Science topic

Reagents - Science topic

Explore the latest questions and answers in Reagents, and find Reagents experts.
Questions related to Reagents
  • asked a question related to Reagents
Question
4 answers
Hey,
I would like to know more about HRM analysing. can anyone assist me regarding the analysing the data which were taken from this experiment?!
I have used MeltDoctor reagent and I calibrated quantstudio 3 with HRM plate in advance. Attached you can see the result from my samples.
Thank you in advance.
Fatemeh
Relevant answer
Answer
How big is your HRM amplicon? And what are you targetting? A single SNP? Methylation?? From the melting curves, it looks like your amplicons might be quite large ??
  • asked a question related to Reagents
Question
1 answer
For my protocol, at end-point I have to do tail-vein injections of TRITC-labeled dextran (100 ml of 10 mg/ml) and FITC-labeled lectin (50 mg). However, the protocol I'm following states that they inject one, followed by a 10 min circulation and then the other followed by a 5 min circulation. Tail-vein injections on black mice can be challenging especially two. I know some fluorescence reagents can be mixed but I just want to confirm this can be done.
Relevant answer
Answer
Yes sure
  • asked a question related to Reagents
Question
2 answers
I am currently trying to do chromosome counts on algal cells. However, I don't have the reagents necessary to extract full protoplasts. But I can make small holes which can allow the DAPI stain to get through. Is there a methodology to do chromosome counts with the cell wall still intact?
Relevant answer
Answer
We stain the roots with hematoxyllin (this is mitosis). Everything OK with cell walls. (Dissolve 1 gram of hematoxylin powder in 50 µl of distilled water and leave for 3 days. Then add 50 µl of glacial acetic acid and hold the iron clip for a couple of minutes). Boil the roots in hematoxylin over an alcohol lamp.
  • asked a question related to Reagents
Question
7 answers
I am a first year Ph.D. student in Cell and Molecular Biology department, UT. I am going to start a project on Breast Cancer and for this I need to be skilled on cell culture. I never did cell culture in my undergrad and now after shadowing my PI and seniors in the lab, I started my own session. But unfortunately my cells are getting contaminated. I wash my hands, rub my hand with alcohol, sterilize my hood with alcohol, spray alcohol in every bottle of reagents I use, Using auto pipet ( Big pipet). What else I am missing? I am feeling so pissed off because my seniors are doing the same but they are not facing the contamination. Please help me to overcome this situation.
Relevant answer
Answer
Hello Sadia Haque Khan,
Even if you had the opportunity to work along with members experienced in cell culture, since you are new to cell culture, it will take a while till you develop the skills of handling cultures.
Keep following the sterile practices learnt from your seniors. Observe your seniors when they handle cultures. You should record even the minute details such as the way they open the knob of the culture flask during feeding the cultures or subculturing. For instance, the knob of the culture flask should be kept in such a way that the inner side of the knob faces upwards and does not touch the surface of the laminar hood. The pipet tip should not touch any non-sterile surface, and so on and so forth. You will be able to master these skills with practice.
Till you master these skills, I suggest you add antibiotics to the growth media which will help prevent contamination. Generally, antibiotics are not supposed to be added to cultures. Once you get the confidence of handling cultures without any contamination, you may discontinue the use of antibiotics.
Good Luck!
  • asked a question related to Reagents
Question
2 answers
If an unknown powder or solid sample has given a yellow color result, what INORGNIC or METALLIC COMPOUNDS ARE INDICATED. I am not interested in any controlled organic substances , only inorganic substances. Any shade of yellow qualifies. TY.
Relevant answer
Answer
Salts of palladium and gold I think,
and Inorganic or metallic compounds typically do not produce significant color changes with the Marquis reagent
  • asked a question related to Reagents
Question
1 answer
I am extracting rna, from sheep goat blood with rna later, I use RNeasy Plus Universal Mini Kit (50) however while I get a good ratio of 280/260, the concentration is very low, does anyone know what blood concentrations and QIAzol Lysis Reagent I need to get a good concentration; Thank you very much
Relevant answer
Answer
To improve your RNA yield, ensure you're using an appropriate amount of blood and QIAzol Lysis Reagent. Generally, for blood samples, a starting ratio of 1 mL of blood to 3 mL of QIAzol is recommended. Also, consider optimizing your RNA extraction protocol by adjusting parameters such as centrifugation speed and duration, as well as the volume of elution buffer used. Additionally, factors like the age of the blood sample and the health of the animals can affect RNA yield.
  • asked a question related to Reagents
Question
1 answer
Hello.
I am new to the GSH assay and I am learning about it online. I performed it this week for the first time.
