concern, then store the sample at -20 instead.
  • asked a question related to Buffer
Question
5 answers
I am new to CO-IP.
I am following this protocol for CO-IP
Lysis buffer 1
0,25ml 1M Tris-Cl
0,15ml 5M NaCl
0,5ml 10% NP40
0,25ml 10% Nadeoxycholate
MQ 3,85ml
1/2 pill EDTA free
Lysis buffer 2 (high salt)
250ul 1M Tris-Cl
500ul 5M NaCl
50ul 10% NP40
25ul 10% Nadeoxycholate
MQ 4.2ml
1/2 pill EDTA free
Lysis buffer 3 (low salt)
250ul 1M Tris-Cl
50ul 10% NP40
25ul 10% Nadeoxycholate
MQ 4.75ml
1/2 pill EDTA free
Day 1
1- Resuspend in medium
2- count cells
3- Wash once with cold PBS
4- Add 1ml cold lysisbuffer 1 with EDTA free protease inhbitor.
5- transfer to an eppendorf tube
6- Add 5ul benzonase
7- Incubate 30min @ 4Co in a 50ml tube on a roller bank
8- Pass 3-5x through a blue needle
9- Spin 10 min at 10000g at 4Co
10- Transfer supernatant to a new tube
11- Save 150ul for total lysate
12- Add 850ul to 50ul beads agarose A and add 10ul Ab
13- Spin 16 rpm o/n @ 4oC
Day 2
1- Short spin 20"
2- remove supernatant
3- Wash 1x with lysisbuffer 1
4- Short spin 20"
5- remove supernatant
6- Wash 1x with lysisbuffer 2
7- Short spin 20"
8- remove supernatant
9- Wash 1x with lysisbuffer 3
10- Short spin 20"
11- Remove all wash.
12- Add 150ul elution buffer (0.1M glycine Ph2)
13- Incubate 10' @ RT with rotation (do not exceed 10')
14- Short spin 20"
15- Transfer to a new tube
16- Add 15ul neutralization buffer (Tris HCL ph8.1)
19- Add loading buffer and Boil samples @95oC for 5'
20- Short spin and load on a 4-20% gel
the question is Why I m seeing this massive band along the membrane and how can I optimize the protocol.
thank you in advance
Relevant answer
Answer
Why is the protease inhibitor EDTA free? How does it make sure that metal ion dependent proteases are inhibited as well? They could chop down anything including capture antibodies.
Concerning "then Input there is no band at all" - did you check the protein transfer after Western Blot via ponceau to see if protein transfer was successful in general?
To optimize the protocol, I would prepare the beads first by blocking them via resuspension in albumin solution, wash once, then incubate with antibodies, then add to sample. otherwise a lot of unspecific binding might occur.
  • asked a question related to Buffer
Question
3 answers
I am been working for a couple of months without success on setting up an assay based on GTPases loading with bodipy gdp and then measure the exchange to GTP in presence of various GEFs.
Reading on literature, the idea is that once the Bodipy GDP is loaded onto the GTPase, there is a significant increase in fluorescence (compared to Bodipy GDP alone) , which decreases when adding GEFs, which exchange it to GTP and thus releasing the Bodipy GDP.
I have been stuck on the first step, because after incubating my GTPase with Bodipy GDP I saw that there was no difference in fluorescence compared to bodipy gdp alone.
Among different protocols that didnt work, here is one:
I store my GTPases in a simple Tris based buffer, tried to buffer exchange them into HEPES buffer that the paper uses but nothing. There are other assays that instead of Hepes use Tris, I have tried those too but nothing.
i think my GTPases cannot load the GDP for some reason and i dont understand why.
If anybody had been through this assay and would like to share any tip in protein storage handling or the assay i would be grateful.
thank you!
Relevant answer
Answer
I have had success in some cases using polarization/anisotropy to measure binding of Bodipy-labeled ligands to proteins. Other fluorescent dyes can also be useful, such as tetramethylcarboxyrhodamine (TAMRA).
Attaching a large fluorescent dye like Bodipy can alter the ability of the ligand to bind, or its affinity. You should also try other fluorescent probes, if they are available.
Finally, I suggest you analyze the purity of your supply of Bodipy-GDP. Commercially supplied probes are not always of the highest quality. You may be able to find a TLC or HPLC method in the literature. It may be possible to repurify the supply using some sort of chromatography. I did this with Bodipy-penicillin using a column of Sephadex LH-20.
  • asked a question related to Buffer
Question
1 answer
hello ,I want to use exogenous ligands to activate receptors on the surface of the U87 cell line. How can I remove endogenous ligands before the experiment to ensure a low baseline receptor activation level? I tried treating the cells with a glycine buffer at pH 2.7 (30 seconds * 3 times), but after treatment, my cells died. How can I optimize my experimental conditions? thanks for your kindly help.
Relevant answer
Answer
As long as the affinity of the ligands is not too high, simple dilution should allow them to dissociate. Remove the medium containing the ligand and replace it with a large volume of fresh medium. Allow some time (half an hour?) for dissociation. Repeat this process a few times.
  • asked a question related to Buffer
Question
3 answers
I want to prepare reduced form of NADPH. I am using 100mM of Tris (pH8.0) to make the buffer but the absorbance is showing to be 1.6 at 340nm which should supposed to be 0.6.
can someone please help me to resolve this?
Relevant answer
Answer
Hi there,
The best thing to do is to perform a scan between 250nm and 400nm so that you will get both specific absorbance peaks of NADPH at 260 and 340 nm. Provided you run a blank first, the absorbance at 340nm will allow you to deduce the concentration of the reduced form and the ratio 260/340 will be indicative of the possible presence of the oxidized form... The ratio is 2.6 if 100% reduced, it tends to increase if oxidization occurs...
NB: both peak values have to be within the linear range of absorbance of the spectrophotometer...
  • asked a question related to Buffer
Question
3 answers
Hi everyone,
Recently, I bought a new cell line named Tenocytes from a company.
I followed the manufacturer's instructions and used their medium and coating buffer.
However, I observed that the cell was not attached to the bottom, as shown in the pictures I attached below.
As you can observe here, I saw all cells are still alive. However, they do not attach to the bottom.
I would greatly appreciate your suggestions or any advice for my experiment.
Best regards,
Relevant answer
Answer
Dear Tina Trinh ,
I would highly recommend to get in tough with the company asap, since they are stating this at the end of the link you have provided:
"Due to the sensitive nature of primary cells and cell lines, all quality related issues about the cells products must be reported back to us within ONE month period after receiving the products, no quality warranty (i.e. replacement of cells) will be provided after the ONE-Month period. Thank you for your understanding."
They should check their batch of the cell stock and give you maybe more clear thawing protocol.
We are usually thawing (different cells) like this:
Thaw the cells 30-60s with in a 37°C water bath (ice should/must still be visible which will keep the cells still cool). Than we are are transferring the cells into 10 ml pre-warmed medium I do use a pipet for that set to 800 µl. After 3-4 pipetting steps adding warm medium into the vial and removing than cells an the medium into the 10 ml reservoir, I spin the cells down (5 min 250 x g) aspirate the medium and plate the cells in new medium into the cell flask or dish. On the next day the medium is exchanged to get ridge of the rest of the DMSO.
Best wishes
Soenke
  • asked a question related to Buffer
Question
3 answers
I am working on quantifying alkaloids in a plant extract. I want to prepare a phosphate buffer solution with sodium phosphate dibasic dihydrate with citric acid at pH 4.7. However, sodium phosphate did not dissolve in water. Kindly request a method to dissolve it properly.
Thank you!!
Relevant answer
Answer
  1. Assessing Water Quality: Ensure that the water used is of high purity. Impurities in water can significantly affect solubility and buffer pH. Deionized or distilled water is recommended.
  2. Temperature Adjustment: Increasing the temperature of the solvent (water) can enhance the solubility of sodium phosphate dibasic dihydrate. Heating the water to 30-40°C before adding the salt may facilitate dissolution without decomposing the salt or affecting its buffering capacity.
  3. Incremental Addition: Gradually add sodium phosphate dibasic dihydrate to the water while continuously stirring. This method prevents the formation of aggregates that are difficult to dissolve.
  4. Use of a Magnetic Stirrer: Employ a magnetic stirrer for uniform mixing. Continuous stirring for an extended period may be necessary until complete dissolution is achieved.
Adjusting to Desired pH with Citric Acid
  1. Preparation of Citric Acid Solution: Dissolve citric acid in deionized or distilled water to create a concentrated solution.
  2. Gradual Addition to Phosphate Solution: Add citric acid solution dropwise to the sodium phosphate solution under continuous stirring. Monitor the pH closely using a calibrated pH meter.
  3. pH Adjustment: Continue the addition until the pH reaches 4.7. The precise amount of citric acid needed can vary; thus, incremental addition and continuous pH monitoring are crucial.
Troubleshooting
  • If solubility issues persist, consider using a sonication bath to facilitate dissolution. Ultrasonic waves can help break down aggregates, enhancing solubility.
  • Verify the purity and quality of the sodium phosphate dibasic dihydrate. Impurities can significantly impact solubility and buffer efficacy.
  • Reassess the concentration of sodium phosphate dibasic dihydrate. Overconcentration may exceed the solubility limit under the given conditions.
  • asked a question related to Buffer
Question
2 answers
Exploring the release rate of paclitaxel from a mesoporous silica nanoparticle, prior studies have employed various release mediums, such as PBS buffer, or PBS supplemented with Tween or SDS at varying ratios. I am intrigued by how paclitaxel, being highly hydrophobic, can dissolve in PBS (pH 7.4) without the aid of a surfactant. Can you elucidate the rationale behind selecting the optimal release medium, considering the notion that it should mimic the cellular environment?
Relevant answer
Answer
Thanks a lot, Alvena Shahid
  • asked a question related to Buffer
Question
1 answer
Hi, I'm doing experiments reconstituting membrane proteins to liposome.
And have a few questions.
1. If I use buffer while hydration of lipid, buffer can encapsulated into liposome. Then, detergent treatment for membrane protein reconstitution (I usually use 0.75% OG, n-octyl-beta-D-glucoside) results in leakage of buffer from liposome?
2. Will buffer leak from liposome during or after detergent removal by dialysis?
Relevant answer
Answer
0.75% OG is approximately equal to the critical micellar concentration, so I would expect there to be some permeabilization and even disruption of the liposome membranes. This will cause the internal and external liquids to equilibrate.
Once the detergent is removed by dialysis, the internal compartment should become isolated from the external compartment again (assuming that the lipids chosen are capable of forming sealed liposomes).
If you need to create a situation in which there are different aqueous compositions inside and outside, you can dialyze the liposomes against a different buffer after the detergent removal stage, or pass the liposomes over a gel filtration or desalting column (e.g. PD-10) equilibrated with the desired external buffer.
  • asked a question related to Buffer
Question
3 answers
I digested my cas9 vector by adding 5ug of vector, 3ul of NEB 2.1 buffer, 3ul of BbsI enzyme and completed to 30ul with ultra-pure H2O.
I incubated overnight at 37C and added 1 ul of CIAP incubated for 10 min at 37C.
I ran my digested vector on an agarose gel and there is no visible band.
Although I can see a band for the intact cas9 vector, I don't for the digested one.
I had already made this digestion and I could see a band before, now it disappeared and I tried to make some new digested vector and there is no visible band as well.
I don't understand what is happening, if anyone has any idea.
Relevant answer
Answer
did You not see any band before the gel extraction? Or after the gel extraction? That will
tel you if the problem is the digestion or the gel extraction step.
  • asked a question related to Buffer
Question
1 answer
Further, how does one calculate the pH of such a buffer solution? Using a pH meter is impossible due to the high viscosity of the buffer solution... Any chemists here?
Relevant answer
Answer
The pH will be determined by the phosphates and sodium glutamate, since sucrose has no ionizable groups. If you measure the pH of the solution made without sucrose, it should accurately represent the pH of the complete solution.
  • asked a question related to Buffer
Question
1 answer
I am blocking some microwells with PBS + BSA 1.0 % and then sensitizing them with LPS. Please let me know what range of absorbance values I should expect for a microwell without sensitizing with LPS, but with the blocking buffer used.
Relevant answer
Answer
You haven't said much about the experiment, but I'll guess you are doing an ELISA-type measurement. The question is, what is the background absorbance in the negative control (no LPS) blocked well. It's impossible to know for sure in your experiment, but in my experience a negative control well had a low absorbance of less than 0.1 in the case of an HRP-conjugate using TMB for detection.
  • asked a question related to Buffer
Question
3 answers
Hello everyone,
I use several pcDNA3.1 expression vectors to transfect cells.
The vectors were prepared by midi-prep a year ago and diluted in TE buffer.
Now that I run new experiments, I decided to measure plasmid concentrations again, prior to transfection.
All their concentration have droped by 2 to 3-fold.
260/280 ratio are still good (over 1.8), but strangely 260/230 ratio have risen (from 2 to 2.3~2.5).
Given the good 260/280 ratio, the presence of EDTA in the buffer and the -20°C storage, I'm pretty sure it is not degradation.
It could be adsorption of DNA on eppendorf tube wall but given the 100~500ng/µL range of concentration, I don't think any tube surface could sequester this much vector quantity.
Anyway I heated my vector for 15min to 60°C and votexed it without increasing the measured concentration ?
The only thing I see would be freeze/thaw cycle maybe ? (I did 10 to 20 such cycles...)
Should I add glycerol to my TE so that freezing and ice crystals don't shear my vector ?
Or just aliquot my vector?
Where did my vectors go guys ???? ^^
Thanks for the help you can provide,
Philippe.
Relevant answer
Answer
But remember it might be the original measurement that was off. Lastly it might not matter, use the lower concentration as real. and It won’t hurt if there is a bit more than expected Unless you need to be very quantitative
  • asked a question related to Buffer
Question
2 answers
Typical neutralization buffer used for Protein A/G affinity separations is 1 M Tris/HCl pH 9.0
Any alternate buffer for 1 M Tris/HCl ?
Relevant answer
Answer
Yes, thanks for your suggestion. will Try!
  • asked a question related to Buffer
Question
1 answer
I am doing protein estimation from fish tissue by Bradford's method but after adding the reagent cbbg- 50mg, ethanol-50ml, ortho-phosphoric acid- 100ml in 1L soln precipitating clot is shown even in the standard BSA solutiin. Each test tube contain. 0.1 ml sample+0.9 ml phosphate buffer+5ml bf reagent.
Relevant answer
Answer
One possibility is that the amount of protein added is too high. Try diluting the sample with water and doing the assay again.
Another possibility is that the amount of other substances besides protein is the problem. This could be nucleic acids and fats, for example. Some additional preparation of the sample may be needed.
  • asked a question related to Buffer
Question
3 answers
I have been running a series of ITC to look at the binding affinity of Fluoxetine Hydrochloride (Prozac) (0.5mM) with Human Serum Albumin (0.02mM). Prozac is my ligand and HSA is in my sample cell. I am using a 0.1M Tris base buffer at pH 7.4. I have been adding runs at different concentrations of ethanol (4%, 8%, 12%, 16%, in both the cell and syringe) to see if alcohol will affect binding, and the magnitude of the peaks significantly increased in the negative. Then I ran a "water into water" using buffer at 12% EtOH in both the syringe and the cell and I still was seeing large peaks. I am interpreting these as the EtOH and my buffer were reacting. But I don't understand how that could be if both the sample cell and titrant were at the same concentration of ethanol. Does anyone have any idea what could be happening? I am an undergraduate Biology major and Chemistry has not always been my strongest subject.
Relevant answer
Answer
Isothermal Titration Calorimetry (ITC) is a powerful technique for studying intermolecular interactions, including protein–ligand binding. Let’s delve into your specific case and address the interaction between Tris buffer and ethanol in your ITC experiments.
  1. Buffer Choice:Tris base buffer is commonly used in ITC experiments. However, it’s essential to select a buffer that maintains the solubility and stability of your biomolecules. Ensure that the buffer contains any necessary salts, cofactors, or additives required for binding studies 1. Tris buffer typically has a high enthalpy of ionization, which can impact the heat measurements in ITC 2.