My protocol said to add DTNB and GR to my GSH standards + samples - allow 30 sec the conversion of GSSG to GSH - and THEN - add NADPH and measure absorbance.
Is not NADPH essential for the conversion of GSSG to GSH? Why adding it later?
Also. Can I add just DTNB and measure the absorbance (initial GSH) and then add GR+NADPH and measure absorbance again to measure the total GSH (Initial GSH + reduced GSSG)?
So I can calculate also GSSG (GSHt - GSHi) with this assay (without using vinylipyridine) ?
Relevant answer
Hello Ambra.
Ellman's reagent (DTNB) reacts with any molecule that has a free thiol group (-SH), generating TNB (increasing yellow color), with a gain in absorbance that can be read at 412 nm. Due to the non-specificity of DTNB with the different thiols in your sample extracts (cysteine, GSH, coenzyme A), it is necessary to evaluate glutathione concentrations using an exogenous GR. Depending on the concentration of glutathione in your sample extract, you will obtain a proportional slope (delta A/delta T) on the spectrophotometer. Therefore, in addition to analyzing your samples, it will also be important for you to perform a calibration curve using exogenous glutathione (analytical standard), so that you can later interpolate your data obtained with biological samples using this calibration curve. Answering your questions, the reason why NADPH is the last reagent to be added is precisely because before it is added, all the GSH in your sample will be in the form of GSSG (as it has already been completely oxidized by DTNB). When adding NADPH, GR will begin to convert it into GSH, which then reacts with DTNB until all this NADPH is exhausted, thus generating a slope proportional to the concentrations of its glutathione, as mentioned previously. To be able to differentiate the initial GSH from the initial GSSG, you must perform a new test using vinylpyridine. Vinylpyridine has the ability to complex with the initial GSH, leaving only the GSSG available for you to carry out the cyclic reaction with GR, NADPH and DTNB again. This way, by calculating the concentrations of total glutathione and GSSG, you can determine the initial GSH concentration: Total glutathione = GSH + GSSG. It is important to remember that each GSSG is equivalent to 2 GSH. If you have any specific questions, you can ask them in the comments below.
Best Regards.
  • asked a question related to Reagents
Question
1 answer
Hello to everyone!
For the experts of the DNA fiber technique, I am having issues with my fibers. As you can see in the picture, the fibers are all together as a mass and they have not separated nicely. I have had beuatiful fibers in the past but now I am unable to reproduce them (I have not changed the technique or any single reagent whatsoever!). I am aware that the envrironment does play a role for the experiment so I have tried to conduct the experiment in a warm ish place. It looks like either a) there are too many fibers (they should be 8x105 as many protocols state) or b) the lysis buffer has not worked? I am using the technique from this paper. https://www.sciencedirect.com/science/article/abs/pii/S0076687917301143
The cells that I am using is HeLa, a widely used cell line for this tecnhique. I also make sure that the fibers ''travel'' slow enough (5 mins) along the slide because some papers mention that when the fibers go down quickly, they dont get separated therefore they are not analysable (the problem I have!)
Any help would be much appreciated!
Relevant answer
Answer
I have earlier faced issues of this sort. I still am facing it while performing S1 nuclease DNA fiber variant.
My advisor suggested dilution with unlabelled cells. I tried to dilute the labelled cells with unlabelled cells. It has given me good results with the normal fibre experiments (except S1 nuclease variant, where nuclei tend to clump). You will get better spreads and data if you follow this. Hope this helps.
I would also appreciate if anyone could let me know tricks with the S1 nuclease experiment.
Ganesh Pai B
  • asked a question related to Reagents
Question
2 answers
I am trying to extract DNA from serum. I found an article that claimed simple extraction can be done in a single tube -
Here they used a solution containing 6M NaI/13mM EDTA/0.5% sodium N-lauroylsarcosine/10 µg glycogen as a carrier/26mM Tris-HCl, pH 8.
But currently, I don't have NaI and glycogen. So, I am thinking of making a solution with KI + EDTA + sodium lauroyl sulfate + Tris-HCl, pH 8. And finally, use Na-acetate and absolute ethanol for the precipitation of DNA.
What consideration should I take into account to use alternative reagents?
And, in their protocol does it mean the final solution containing all reagent should have a pH 8 or it just means the use of Tris-HCL with pH 8?
Relevant answer
Answer
NaI can be replaced by KI, since iodide reduce Tm of protein and cause instability of folded protein, which had its purpose for protein denaturation, but potassium may increase the pH. For sodium N-lauroylsarcosine, as long as sodium lauroyl sulfate provide a membrane disruption ability, then the DNA should be released as the same way as original protocol. All your reagents should be in tris-HCl and adjusting to a final pH 8, because in this pH tris has its best buffer ability.