  2. Ethanol Concentrations:You’ve added different concentrations of ethanol (4%, 8%, 12%, 16%) to both the sample cell and syringe. The magnitude of the peaks increased negatively, suggesting an interaction between ethanol and your system. It’s intriguing that even in a “water into water” control (both syringe and cell containing 12% EtOH), you observed large peaks.
  3. Possible Explanations: Ethanol–Buffer Interaction:Ethanol might indeed interact with the Tris buffer, affecting the heat measurements. Even if both the sample cell and titrant have the same ethanol concentration, local variations or specific interactions could occur. Ethanol could alter the buffer’s properties, leading to unexpected heat changes. Buffer Stability:Tris buffer stability is crucial. Ensure that the buffer remains stable during the experiment. Changes in buffer properties (pH, ionization) due to ethanol could impact the ITC results. Equilibrium Shifts:Ethanol might influence the equilibrium between ligand and protein. If ethanol affects protein conformation or ligand binding, it could lead to altered heat signals. Instrument Calibration:Verify the instrument calibration, including chemical calibration. Incorrect calibration could introduce systematic errors. Sample Cell Contamination:Ensure that the sample cell is free from contaminants (e.g., residual compounds from previous runs). Contaminants could contribute to the observed peaks.
  4. Suggestions: Run additional controls:Blank runs with buffer only (no ligand or protein) to assess baseline heat. Ethanol-only runs to understand its impact on the system. Investigate the buffer–ethanol interaction further:Vary ethanol concentrations and observe the heat changes. Consider using alternative buffers with lower enthalpy of ionization. Collaborate with experienced chemists or biophysicists:Seek guidance from colleagues or mentors who specialize in ITC. They can provide insights based on their expertise.
Remember, scientific exploration often involves unexpected findings. Your curiosity and willingness to learn are commendable! Feel free to seek additional advice from experts in your field. 🌟
  • asked a question related to Buffer
Question
1 answer
Hi! I used the NEB PNGase-F denaturing protocol to denature some lysates and ran them on Western. I usually load 20µg of protein on western; so I did that with my un-denatured lysate control as well as my lysates, using the same amount of the lysate/denatured lysate in the sample prep
2µL lysate/denatured lysate
2µL DTT
3µL lammeli buffer sds (6x)
8µL MilliQ water
Samples were then briefly vortexed, spun, and boiled at 95C for 5 minutes before being run on a gel and probed for the protein. The denatured protein samples show the correct band I want, but when using GAPDH as my loading control, it is evident that the same amount of protein was not loaded. I cannot find anything on the internet about using different amounts of protein for loading after this...
I also made a silly mistake after and thought I could BCA my denatured samples to get the exact concentration of them, but all the glycerol interfered with the BCA and gave super high protein results.
Relevant answer
Answer
FYI, the DTT is what actually interfered with the BCA assay, not the glycerol.
Following the denaturing protocol and the subsequent PNGase F treatment results in the lysate sample being diluted. Did you take that dilution into account?
  • asked a question related to Buffer
Question
3 answers
I want to check the mTORC activity and for that I am using rat kidney. I used cell lytic buffer from Sigma Aldrich to extract the proteins from rat kidney (50mg sample homogenized in 200ul cell lytic buffer with Halt protease and phosphatase inhibitor cocktail followed by centrifugation at 13000 g for 15 minutes), took the protein concentration by BCA assay, and then ran SDS-PAGE at 80V for 1.5 hours. I used BioRad's 10X Tris/Glycine/SDS electrophoresis buffer. I then did the transfer to PVDF at 0.18A for 1.5 hours. I used 10X Tris/Glycine as a transfer buffer. I incubated the blot with S6 primary antibody at 4 degrees overnight, washed with TBST 3 times and incubated with secondary antibody for 2 hours at 4 degrees followed by TBST wash 3 times.
When I developed the membrane, I see the bands are not aligned. I see it smeared. Can anyone help me point out what could be the issue? Shall I lower the voltage while running the gel? or the problem could be something else?
Relevant answer
Answer
Definitely looks overloaded. Find the right concentration that provides a clean lane. Choose one sample, and dilute 1:1 in a series starting at the concentration you show above and performing 6 - 8 dilutions. Ideally, you will find the dilution that gives you a non-saturated cleanly run lane. Then go back and use that same dilution/concentration to repeat all of the samples.
In general, it's best to run ranging pilot experiments to determine your dynamic range. You have to be able to quantify/interpret an effect and you are not able to do so with saturated blots. Give it a try and best of luck.
  • asked a question related to Buffer
Question
5 answers
I've tried to purify recombinant protein from E.coli and I got a good single band in small-scale while several unspecific bands presented in LARGE-SCALE. I tend to Re-Purify the targeted protein (which still in Elution buffer that contain 250 mM imidazole) using Ni-NTA agarose gel.
Should I exchange elution buffer into PBS before Re purify the protein?
Relevant answer
Answer
The contaminating proteins were present in the small-scale prep, just at a scaled ratio making them hard to see until to increased the prep size. If you have access to SEC this is the shortest path to both desalt and polish your protein. It all depends on the size of the other contaminants. May need to add an IEX step to refine. I agree with others, not much to gain from just rebinding to Ni-NTA, but if you wanted to you simply need to dilute the imidazole down to a concentration that is permissive to rebinding.
  • asked a question related to Buffer
Question
10 answers
Hello everyone,
I need a citrate buffer with an optimal pH of 4.8 for the enzymes I'm working with. I'm using citric acid monohydrate (molecular weight: 210.14 g/mol) and adjusting the pH with NaOH. I'm preparing it as a 10x concentration and diluting it to 1x in the final volume (15-200 µl).
However, I've come across recipes for citrate buffer that use both sodium citrate and citric acid.
My question is whether the buffer I'm making will be strong enough to maintain a pH of 4.8 when I dilute it to 1x in my sample. Is the recipe with sodium citrate and citric acid a better option for buffering at pH 4.8?
Relevant answer
Answer
We used sodium citrate and citric acid to prepare citrate buffer . There is a Table for buffer preparations in Methods in Enzymology Vol 1 .
  • asked a question related to Buffer
Question
4 answers
I'm currently using a Qiagen Kit for DNA extraction, I saw white precipitate in the P1 buffer, I was wondering if it's normal and will it effect my DNA yield ?
Thank you in andvance
Relevant answer
Answer
(late reply but hope this may help anyone come across the same issue)
I have similar experience with Qiagen plasmid prep kit, but I realise my precipitate was due to the LyseBlue added to the P1 buffer. An unopened bottle of P1 with no added LyseBlue should be clear.
  • asked a question related to Buffer
Question
3 answers
Hi Everybody. I'm senior in a University in Vietnam. This is fisrt time I do SDS-PAGE, so I want to ask about the correction of my loading buffer recipe for SDS-PAGE as a following file.
In addition, If 0.5M Tris-HCl pH 6.8 isn't available, Can I replace it by 1M Tris-HCl pH 6.8 or any other concentrations of Tris-HCl pH 6.8.
Also, how should I mix these components? (In order or out of order)
(Samples I am using in my research is piglet intestines)
Relevant answer
Answer
Both answers are correct
  • asked a question related to Buffer
Question
2 answers
The detailed Western Blot procedure
The membrane is first rinsed briefly with 20ml of TBS, followed by a 30-minute incubation, shaking with 10ml of Blocking Buffer. During this incubation, the volume of primary antibody needed for a 1:5000 dilution into 10ml of blocking buffer is calculated, and 10ml of blocking buffer is prepared for the antibody. Subsequently, the Blocking Buffer is removed, and the membrane is incubated for 45 minutes, shaking with 10ml of primary antibody in Blocking Buffer (Rabbit anti-ADH diluted 1:5000). After removing the antibody, the membrane is washed three times for 5 minutes each with 10ml of Blocking Buffer. Meanwhile, a 1:5000 dilution of the secondary antibody in 10ml of blocking buffer is prepared. The membrane is then incubated for 45 minutes, shaking with 10ml of secondary antibody in Blocking Buffer (Alkaline Phosphatase conjugated Goat anti Rabbit diluted 1:5000). Following the antibody removal, the membrane undergoes three 5-minute washes with 10ml of TBS/T. Finally, 5ml of BCIP/NBT liquid substrate (Sigma-B1911) is added, and incubation continues until color develops, with the reaction being stopped by rinsing with distilled water.
I was told I might have washed it with a different TBS (10mM one instead of 5mM)
What could the reason for such a different bands on the well.
Relevant answer
Answer
Some possible reasons for seeing extra bands are (1) insufficient blocking, (2) an insufficiently specific primary antibody, (3) too high a concentration of the primary antibody, (4) insufficient quality of the antigen, containing multiple aggregates and proteolytic fragments.
Check by SDS-PAGE if the supposedly purified antigen runs as a single band using a heavy loading to see minor bands.
  • asked a question related to Buffer
Question
2 answers
I'd like to prepare 2.5% glutaraldehyde/2% paraformaldehyde in 0.1 M sodium cacodylic acid. We can buy this pre-made but we only need to use low volume at a time so buying pre-made buffers (minimum order of 100 mL) is not cost-effective. So I plan to prepare this buffer myself and just store by freezing. Can I do this? Will this affect the property of the buffer? Any tips for storing this long term are highly appreciated.
Relevant answer
Answer
Lawrence Ekebafe Thank you so much for sharing this paper.
  • asked a question related to Buffer
Question
1 answer
Due to the high cost of Magnetic IP Kits, our group has decided to make the buffers ourselves. We intend to use SDS Lysis buffer for cell lysis, which should work just fine. However I struggle to find clear information on which wash buffer to use. There are lots of suggestions includung plain PBS, PBS-T, Lysis buffer or a self-made wash buffer (Tris, EDTA, EGTA, Triton x100, NaCl and protease inhibitors) Does anybody have experience or suggestions for which wash buffer to use?
Thank you in advance
Relevant answer
Answer
I assume you're using protA/G mag beads. Pretty robust, but depends on your IP Ab. Start by using some anionic detergent (Triton X100, or NP40 around 0.1%) and isotonic conditions, especially if you intend to move to Co-IP xpts.
  • asked a question related to Buffer
Question
2 answers
I have ordered the SOD colorimetric kit (invitrogen) and a GSH/GSSG ratio assay (abcam, ab205811) and was hoping to use the same brain tissue for both kits. In the methods provided the by the manufactures, the GSH/GSSG assay requires PBS/0.5% NP-40 and the SOD assay just requires PBS. Does anyone have experience with these kits and can advise? Could NP-40 be left out if cells are throughly homogenised with a ultrasonic homogeniser since this is just a detergent?
Really appreciate any advice.
Relevant answer
Answer
Be careful. Although you may achieve sufficient cell lysis to release cell's glutathione and enzymes into the medium, glutathione oxidizes very rapidly in neutral pH. I am not sure about the specific kit you mentioned, but a common practice in measuring glutathione with colorimetric methods is to prepare the cell lysate in acid (e.g., trichloroacetic acid or sulfosalicylic acid). The use of acid serves two purposes, 1. to keep the pH and avoid oxidation, and 2. to precipitate proteins that might interfere in the assays.
  • asked a question related to Buffer
Question
4 answers
I do ELISA many times, and my result is not the same. I realize that when I harvested the supernatant, the volume each well was different. For example I stimulate cells with 200 ul media, when I harvest it, the volume in each well is different and less than 200 ul. So my concern is, should I make all the supernatant at the same volume (200 ul) by adding media, then dilute it with blocking buffer as I want? Or just dilute with blocking buffer directly without making the same volume first?
Thank you very much 🙏
Relevant answer
Answer
Thank you very much for giving me the answer.
  • asked a question related to Buffer
Question
1 answer
Hello! I am not sure whether or not my protein samples are reduced or not. Based on the information below, can someone please let me know if they are reduced?
Sample setup:
__ uL total cell lysate (exact amount calculated from BCA assay)
1 uL Thermo Fischer 10X Sample Reducing Agent
2.5 uL Thermo Fischer 4X LDS Sample Buffer
1X RIPA buffer to dilute sample to 10 uL
Relevant answer
Answer
Dear Kristen,
Protein samples are typically reduced before loading onto SDS-PAGE using a loading buffer containing a reducing agent such as DTT or b-mercaptoethanol (and the sample is also often boiled to further denature the proteins).
If you dilute your protein sample first with RIPA buffer and then add Sample Reducing Agent (and perhaps boil it for 60 - 90 seconds) all of the proteins should be reduced. You can then add some of the Sample buffer and load directly onto your SDS-PA gel for electrophoresis. (I am sure that Thermo Fisher would have supplied a standard protocol for this!).
You should take care to keep all of the buffers at the correct temperature, and always check that the Reducing Agent always smells a bit like rotting eggs as DTT and b-ME go off quickly (I always store these in the fridge but Thermo Fisher may recommend freezing the Reducing agent).
Andrew
  • asked a question related to Buffer
Question
3 answers
I am trying to optimize my control titration for ITC so that I can ensure my ligand to protein traces are reliable.
My small molecule is stored in DMSO. I have been very careful to avoid buffer mismatch. In short I make a 2x stock solution of my compound in 20% DMSO diluted with protein dialysis buffer, and a 2x stock solution of 20% DMSO (without compound) diluted with protein dialysis buffer. The 2x compound stock is added to an equal volume of protein dialysis buffer (species in syringe) and the 2x stock 20% DMSO in protein dialysis buffer without compound is added to an equal volume of protein dialysis buffer (for control) or 2x protein in dialysis buffer (species in sample cell). This results in a final 10% DMSO and 1x ligand/protein solutions or for my control 1x ligand and buffer (for control experiment). This is also all done with locked pipettes.
Upon titrating compound into buffer I am seeing a significant heat transfer but it is not constant or high with a slight linear decrease, which I know is indicative of buffer mismatch.
Is it possible that this compound into buffer just produces a large heat release intrinsically?
I have attached the raw data below.
Relevant answer
Answer
If the ligand-into-buffer thermogram is significantly different to the ligand-into-protein thermogram, then, as a first approximation, you may subtract the first from the second thermogram. This will provide you the net heat effect "mostly" coming from the interaction. However, this is an approximation since the ligand self-dissociation will be coupled to the interaction with the protein and will result in an apparent interaction affinity and enthalpy that do not correspond to the intrinsic interaction parameters. However, if you are just concerned about demonstrating there is interaction, that is fine, and, at least, you have some approximate estimation for the interaction parameters.
A more appropriate alternative would be determining the self-association parameters from the ligand-into-buffer thermogram, and then develop a model in which ligand dissociation is coupled to the protein binding. Using the self-association parameters as known parameters, you may determine the protein binding parameters.
  • asked a question related to Buffer
Question
1 answer
Hello,
I have recently switched from doing ligand into protein for ITC because my ligand is in DMSO and has low solubility at 10% DMSO; therefore, now I am doing protein into ligand -- the heat transfers are much better and seem to correlate to the protein concentration loaded in the syringe which is good to see.
However, at the end of my titration regardless of the concentration of ligand or protein added there is an increase in heat transfers. I am also seeing this in my protein injections into buffer alone.
Any thoughts on why this might be happening?
Relevant answer
Answer
Hi, what is the oligomeric state of your protein? Maybe there is a possibility that you observe some heat of protein association?
  • asked a question related to Buffer
Question
2 answers
I have tested enzyme activity (b-galactosidase) with cation ions in different buffer and the results differ dramatically. For example, while I used MES buffer calcium acts as an activator but when I dissolved my enzyme in phosphate buffer calcium acts as an inhibitor.
is there any explanation besides I done something wrong?
Kindly ask for help and thank you for any answer.
Relevant answer
Answer
the pH and ionic strength of the buffer can also impact the enzyme's activity by affecting its conformation and the interactions between the enzyme and its substrate. Therefore, the specific ions and their concentrations in the buffer can have different effects on enzyme activity depending on the specific enzyme and its substrate.
  • asked a question related to Buffer
Question
5 answers
An unopened Sigma-Aldrich (P4557) phenol solution bottle was shaken (prior to the addition of the Equilibration Buffer) and a gel-like layer formed at the bottom of the bottle. The upper phase is still liquid. The bottle was shaken briefly after the phenol solution was taken out of +4 C. What should be done? Should it be heated in order for it to return to liquid?