Best
  • asked a question related to Reagents
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 Reagents
Question
2 answers
I’m currently using a protein (36 kDa) which needs to be unfolded during a labelling reaction. unfortunately the protein precipitates completely upon denaturation with EDTA which chelates the zinc ions holding its structure together. I’ve tried the reaction at 37, 40, and 55 degrees Celsius all with the same issue.
The exact same protocol (37 degrees, same edta:zinc ratio, time course) works for smaller constructs of the protein. I typically unfold, reduce, and then add the labelling reagent sequentially for 50 minutes each (total 2.5 hours shaking at 37). My protein concentrations have been between 20 and 200 micromolar, and the pH is maintained at 7.8 in 100 mM ammonium bicarbonate buffer (no salt) for optimal labelling.
I need the protein to remain in solution for downstream experiments after the labelling reaction. How can I denature the protein while keeping it soluble?
The protein pI is 7.06, which may be too close to the reaction buffer, especially considering that the other constructs had pi’s at 5.3 and 6.6. I’m considering testing a higher pH, unfolding with EDTA and a detergent, and increasing the salt concentration. Ideally I’d have as low a salt concentration as possible for downstream mass spec, and maintain the pH between 7.5 and 8.5 for optimal labelling with the different reagents. I’d appreciate any feedback or advice!
Relevant answer
Answer
There are 2 main reasons why the protein could be precipitating: electrostatic interactions or hydrophobic interactions.
Increasing the salt concentration would probably overcome electrostatic interactions. You would have to remove the salt later, which is easy to do by passing the column through a desalting column or by dialysis. Changing the pH by several units could also help, but you have explained why this is a less desirable approach for you.
In my opinion, the more likely reason for the insolubility is hydrophobic interactions, since the interior parts of proteins are hydrophobic. You could probably keep the protein in solution using a detergent, but removing detergent later can be troublesome. There are a few detergents that are compatible with mass spec, however. The other approach would be to add a strong chaotrope, either urea or guanidine-HCl, at several molar. These are small molecules that can be removed later. You would have to experiment to find the minimum necessary concentration to keep the Zn2+-free protein in solution, especially if you don't want to completely unfold the protein, since refolding a completely denatured protein can be a challenge.
  • asked a question related to Reagents
Question
10 answers
The Griess assay is not working for the standard curves. I am using 100uM-1.56uM standards. When I add the Griess reagent the high concentrations are yellow. Lower concentrations go very dark pink and crystals form at the bottom. What am I doing wrong?
Relevant answer
Answer
I have the same problem. Please advice
  • asked a question related to Reagents
Question
1 answer
I have tried isolating ventricular cardiomyocyte by perfusing type II collagenase through adult mice hearts. When looking at the extracted cells under 20X magnification, I don't see the typical rod-shaped cardiomyocytes I was expecting. Instead I seem to have multiple rounder debris. Was I able to extract the myocytes, but they got damaged during the isolation or am I looking at totally different kinds of cells ?
I know immunostaining would be the way to test, however I do not have the reagents on hand.
Relevant answer
Answer
On looking Slide at first instance , it’s is not typical rod cell myocytes. Might be the fault at isolation , proccessing. Better to repeat the sampling
  • asked a question related to Reagents
Question
4 answers
I am planning to synthesize AgNP using plant extract as a bioreductor.
I have come across several papers recommending a neutral to alkaline pH range (pH 7-8) to achieve silver nanoparticles with characteristics such as small size (<100 nm), uniformity, and stability. In green synthesis approaches, is it acceptable to use chemical reagents to adjust the pH, or are there alternative methods available?"
I would greatly appreciate any insights or advice on these questions. Thank you in advance for your help.
Best regards,
Relevant answer
Answer
You can try to achieve this pH by increasing the temperature, pressure, or adding another extract.
  • asked a question related to Reagents
Question
4 answers
I am planning to synthesize AgNP using plant extract as a bioreductor. In the green synthesis approaches of silver nanoparticles, should I focus solely on preventing the degradation temperature of reducing metabolites such as phenolic compounds?
I would greatly appreciate any insights or advice on these questions. Thank you in advance for your help.
Best regards,
Relevant answer
Answer
Silver ions are well reduced at room temperature. You are right, you can try to optimize by increasing the temperature or using an autoclave, if you need to develop the necessary production technology.
  • asked a question related to Reagents
Question
4 answers
Hello,
I am currently attempting to obtain amplifications of SoxC from a species of onychophoran belonging to the Peripatidae family using primers designed from a sequence of its sister family, Peripatopsidae. The sample I am using is cDNA, and I have tried different concentrations of reagents and cycling times in the thermocycler. However, I have not obtained any amplification yet. I would like to ask how you would start standardizing reagent concentrations and the thermocycler program for a set of primers generated from the sequence of another species for a conserved gene like SoxC (700 bp amplicon).
I have COI as positive, and before using any cDNA for SoxC, COI was amplified.
Due to resource optimization, the primers I use for SoxC had to be initially designed with the sequences of the promoters SP6 and T7.
So these are my primers:
SoxC-F5: ACGCCAAGCTATTTAGGTGACACTATAGGCGGCTACGGATCTTACACA
SoxC-R5: CAGTGAATTGTAATACGACTCACTATAGGGGGGAAATCAAAGTGCGAGCC
none exceed 50% CG.
Additionally, I am conducting tests with cDNA extracted from different tissues and stages to increase the likelihood of finding my sequence, but I still fail to obtain amplification.
Relevant answer
Answer
I would start by amplifying a common housekeeping gene to check that there is cdna and that it can be amplified. You can waste a lot of time worrying about the pcr conditions when actually the problem may be at the template part of the process. Then I would start with a low annealing temperature (8c below the melting temperature) and too many cycles (36) because it is easier to clean up a working but dirty pcr than to guess where the problem might be when there is no product, Check if the template sequence is very high GC or AT in which case secondary structure may be a problem and a high GC buffer ( betaine plus dmso) may help and also check if any known snpd exist under the 3' ends of either primer sequence in your species. If the primers are old and inherited from another researcher they may be degraded so try amplifying a gdna sample just to prove that the primers can work if the product is not too large
  • asked a question related to Reagents
Question
5 answers
I am working on expression and purification of one cytoplasmic protein with His tag, in E coli Bl21(DE3) host cell. Here is SDS picture. Line 2 and 9 are cell lysate, 3,4,7,8 wash steps and 5,6 are elutes using different purification procedure. The expected size is 48 kDa. For protein extraction I used a high pressure homogenizer, also I didn’t use any inhibitors. I was told to try to use Bugbuster protein extraction reagent supplemented with benzonase, do you think it might help?
Relevant answer
Answer
Proteins don't always run at the expected molecular weight on SDS-PAGE. I'd suggest sending a sample of the mostly purified protein off to a mass spec lab to determine the molecular weight of the whole protein.
Meanwhile, it wouldn't hurt to include protease inhibitors when you lyse the cells. There might be a protease-sensitive site near the N- or C-terminus.
  • asked a question related to Reagents
Question
2 answers
We used to use the GenomONE HVJ-Envelope reagent from Cosmobio for SCNT / cell fusion experiments. It's been discontinued for years, and they don't know if it's ever coming back. Do you know of a commercial source? Do you have practical experience in making this reagent? Do you have some leftover that you don't need? We can pay well for all of the above.
Relevant answer
Answer
Sorry I didn't. We're going to draw down our last stocks of it soon. Then we will have no choice but to hire a full time biochemist to make it... I'll be sure to check back into this thread, if we're ever successful with that.
  • asked a question related to Reagents
Question
2 answers
I am measuring primary aromatic amines by UV-VIS and using the NEDA solution as my coupling reagent. The method says that the reagent needs to be made fresh every day. However, the solution is sold commercially from 0.1-1.0% with a shelf life of 6-12 months. Why does it have to be made fresh? What is its stability? Is it a temperature or UV dependent reaction? Can storing in a brown(amber) bottle and/or refrigerating allow for longer use?
Relevant answer
Answer
In such cases, it is preferable to prepare it daily and avoid its instability as a result of it being affected by various conditions such as light, temperature, etc. It is best to prepare the amount as needed daily
  • asked a question related to Reagents
Question
2 answers
Besides extracting the poorly crystalline Fe- and Al-(hydroxy)oxides, does 0.2 (M) ammonium oxalate-oxalic acid (pH 3.25) buffer extract poorly crystalline Mn-oxides too?
Relevant answer
Answer
Aromatic nitriles are versatile intermediates in organic chemistry, serving as precursors to amines, amides, and carboxylic acids among other functional groups. The selective reduction of these nitriles to primary amines in the presence of other functional groups like OH requires careful consideration of reagents and conditions to achieve high selectivity and yield. The challenge is compounded by the resonance stabilization of the nitrile group when attached to an aromatic system, which can impede reduction. Additionally, the presence of an OH group introduces the risk of over-reduction or side reactions, necessitating a strategy that can accommodate both functionalities without necessitating protection of the OH group.