Relevant answer
Answer
All you need to do is to heat it up slightly, but not with real flame, do that by gently stirring it until it returns to liquid. If this does not work, contact the supplier/company for assistance. Thank you.
  • asked a question related to Buffer
Question
1 answer
I am trying to create a workflow for flow cytometry experiment sample prep. I intend to use LIVE/DEAD™ Fixable Aqua Stain and an antibody cocktail for extracellular antigens.
I intend to use the viability dye first. The protocol I found does not seem to mind the protein content in buffers, as the cell washing is performed with PBS (5-10% fetal cell serum(I imagine they meant "calf")), and the cells are suspended in the same buffer before viability staining. I read that the presence of proteins in the buffers might contribute to a higher background, is that correct? How should I adjust the solution (for washing and resuspension) if that is correct?
Additionally, I intend to perform antibody titration for the antibodies I'm using. There are a few questions here too. We have no prior experience with these antibodies in the lab. Questions:
1. Is it important to do titration for every antibody in the cocktail?
2. Do I keep the same general sample prep with the antibody cocktail, only swapping the antibody cocktail with a single antibody that's in different concentrations? Do I also apply the viability dye as well?
3. The antibody cocktail I am to use includes dyes such as SB and BV etc, which require a special staining buffer or blocking buffer. Do I need these special staining buffers or blocking buffers for titration as well? Is it necessary since titration typically only uses one dye at a time (together with viability dye?)?
I know the information above might not be detailed enough but could you share your personal experience related to these scenarios? The protocol that I base my protocol on comes from this page
Thanks a lot for your input!
Relevant answer
Answer
Hello,
You can do your viability staining step as the first step of your FC staining, at the same time as your CD16/CD32 blocking step. You don't have to use fetal calf serum or BSA at this point.
For your other questions:
1. Titration is an important step if your plan to use your antibody panel often. Usually, the concentration proposed by the supplier is in the correct range, but the cell you used and the supplier's are for sure different, hence, antibody concentrations might be in need of adjustment.
2.You can prepare a master mix of antibody and prepare a serial dilution of this master mix. You don't have to prepare different dilutions of 1 antibody while the rest are the same, and repeat this step for each antibody. Titration you antibodies with your complete panel, this way you can take in account the signal leak for other fluophores. Titration of the viability dye is less important in my opinion. Supplier's proposed concentration for the viability dye should be enough.
3. Do your titration in the buffers you plan to use in your real experiment
  • asked a question related to Buffer
Question
3 answers
If someone has done some comparative experiment I would be really grateful for results and impressions. I need to detect low abundant microRNAs from plants. I bet that there is no difference among them but who knows :)
Relevant answer
Answer
Hsu Lei Wai Well, I typically use 25-30mL for larger blots. But this is dependent on the size of the bottle you are using and the width of your blot. Even though the protocol suggests lower volumes, I try to make sure the membrane is sufficiently covered with buffer while it is rotating.
  • asked a question related to Buffer
Question
1 answer
Hello,
I want to use a commercial preparation of recombinant PNGase, but I do not have the buffer. What cofactors or buffer conditions does PNGase F require? I will add the enzyme to cell lysate and then run western.
Thanks!
Relevant answer
Answer
Here is the protocol from New England Biolabs.
file:///Users/adam/Downloads/PNGase%20F,%209PIV483.pdf
  • asked a question related to Buffer
Question
2 answers
I have tried to achieve a separation of the nucleus and cytoplasm by using RLN buffer+ NP40 (0.2%), and then proceeding with GeneMATRIX universal RNA purification kit (the same used for total RNA extraction) and followed cell culture RNA purification protocol. I have managed a separation with this protocol in HeLa cells, but SH-SY5Y cells seem to be very sensitive. I have added the protocols and would appreciate any suggestions.
Relevant answer
Answer
It's commendable that you're trying to optimize your protocols for the separation of nucleus and cytoplasm in different cell types. Since you've had success with HeLa cells but are encountering sensitivity issues with SH-SY5Y cells, here are some suggestions to consider:
  1. Optimize buffer conditions: The composition of RLN buffer can affect the efficiency of nucleus-cytoplasm separation. You may need to adjust the concentration of NP40 or other components in the buffer to better suit the sensitivity of SH-SY5Y cells. Try varying the NP40 concentration (within a range that maintains cell integrity) or consider using alternative lysis buffers optimized for neuronal cells.
  2. Gentle cell lysis: SH-SY5Y cells may be more sensitive to harsh lysis conditions. Ensure that you're performing gentle cell lysis to minimize cellular damage. You can try reducing the duration or intensity of cell lysis or exploring alternative lysis methods that are less disruptive to neuronal cells.
  3. Optimize cell harvesting: Ensure that you're harvesting SH-SY5Y cells under conditions that preserve cell integrity. Avoid over-trituration or excessive mechanical disruption during cell harvesting, as this can lead to increased sensitivity to lysis conditions.
  4. Assess cell viability: Before and after cell lysis, assess the viability of SH-SY5Y cells using appropriate viability assays (e.g., Trypan blue exclusion, MTT assay). This can help determine whether the sensitivity issues are due to cell death induced by the lysis procedure.
  5. Consider alternative methods: If you're still facing challenges, consider alternative methods for nucleus-cytoplasm separation in SH-SY5Y cells. Techniques such as differential centrifugation, gradient centrifugation, or commercially available kits optimized for neuronal cells may provide better results.
  6. Consult literature: Look for published protocols or studies that have successfully performed nucleus-cytoplasm separation in SH-SY5Y cells. These resources can provide valuable insights and guidance for optimizing your protocol.
By systematically optimizing your protocol and considering the specific characteristics of SH-SY5Y cells, you can improve the efficiency and sensitivity of nucleus-cytoplasm separation for downstream applications.
  • asked a question related to Buffer
Question
3 answers
I was running PCR 2 lines of fax1 potential mutation (with positive and negative controls).
However in the results I have not been able to detect any DNA bands in the gel electrophoresis with one exception.
I had 2 amplification mixes - one for the fax1 gene and one for the tfax1 mutation (insertion of tDNA).
I used 1.5% agarose gel in 1*TAE buffer with 2ul GelRed per 100ml gel.
and inserted 20ul sample in each of the gel pockets.
Where could the problem be?
(i apologies for any unclarities and mistakes in my question)
Thank you in advance!
Relevant answer
Answer
you have a large primer dimer blurred band which removes primer from the pcr.Try using less primer and use a hot start polymerase. Also check the od260/280 of you dna to check that its quality is good and that you are not adding too much dna to your pcr and poisoning the pcr reaction
  • asked a question related to Buffer
Question
4 answers
I attempted to dissolve 20 mM DTNB(5,5 - Dithiobis(2-nitrobenzoic acid) in 1X PBS buffer(0.1M pbs, pH 7.3), but it was unsuccessful.
Even after adding DTNB to the PBS buffer and vortexing for about 40 minutes, it did not dissolve. Then, I placed the DTNB-containing PBS buffer in a tumbler and reacted it for approximately 16 hours, but it still did not dissolve.
DTNB seems to be insoluble.....
Does anyone know a solution to this? I would appreciate any advice you may have.
Relevant answer
Answer
DTNB is a compound that is slightly soluble in water. To ensure that DTNB can be fully dissolved, a buffer solution, such as a buffer with a pH value of approximately 8, is usually prepared first. Then, DTNB is mixed with sodium bicarbonate and dissolved using this buffer solution.
  • asked a question related to Buffer
Question
4 answers
As NaCl substitute in Na+ free buffers, NMDG (N-methyl-D-glucamine) is firstly prepared as a chloride solution or can we directly titrate with HCl in the final buffer?
Relevant answer
Answer
Dear esteemed colleague,
I trust this message finds you well and thriving in your scientific endeavors. Your inquiry regarding the preparation of N-methyl-D-glucamine chloride, a compound of interest in various biochemical and pharmaceutical applications, is both intriguing and significant. Below, I outline a methodical approach to synthesizing N-methyl-D-glucamine chloride, ensuring clarity and precision at each step:
1. Objective:
The goal is to prepare N-methyl-D-glucamine chloride by introducing a methyl group into D-glucamine, followed by the addition of hydrochloric acid to form the chloride salt.