Methodologies for Reduction
1. Catalytic Hydrogenation
Catalytic hydrogenation using hydrogen gas (H₂) and a palladium catalyst (Pd/C) is a widely adopted method for reducing nitriles to primary amines. However, the effectiveness of this method in the presence of an OH group and an aromatic nitrile can vary based on the substrate and catalyst used. Palladium on carbon (Pd/C) under mild conditions can offer a route to selectively reduce the nitrile without over-reducing the OH group, though the reaction may require optimization of pressure and temperature to achieve the desired selectivity (Rylander, 1979).
2. Chemoselective Reduction Agents
Selective reduction agents such as Nickel Boride (Ni2B), formulated by the in situ reaction of Nickel Chloride (NiCl₂) with Sodium Borohydride (NaBH₄), have shown promise in reducing nitriles to amines while preserving other functional groups like OH (Girard et al., 1998). This method leverages the chemoselectivity of nickel boride for the nitrile functionality, potentially offering a viable pathway for selective reduction in complex molecules.
3. Transfer Hydrogenation
Transfer hydrogenation represents an alternative strategy, using formic acid or ammonium formate as hydrogen donors in the presence of a suitable catalyst, such as Ru, Rh, or Ir complexes. This approach has been successful in reducing aromatic nitriles to primary amines under relatively mild conditions, with the potential for high selectivity (Casey et al., 2007). The compatibility of transfer hydrogenation with various functional groups, including OH, makes it an appealing option for selective reductions.
4. Avoidance of Protective Groups
The strategic avoidance of protective groups is a guiding principle in the design of reduction methodologies. While protective group strategies can offer a route to selective reductions, they introduce additional synthetic steps, increasing the complexity and time required for the synthesis. The methods outlined above represent approaches that, in principle, obviate the need for protecting the OH group, thereby streamlining the synthetic process.
References
  • Rylander, P. N. (1979). Hydrogenation Methods. Academic Press.
  • Girard, C., Onen, E., Aufort, M., Beauviere, S., Samson, E., & Charette, A. B. (1998). Nickel Boride, a Versatile Reducing Agent. Journal of Organic Chemistry, 63(23), 8108-8109.
  • Casey, C. P., Singer, S. W., Powell, D. R., Hayashi, R. K., & Kavana, M. (2007). Transfer Hydrogenation Catalyzed by Chiral Rhodium Complexes: Enantioselective Reduction of Aromatic Nitriles. Journal of the American Chemical Society, 129(20), 6477-6484.
  • asked a question related to Reagents
Question
1 answer
Dear colleagues, help me figure this out.
We are trying to analyze and sort antigen-specific cells using Flex T technology (Biolegend). After UV exchange, the efficiency is 60%. Next, according to the manufacturer’s protocol, we separate and carry out conjugation with streptavidin-PE and steptavidin-APC. And when using these reagents, we obtain a high level of nonspecific staining for each of the fluorochromes.
I can’t figure out why there could be such pronounced non-specificity and how to deal with it. I would be grateful for any suggestions)
Relevant answer
Answer
Flex-T analysis is a method for identifying and characterizing antigen-specific T cells. Here's a breakdown of the process:
1. Sample preparation:
  • Peripheral Blood Mononuclear Cells (PBMCs): Blood is drawn from the patient, and PBMCs are isolated. These cells contain various immune cells, including T lymphocytes.
2. Antigen stimulation:
  • Specific antigen: The isolated PBMCs are exposed to the antigen of interest. This can be a pathogen-derived peptide, tumor antigen, or any molecule against which the immune response is directed.
3. Expansion of antigen-specific T cells:
  • Cytokine cocktails: The PBMC culture is supplemented with specific cytokines (signaling molecules) that stimulate the growth and proliferation of T cells that recognize the introduced antigen.
  • Bead-based enrichment: Magnetic beads coated with antibodies against activation markers on T cells can be used to enrich the population of antigen-specific T cells.
4. Detection and characterization:
  • Flow cytometry: Cells are stained with fluorescent antibodies specific for various surface markers on T cells. This allows identification of the expanded population of antigen-specific T cells based on their unique marker expression profile.
  • Functional assays: Proliferation assays or cytokine secretion assays can be performed to assess the functional capacity of the expanded T cells. This helps determine their ability to respond to the specific antigen.
Additional considerations:
  • Major histocompatibility complex (MHC) tetramers: These reagents can be used to directly identify and quantify antigen-specific T cells based on their interaction with the specific antigen presented in the context of MHC molecules.
  • Next-generation sequencing: This technique can be employed to analyze the T cell receptor (TCR) repertoire of the expanded population, providing insights into the diversity of antigen-specific T cells.
Resources for further information:
  • A detailed protocol for Flex-T analysis can be found in the scientific literature. Search for research articles containing keywords like "Flex-T analysis" or "antigen-specific T cell expansion."
  • Consider consulting with an immunologist or a flow cytometry core facility for guidance on implementing Flex-T analysis in your research setting.
Important Note: Flex-T analysis is a complex technique requiring expertise in cell culture, flow cytometry, and immunological assays.
  • asked a question related to Reagents
Question
1 answer
Dear All,
We recently purchased Bradford's reagent from SRL. We didn't know the concentration of the solution, if it is 5X or 1X. Nothing is mentioned on the bottle. Also, how to standardize using BSA, how much concentration of Bradford's reagent to use. Please help. Thank you.
I have attached the picture for reference.
Relevant answer
Answer
I would start by guessing that it is 1X, since no other information is available. You could try using the method given in the original paper by Bradford. https://hoffman.cm.utexas.edu/courses/bradford_assay.pdf
I use a microplate version, in which a 10 µl sample in water or a non-interfering buffer is mixed with 200 µl of 1X Bradford reagent, and read at 595 nm after 5-10 minutes. The amounts of BSA depend on the quality of the Bradford reagent, but I use either 0.5-5.5 µg or 1-11 µg to make the standard curve. I fit the absorbance vs. µg BSA data with a 2nd order polynomial, but a linear fit may work well, especially for the lower concentration range.
  • asked a question related to Reagents
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 Reagents
Question
2 answers
I am differentiating monocytes into macrophages from PBMCs, and every time I have encountered these bacillus-shaped elements when I looked them in the microscope at 40x. They are not typical bacteria because I have taken the supernatant from some wells and incubated it with LB medium, but nothing grew. Additionally, the medium did not turn yellow. I have seen it in different cultures in which I used different reagents and donors. Cells do not look atypical.
I left you a video of the culture
Could anybody help me to identify this elements? Are they normal? Some advice?
Thank you very much!
Relevant answer
Answer
Thank you very much B. C. Menarim !
We are taking your suggestion
  • asked a question related to Reagents
Question
2 answers
I'm currently trying to make a standard curve for potassium ion detection. For my reagent I'm using 25 mM of sodium cobalthexanitrite with potassium dihydrogen phosphate as a substrate in a 40:60 ratio and measuring at 420nm after incubating the mixture in the dark for 10 minutes. Unfortunately, it's not really working and I'm not getting any precipitation. Is there something more I need to add or a step I've missed? Is my wavelength incorrect?
Relevant answer
Answer
Adam B Shapiro thanks this looks very helpful for me
  • asked a question related to Reagents
Question
2 answers
while performing Bradford assay , I noticed the OD of water plus Bradford reagents was higher then the Bradford alone. So Is there any possibility that if by chance water is contaminated then is there a chance that it will react with Bradford reagent and give different absorbance?
Relevant answer
Answer
The spectrum of the Bradford reagent is affected by dilution. This is probably why you saw the effect of water. When performing the Bradford assay, it is important to make sure that all samples have the same volume, so that the dilution of the Bradford reagent is the same in all cases.
  • asked a question related to Reagents
Question
1 answer
I have three SDS classifications which are Sodium dodecyl sulphate, ACS reagent, ≥99.0%, Sodium Dodecyl Sulphate, >85.0% (T), Sodium dodecyl sulphate, >97.0% (GC)(T). Could somebody tell me which SDS is appropriate for nickel composite coating? The goal is to prevent particle agglomeration
Relevant answer
Answer
The question is: what are the contaminants in the less pure species and are their compositions reproducible? Generally, I would suggest to make the first experiment with a small batch of the highest purity and then see if the lower purity also does the job with the perspective for upscaling, but if the contaminants are not reproducible, the whole coating quality probably won't be.
  • asked a question related to Reagents
Question
2 answers
Hello,
I have some problem when using RIPA buffer during reading BCA assay. I'm getting high value of the RIPA's absorbance so I'm getting negative values.
I have Standard curve diluted with PBS and 100ul samples with 200ul of RIPA(X2) and proteinase inhibitor (0.5ul). Then taken the 25ul for the assay plate+200ul WR (reagent A+B).
What is the right blanks for me? (should I substract PBS value from the standard curve absorbance and substract RIPA+PBS+PI from the unknown samples absorbance value?)
Maybe:
Blank 1 - PBS (for standard curve)
Blank 2 - 100ul PBS (instead the sample) + 200ul RIPA(X2) + 0.5PI (for unknown samples)
What can be the problem? and do you have some suggestions how to calculate it?
Also what is the right graph type? 4PL/point to point/linear regression?
Thank you!