2. Required Chemicals and Equipment:
  • D-Glucamine (also known as chitosamine)
  • Methylating agent (e.g., iodomethane, dimethyl sulfate)
  • Hydrochloric acid (HCl), preferably anhydrous
  • Organic solvents (e.g., methanol, ethanol)
  • Standard laboratory equipment (e.g., reaction flasks, magnetic stirrer, reflux setup, rotary evaporator)
  • Protective laboratory gear
3. Synthesis Overview:
The synthesis involves two main reactions: methylation of D-glucamine to form N-methyl-D-glucamine, followed by the formation of its chloride salt.
Methylation Step:
  1. Reaction Setup: Dissolve D-glucamine in an appropriate organic solvent under an inert atmosphere to prevent oxidation. Methanol is often used due to its compatibility with the reagents.
  2. Methylation: Add the methylating agent to the D-glucamine solution gradually while maintaining stirring and controlling the temperature to prevent excessive reaction rates that could lead to by-products.
  3. Monitoring: Monitor the reaction progress via thin-layer chromatography (TLC) or high-performance liquid chromatography (HPLC) to ensure complete methylation.
  4. Completion: Once the reaction is complete, remove the excess methylating agent and solvent under reduced pressure using a rotary evaporator.
Salt Formation Step:
  1. Hydrochloric Acid Addition: Dissolve the N-methyl-D-glucamine in a minimal amount of anhydrous ethanol or another suitable solvent. Slowly add anhydrous HCl gas or a concentrated solution of HCl in the same solvent to the solution, controlling the addition to maintain the reaction's temperature and prevent local excesses.
  2. Salt Precipitation: The N-methyl-D-glucamine chloride typically precipitates out of the solution as a solid. If not, cooling the mixture can facilitate precipitation.
  3. Filtration and Washing: Filter the precipitated salt and wash it with a cold solvent to remove any unreacted starting materials or by-products.
  4. Drying: Dry the solid under vacuum or in a desiccator to obtain the final N-methyl-D-glucamine chloride product.
4. Characterization:
Characterize the synthesized N-methyl-D-glucamine chloride using techniques such as nuclear magnetic resonance (NMR) spectroscopy, infrared (IR) spectroscopy, and mass spectrometry (MS) to confirm its structure and purity.
5. Safety Considerations:
  • Handle all chemicals, especially the methylating agents and HCl, with care, using appropriate personal protective equipment (PPE).
  • Conduct the synthesis in a well-ventilated fume hood to avoid exposure to hazardous fumes.
  • Dispose of all waste materials according to local regulations and guidelines.
In conclusion, the preparation of N-methyl-D-glucamine chloride requires careful attention to reaction conditions, reagent selection, and safety precautions. By following the outlined protocol, you should be able to synthesize this compound efficiently and safely.
Should you require further clarification or assistance with your synthesis, please do not hesitate to reach out.
Warm regards,
Reviewing the protocols listed here may offer further guidance in addressing this issue.
  • asked a question related to Buffer
Question
2 answers
I have acetic acid glacial 100% GR solution
Relevant answer
Answer
If you have a pH meter in your lab, you can prepare the solution by mixing two solutions - 0.1 M sodium acetate and 0.1 M acetic acid (diluted 1:174 from glacial acetic acid, which is 17.4 M). Gradually add one solution to the other in a beaker with magnetic stirring while measuring the pH until the desired pH is reached. The acetate concentration will be 0.1 M.
Another approach using the pH meter is to add glacial acetic acid dropwise to a solution of slightly higher than 0.1 M sodium acetate until pH 5 is reached, then adjust the volume to bring the sodium acetate concentration to 0.1 M.
Make sure to calibrate the pH meter with pH 4 and 7 standards before beginning. Also, since mixing solutions will cause the temperature to change, and pH is temperature-dependent, you may have to wait for the temperature to return to ambient temperature before completing the titration.
  • asked a question related to Buffer
Question
3 answers
i ran a 0.8% agarose gel for 130V, half an hour and loaded 5ul of DNA 1kb ladder which looks very very dodgy and has smeared at the higher kb bands - anyone got any ideas what happened and went wrong :(
side note - i didnt load my samples into every lane yet each lane is full?? is that because of too much buffer covering the gel or an issue with making the gel?
Relevant answer
Answer
From the distance that your dna ladder has run I think that you are running the gell at too high a current.Run at a lower voltage and the larger bands should improve. I think that the most likely reason for all lanes being populated is that you poured the gel too hot and the tray warped so that the comb teethe are touching (or very close) to the bottom of the gel tray. Thus when you remove the comb the bottom of the gel is torn and dna can flow under the gel into the next well. Allow the gel to cool more before pouring and make sure that it is well set before removing the comb. Also watch the samples during loading to see if they are spreading. The blue loading dye will show if this is happening. I agree with Péter Gyarmati that a denser gel would help .0.8% is good for larger molecules but your amplimer is quite small
  • asked a question related to Buffer
Question
5 answers
Recently we’ve noticed these patterns on our separating gel 15% and I’m wondering if anyone can provide insight into it. Is it normal? And if it isn’t what could we possibly be doing wrong?
We’ve already changed our Temed, made a fresh new AP and buffer too.
Relevant answer
Answer
”hi, do you pour your gel from one point? This also looks like you choose one point and pour gel to the cast. i would recommend you to move through the cassette when you are pouring gel. If you continuously pour gel from one point, the first amount of gel starts polymerization and if you continue to add more gel under this part, the weight causes shrinkage . So you can try to move your pipette during gel pouring from one side to other side of the cassette. And be sure that you are pouring your gel immediately before your mix is ready.
  • asked a question related to Buffer
Question
3 answers
Hello everyone,
This is my first question and very curious to learn why my GST fused protein shows band in the cell Lysate flow through, lysis buffer, wash buffer, but absolutely no bands on the elution buffer on the SDS PAGE? I checked the concentration in the A280 nano drop the concentration shows high absorbance with clear peak intensity?
It is a GUS -GST recombinant protein and I'm using the GST resin columns for the purification process. I have been following the exact protocol suggested by them.
1. Is the resin column unable to bind with the GST fused protein?
2. Should I increase the concentration of glutathione in the elution buffer to elute out the tightly bound GST bound protein?
3. Should I remove the NaCl salt from the wash buffer?
4. Do you think that having EDTA could interfere between releasing the GST bound protein from the column?
5. Should I spin concentrate the solution and run SDS PAGE again on the concentrated samples to see the bands?
Any suggestions would be greatly appreciated.
Thank you for the support in advance.
Best
Relevant answer
Answer
Either the target:
A. doesn't bind (as you see it in the flow-through); or
B. it binds and saturates the column, and then excess runs through (as you see it in the flow-through); AND then
C. it never comes off (as you don't see it in the eluent); or
D. the bound protein properly elutes but then something happens to it (as you don't see it in the eluent).
To check possibility C (see whether your protein sticks to the beads) you can boil a little resin in SDS-PAGE sample buffer and load the supernatant onto your gel. Check the resin after washing and after elution.
Now, let me go through your questions.
1. This is possible (my A, above), typically because of something weird in the sample or sample buffer. Double-check pH. Make sure there's no GSH in the sample (e.g., cell growth medium). No chaotrope or detergent.
2. That can't hurt. GSH is fairly cheap. Watch the pH. You can also include 0.05% Tween 20 to help break up non-specific stickiness (my B, above).
3. You can try that. It probably will reduce stringency of washing, which may give you more non-specific binding.
4. No. Don't leave out the EDTA. It won't inhibit elution, and i's important to inhibit proteolysis.
5. That shouldn't be necessary. Be sure to include protease inhibitors (in addition to EDTA) in the eluent (my D, above).
  • asked a question related to Buffer
Question
3 answers
My current protein concentration is 13mg/ml. To load 30ug protein for SDS PAGE, only 2.15uL is needed, can i load 2.15uL sample + 2.15uL 2X Laemmli Buffer = 4.3uL in the well or do I dilute the sample first so that more volume is needed?
If so, how do I dilute the cell lysate, or can I just use PBS?
Help!