Relevant answer
Answer
The BCA assay is not compatible with reducing agents. If there is dithiothreitol, mercaptoethanol, or other reducing agent in your samples, it will interfere. See the product literature for a compatibility table.
The blank should ideally be of the same exact composition as the sample, so it should include RIPA(X2) and protease inhibitors in the same proportion as the samples.
  • asked a question related to Reagents
Question
4 answers
I am working on the effect of heavy metals on plants. However, to check the activity of antioxidant enzymes I need to prepare reagents in bulk. Buying an assay kit is not affordable. Can someone kindly suggest a protocol for reaction mixture (reagents) for the above-mentioned assays (Please specify the volume of each reagent and its concentration)
Relevant answer
Hi Akshatha Banadka , did you get the protocols? If yes, could share with me, please?
  • asked a question related to Reagents
Question
1 answer
Crosslinking on surfaces makes the enzyme reusable multiple times, I wish to know whether this is possible for a chemical reagent. Once fixed or coated on a surface, can the reagent be used multiple times for a microassay? And if this can be done, please direct me to a source. A research paper/any kind of literature would be helpful.
Relevant answer
Answer
Industrial chemistry often uses immobilized enzymes for synthesis. See https://en.wikipedia.org/wiki/Immobilized_enzyme.
  • asked a question related to Reagents
Question
7 answers
I use a DNA/RNA synthesizer from LGC (Biosearch), a MerMade6, to make oligos. The machine has operated for around 16 months and produced about 1500 oligos during this period. As you can see, it is not a heavily used machine.
All preventive maintenances were ok, and the machine was operating just fine and yielding very good quality oligos......
But then it started to present a misalignment between the cart/columns block and the capilaries, pouring reagents out of the columns.
If you look closely you can see that, when the cart with the columns moves and gets to the position assigned, it doesn't stop completely, but slides juuuust a little bit instead. Such a mess!
Calibration doesn't solve the problem: after a calibration, reagents will be delivered correctly into the columns in the first cycle, but as with every move the cart slides a little bit, the misalignment starts to sum up with every move and, at the 3rd cycle, reagents are being spilled outside the columns already.
We are in Brazil. Remotely, LGC team couldn't help. They couldn't even find the problem. The visit of LGC engineering team will coast a lot, and while we are working in resources an bureaucracies to get them here, tips from other users would be welcome and much appreciated.
This machine works with step motor / step motion, which is similar to printers and a lot of other machines. Have you ever seen this kind of issue?
Alternatively, do you know other MerMade users you can put me in touch with?
Thanks in advance
Relevant answer
Answer
Thanks, Andrei!
I certainly did! I took pictures and I have a meeting with them scheduled for tomorrow!
Maybe they won't replace it because the machine is out of warranty... But I'll make some noise anyway, that brought us a lot of trouble!
  • asked a question related to Reagents
Question
5 answers
I want to prepare a cell-free system by following the protocol shown at the bottom, in which the tRNA from E. coli (Catalog number: MRE600 provided by Roche Applied Science) is an essential reagent. However, Roche seems to no longer provide this reagent. Therefore, I just wonder if there are any substitutions to this compounds so that I could prepare the cell-free system. Thanks!
Relevant answer
Answer
Did you find another product? I'm on the same search!
I found this at Sigma: Product R1753 (https://www.sigmaaldrich.com/DE/de/product/sigma/r1753)
Thinking about ordering it, although the description makes me feel uncertain.
Best wishes!
Christina
  • asked a question related to Reagents
Question
3 answers
We have expressed and purified the recombinant viral protein found in the insoluble fraction. Please suggest some methods or reagents to solubilize the protein.
Relevant answer
Answer
You could adding NaCl (100-250 mM) to the culture medium and/or modify agitation speed.
Good luck
Maura
  • asked a question related to Reagents
Question
5 answers
We have expressed and purified the recombinant viral protein found in the insoluble fraction. Please suggest some methods or reagents to solubilize the protein.
Relevant answer
Answer
If the protein is in the insoluble fraction because it did not fold properly, which often happens, then it will probably be necessary to completely denature it with concentrated urea or guanidine-HCl, then refold it using any of several techniques. As this process can be difficult to achieve without several experiments, it is often worth spending some time to try to avoid the problem in the first place.
Sometimes, a portion of the expressed protein can remain in the soluble fraction if the expression conditions and/or the expression strain are changed. One approach that sometimes works is to lower the expression temperature. as low as 16oC. This slows down expression, possibly allowing time for the some of the protein to fold properly instead of aggregating.
  • asked a question related to Reagents
Question
1 answer
Is there any recommended protocol (reagents' concentrations) for RT-qPCR in human postmortem brain tissue for the identification of NR2A (NMDAR) subunit? I cannot find neither a protocol let alone ranging concentrations.