Relevant answer
Answer
I diluted my proteins in pbs. It works just fine. Alternatively, you can dilute them in your lysis buffer so that they will have final concentration of 3ug/ul. In that scenario, you can load 10 ul for each well and they all contain 30 ug protein.
  • asked a question related to Buffer
Question
2 answers
I've been attempting to utilize Native PAGE to observe how my protein of interest, Hsp90, interacts with its cochaperones and clients within mouse embryonic stem cells (mESCs). I'm hoping to run quality Natives to determine and characterize the many complexes it is associated with, however after running several native gels and playing around with some of the conditions, I'm still not confident in how great they are because there seems to be a lot of streaking within the lanes especially on the sides (aligned along the walls of the well where sample loading occurred). I run whole cell lysate through, anywhere from 100-150ug of protein though I have loaded less. Using Native PAGE, I know not to expect great resolution, however I usually do see some well defined bands and my NativeMark runs through well. If streaking were to occur I would expect it to happen more uniformly across each lane and not just on the left and right side of the lane, leaving the middle seemingly unaffected?
If I could get any advice from someone that has experience running Native PAGE especially with lysates that would be greatly appreciated. If it helps my lysis buffer is 20mM HEPES pH 7.4, 50mM KCl, 2mM EDTA pH 8.0, and 0.01% NP-40. For making the acrylamide gel I utilize a Tris-HCl buffer at 0.375M pH 8.8 that I use to dilute 30% acrylamide to make a 4-15% gradient gel with a 3.25% stacking (my stock acrylamide is 30% Acrylamide/Bis solution 37.5:1 if that is important). The Native Sample Buffer I use to load is a 4x concentration (125mM Tris-HCl pH 6.8, 50% glycerol, 0.08% bromophenol blue) and the running buffer (10x) is essentially standard running buffer in the absence of SDS (0.25M Tris base, 1.92M glycine)
I've attached images of some of my Natives. The one with the fluorescent Western Blot was from cells transfected with mCherry-Hsp90 and blotted for mCherry.
I also have an undergrad that is attempting to work out a Blue Native PAGE protocol, but that too seems to have similar issues with side streaking along the well borders all the way down the gel. Again any help and advice would be appreciated!
Relevant answer
Answer
Christian Magaña Vergara I'm in the same boat as Seth and seeing as he didn't reply perhaps I can hear your thoughts.
1. Whole cell lysate clarified by a 30min spin at 20000xg
2. Approximately 30ug of total protein
3. My target is NOD2 which has a membrane-associated as well as a cytosolic pool
I've tried benzonase treatment which reduced streaking somewhat. My downstream application is Western blotting, I have not been able to obtain discrete bands on my blots. Any ideas?
  • asked a question related to Buffer
Question
1 answer
1. If we looking for detection of estrogen in my sample. the concentration of antigen in sample should vary but the Test line should be same concentration. If I choose a BSA-bound estrogen of 1mg/ml as concentration and use for development of Test line. Is this correct?
2. For sample preparation can we use Phosphate buffer. Are hormones dissolve in Phosphate buffer?
Relevant answer
Answer
Test line concentrations on LFA are highly variable depending on the assay. I’ve used concentrations as low as 0.125 mg/ml to several mg/ml, so there is no way to answer this question without empirically determining what the optimal test line concentration is. Too high of a testing concentration and you will get false positive results. 1 mg/ml is a good starting point but you should test a range of concentrations.
  • asked a question related to Buffer
Question
1 answer
The buffer contains: 100mM NaCI, 0.1% Triton X-100, 300mM Sucrose, 1mM MgC2, 1mM EGTA, 10mM PIPES (pH 6.8), and 100uM PMSF
Relevant answer
Answer
I would not use it. First, the PMSF is no longer intact. This compound rapidly degrades in water. You could add fresh PMSF to replenish it, but second, the Triton X-100 will have oxidized and could even be destructive due to reactive oxygen species.
  • asked a question related to Buffer
Question
3 answers
Hello every hope you all will be fine.iam reading a paper and iam confuse about that point 5x10^5 dilution.iam giving the method below please seniors help me and clear my mind about procedure.Thanks in advance
Question
The MRSA suspensions were then diluted 5 × 10^5 fold with 1× PBS buffer
Relevant answer
Answer
500,000-fold dilution
This would have to be done in stages.
For example, start with a 100-fold dilution (10 µl into 1 ml, for example).
Then dilute the 100-fold dilution by another 100-fold, which brings it to 10,000-fold.
Then dilute the 10,000-fold dilution by 50-fold to get to 500,000-fold.
  • asked a question related to Buffer
Question
1 answer
Hi guys,
Can anyone recommend a good protein extraction buffer for enzymes? After extraction I would like to test the enzyme for its activity in an assay, so it should still be active in the buffer.
Many thanks in advance!!!
Relevant answer
Answer
There is no such best protein extraction buffer for enzyme, when i work with new protein, typically I use 50 mM Tris.Cl, 10% Glycerol, pH 7.1. and if there is a possibility of aggregation by the known sequence structure (such as any disulfide bond, etc) I will add BME or DTT in the buffer
  • asked a question related to Buffer
Question
5 answers
Hello,
I am doing ITC and getting repeatable curves that look very promising. After reading some articles, it seems the biggest challenge to avoid is buffer mismatch. Is there a way to rule out that my curve is indeed ligand to macromolecule binding rather than buffer mismatch (i.e. shape/values of curve, shape/values of trace, thermodynamic values)?
I am very careful when preparing my samples but because my ligand is a small molecule I can't dialyze. I am pretty certain my buffers are well matched but I just want to be certain.
Any help would be appreciated.
Relevant answer
Answer
Buffer mismatch results in large, similar heat effects, but not in thermogram profiles similar this one you are showing.
Although buffer mismatch should be avoided, there are ways to handle it and reduce its deleterious effect on data fitting. Unless the mismatch is too large, usually good estimates of the interaction parameters can be obtained.
  • asked a question related to Buffer
Question
1 answer
Has anyone used an alternative tranfer buffer, for a semi-dry tranfer? Currently we are using a Thermo-Fisher branded one, but its running out and the recipe/reagents are propietary. Does anyone have an alternative protocol for this?
Relevant answer
Answer
The most commonly used transfer buffer is Towbin's (doi:10.1073/pnas.76.9.4350, in effect SDS-PAGE running buffer with both SDS to increase the mobility of proteins and methanol to reduce it). doi:10.1006/abio.1993.1313 uses separate buffers on the membrane and gel side to resolve this conflict. In my hands, however, Dunn's buffer (10 mM NaHCO3, 3 mM Na2CO3, doi:10.1016/0003-2697(86)90207-1) is cheaper and works better.
I also find that tank blotters work better than semi-dry, probably because the Joulean heat produced by the current is distributed into a larger volume.
  • asked a question related to Buffer
Question
2 answers
I have a few proteins that I produce recombinantly in E. coli cells. All of them are disordered proteins and are stored in the presence of 4mM DTT at -20°C before use. One of them also contains 1.5M urea in the storage buffer. I have found that over time, the concentration of the proteins, measured at 280nm, increases significantly. Is this due to the presence of DTT? I know that oxidised DTT absorbs strongly around 280nm, but my blank also contains DTT. How can I resolve this issue?
Relevant answer
Answer
I agree with Prof. Adam. Sublimation can cause increase in protein concentration.
I also think it might be due to the precipitation/aggregation. Since they are disordered proteins, freezing them at -20C slowly may encourage nucleation and precipitation/aggregation. This is one of the reason, proteins are flash frozen, to prevent precipitation/aggregation.
Another reason could be the presence of urea or DTT. Urea and DTT are chaotropic agents (cause protein to unfold) and usually causes decrease in fluorescence emission intensity in relation to increasing concentrations of the agents. However, there are few proteins that behave contradictorily where fluorescence intensity increases with increasing concentrations. The reason for the increase in intensity is ambiguous. That said, the absorption/emission correlation can be used to understand the change in concentration.
To resolve this, I would suggest checking for change in fluorescence emission with increase in urea/dtt concentration. If you see an increase (and not a decrease like usual), then that should answer the question. You can also try adding glycerol or PEG to see if protein precipitation/aggregation was the reason.
  • asked a question related to Buffer
Question
4 answers
Hi there.
I have to use the pET28a-SUMO vector to get my target protein.
However, I can‘t separate it from SUMO tag.
My protein was about 15 Kda big and has a about 5 value of pI.
Due to the process next is about X-ray diffraction.
How I can i get my protein without tag or without 80% tag?
After tag on/off-columns cleavage, my protein will stick on the Ni column if I want to remove the tag only if I introduce buffer with imidazole. But buffer with imidazole will bring the SUMO tag down even you use 10mM imidazole.
How can i get my protein without SUMO tag ??????
I have tried ion exchange chromatography and It was not work.
6His tag only? Inclusion body.
MBP tag?TEV is trash enzyme and same problem when i remove the tag.
GST tag?A little of protein is soluble.
Relevant answer
Answer
Try to change another protease cleavage sites, such as ULP1
  • asked a question related to Buffer
Question
5 answers
Different oxygen buffers of magmas, such as FMQ and NNO, have been used in the papers. How to transfer the FMQ to NNO if we want to make comparision?
Relevant answer
Answer
Hi Dr. Huang,
Normally, if you know the magma's temperature, pressure, and oxygen fugacity relative to a buffer (e.g., ΔFMQ, ΔIW, ΔNNO), you can calculate the magma logfO2 directly, and then convert the magma fO2 from one buffer to another. Here is a paper that you can get expressions and related constants (Table 1):
B. Ronald Frost; Introduction to oxygen fugacity and its petrologic importance. Reviews in Mineralogy and Geochemistry 1991; 25 (1): 1–9.
Thanks for your good question,
Dian
  • asked a question related to Buffer
Question
1 answer
Lipid vesicle preparation
Relevant answer
Answer
They will probably be abke to multilamellar if you ask, they always allowed us to customize our liposomes.
  • asked a question related to Buffer
Question
8 answers
sdf
Relevant answer
Answer
Thx!
  • asked a question related to Buffer
Question
1 answer
My colleague is trying to study the effects of a protein on pathogen inhibition in N. benthamiana. He is expressing it in either a pSITE vector or pCAMBIA1302 that had the GFP gene removed. They are both in GV3101 strain of agrobacteria. His protein is being expressed fine and he got interesting results. However, when he infiltrated just the vectors as controls, the pCAMBIA gave a similar response as his protein but the pSITE did not give any response. A response was not seen in the no infiltration or buffer only conditions. Has anyone else found that the vector backbone alone can alter plant response?
Relevant answer
Answer
Only suggestion I could give is to cut the pCAMBIA into sections and clone into pSITE to find the region that is causative.
  • asked a question related to Buffer
Question
1 answer
Hi flow users/cell masters,
I'm just wondering if anyone here has experience with staining cytoplasmic cytokines after staining extracellular proteins. So basically what I did in my protocol was staining extracellular markers using PE-conjugated and APC/Cy7-conjugated antibodies, then fix/perm my samples using the 1X fix/perm buffer eBioscience™ Foxp3 / Transcription Factor Staining Buffer Set before staining cytoplasmic cytokine using antibodies resuspended in 1X perm buffer.
I didn't see any APC/Cy7+ or PE+ populations in flow, which is weird coz the markers are supposed to be highly expressed. I saw some posts on Reddit that APC/Cy7 is not stable, but I don't know if PE is unstable either. Also, does anybody know if eBioscience™ Foxp3 / Transcription Factor Staining Buffer Set contains methanol? I couldn't find the info anywhere, if it is, it makes sense then... Since PE and APC tandems are not methanol-resistant.
Thanks in advance to anyone who's going to answer this :)
Relevant answer
Answer
You do appear to mention what you are staining for and why you are using this reagent? I find the eBioscience™ Foxp3 / Transcription Factor Staining Buffer Set is very effective at permeabilising cells for nuclear staining. However, I believe it contains formaldehyde and methanol.
I would suggest you try a detergent based permeabilisation - I find fixation with paraformaldehyde and permeabilisation with 0.1<-> 0.5% Saponin works well for intracellular staining. You can commercial reagents which are QC'd eg BD bioscience or make your own.
best wishes
  • asked a question related to Buffer
Question
1 answer
I have a problem and am asking for advice.I am doing WB. For electrophoresis
I use an 8% separating gel and a 4% thickening gel. The electrophoresis has 2 phases : 30 min-90V , 60 min 110V. Electrophoresis buffer from Bio-Rad of composition 10xTris/Glycine/SDS. For transfer I use Bio-Rad's ready-made but diluted Transfer Buffer. I perform a standard transfer to PVDF membranes (30 min). Membranes are incubated with milk and with tris pH=7.6 and tween. Then as primary antibody I use B act polyclonal antibody from Invitrogen, Lot YD371542, as secondary antibody anti-rabbit IgG HRP Conjugated HAF 008 from R&D, Lot FIN 1922041. Then I use Precision Protein StrepTactin-HRP Conjugated 5,000x. And I add calling reagents.
Why does my 70 kDA stain very clearly and my beta actin stain very weakly ( 42-46 kDa) ?
I make WB from homogenised cardiac tissue. To prepare it, I used Thermo Scientific protease inhibitor at 225 microlitres per 25 ml homogenization buffer.
I attach a blot of beta actin below .
Relevant answer
Answer
Thanks for including the blot pic. Your electrophoresis looks very nice. I suspect the StrepTactin reagent is cross-reacting with something 70k in your samples. To investigate this possibility you might try leaving out that step/reagent, or cutting the MW marker band off of your blot and developing it separately.
  • asked a question related to Buffer
Question
1 answer
I wanted to draw standard curve of Novobiocin in phosphate buffer(7.4), and I made 2,4,8,16,32 and 40 microgram/ml. but It did not show any absorbance in UV spectrometer. I want to know its problem.
Relevant answer
Answer
Even around 390nm?
  • asked a question related to Buffer
Question
10 answers
Hello everyone,
I would like to use platelets in a co-culture with other cells.
I was wondering which are the conditions to keep alive the platelets:
-since I am going to buy my platelets, I was wondering if they can be shipped on ice
-which buffer do I need to keep them alive during the shipment?
-I saw some papers where they were kept in culture with RPMI, do you have any suggestion/ideas about that?
Looking for your help
Thank you in advance
Relevant answer
Answer
Dear Giulia,
How is your experiment going?
I hope all things are fine with you. Good Luck.
  • asked a question related to Buffer
Question
2 answers
I'm attempting to separate two DNA oligos of length 120bp and 100bp. I've been running them with 2x formamide loading buffer for 3 hours at 65V. I understand constant power is important for the temperature of the running buffer to help denaturation. My lab only has a constant Volts or Amperes power supply, so this is what I've been using. I've been considering purchasing a more advanced power supply that has constant power. How important is this for overall denaturing PAGE settings? Is there anything wrong with my current settings? My current method semi-works and I'm considering continued troubleshooting or exploring constant power.
Relevant answer
Answer
maintaining constant power is generally preferred over constant voltage. Constant power ensures a more consistent rate of heating throughout the gel, promoting uniform denaturation of nucleic acid samples. This is particularly important when dealing with DNA fragments that require denaturation.
In constant voltage electrophoresis, as the resistance of the gel changes with time due to temperature variations, the power fluctuates. This can result in uneven denaturation and separation of DNA fragments, especially when dealing with samples of different lengths.
Given your current setup with a constant voltage power supply, there are a few considerations:
Gel Composition: The gel composition, including the percentage of acrylamide, the concentration of denaturing agents (such as urea), and the formamide loading buffer, plays a crucial role in denaturation. Ensure that the gel composition is appropriate for your target DNA fragments.
Run Time: Running your samples for 3 hours at 65V suggests an extended run time, which may be contributing to the separation of your DNA fragments. You may want to optimize the run time based on the expected size difference between your 120bp and 100bp fragments.
Temperature Control: Although constant power is preferable, temperature control is critical. Ensure that the temperature of the running buffer is maintained within the optimal range for denaturation. Formamide loading buffer is often used to assist in denaturation by disrupting hydrogen bonds.
Power Supply Upgrade: If possible, consider investing in a power supply that allows for constant power settings. This can enhance the reproducibility and efficiency of your denaturing PAGE.