Relevant answer
Answer
I have a protocol for mouse brain tissue and NR2A. Would this work for you?
  • asked a question related to Reagents
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 Reagents
Question
3 answers
I have performed 3 RNA isolations on Jurkat cells using the phenol-chloroform principle, using Omega Bio-Tek RNA Solv® & following the protocol. I took care in not disturbing the separated phases when removing two thirds of the aqeous phases but the data I get when I measure the 260/280 & 260'/230nm absorbance ratios lead me to suspect contamination of the samples with phenol or other reagents.
Most of the samples have a 260/280 ratio below 1,6 and only one sample has a 260/230 ratio that comes close to 2 (1,84 to be exact). Subsequent cDNA synthesis & RT-qPCR have not resulted in genes of interest to show any fluoresence at all, only the housekeeping genes in 2 of 7 samples showed up. Problems with primers & RT-qPCR were ruled out as the same setup yielded viable results previously.
Thanks in advance!
Relevant answer
Answer
I agree with Julie Ann Dougherty . RNA quality can be affected by impurities like phenol which can be carried over to samples. Residual phenol is assumed to inhibit PCR reactions by denaturation of enzymes such as polymerases and reverse transcriptase in a concentration dependent manner. So, try to use lower concentration of RNA than what you would be presently using.
You may want to refer to the articles attached below for more information.
Best.
  • asked a question related to Reagents
Question
3 answers
Hi all,
I did a mock transfection using pGL3 vectors with different promoter inserts (plasmids range from 10 to 14kb). However, I have gotten unexpectedly low luminescence ratios (firefly luc / renilla luc) and I was wondering if it would be logically sound to transfect mols of DNA instead of a certain weight for all of them (eg. 200ng per well). Thoughts?
Thanks!
Relevant answer
Answer
No, I haven't. But it is logically sound.
I would just use ~40% more µg of the 14kb plasmid, compared to the 10kb version....
  • asked a question related to Reagents
Question
4 answers
I intend to do a chemical reaction and have seen that ethanedithiol would be a good reagent, however, I do not know how it smells.
Relevant answer
Answer
thank you so much!
  • asked a question related to Reagents
Question
2 answers
I would like to know if it is a common reagent kit for the target amplification of both DNA and RNA, or does it show amplification only for the RNA targets. Will the presence of DNA in the sample affect the reaction and its results?
Relevant answer
Answer
YES, you can, just before starting you have to add a step of RT deactivation (e.g. 94°C for 10 min) at the beginning of your PCR thermoprofile. However, the cost of the run will be more expensive when using RT-mastermix than ordinary PCR-mastermix.
  • asked a question related to Reagents
Question
2 answers
..
Relevant answer
Answer
  • Nucleic acids: These are the building blocks of DNA and RNA viruses, such as herpes simplex virus (HSV), hepatitis B virus (HBV), and human immunodeficiency virus (HIV). Nucleic acids can act as scaffolding proteins or additives for virus-like particles (VLPs), which are virus-derived structures that mimic the form and size of a virus particle but lack the genetic material1. Nucleic acids can also form complexes with viral proteins or nucleic acids to facilitate their entry into host cells.
  • Glycans: These are the sugar chains attached to proteins or lipids on the surface of viruses or host cells. Glycans can affect the release of virions from infected cells by modulating their binding affinity to receptors or enzymes. Glycans can also influence the recognition and clearance of viruses by immune cells by altering their antigenicity or immunogenicity.
  • Peptides: These are short chains of amino acids that can fold into specific shapes and interact with various molecules. Peptides can be designed to mimic or enhance certain features of viruses, such as their envelope glycoproteins, their capsid proteins, or their transmembrane proteins. Peptides can also be used to form fibrils with high stability and mechanical strength.
  • Extracellular vesicles: These are small membrane-bound particles that are released by cells into their surroundings. Extracellular vesicles can carry various molecules from one cell type to another, such as nucleic acids, proteins, lipids, or metabolites2. Extracellular vesicles can act as carriers or mediators for viral infection by transferring viral components from infected cells to uninfected cells.
  • asked a question related to Reagents
Question
3 answers
basically, we are conducting a electrocoagulation experiment and will be using this process to reduce nitrate to a certain level. However, we are struggling on what reagents to use since the nitrate checker from HANNA instruments is not an option since it can only detect from 0 to 75ppm. In the nitrate checker from HANNA instruments, they used malonic acid for their reagent but I'm not sure if its still useable at higher concentrations of 75ppm or do we use Griess reagent?
Relevant answer
  • asked a question related to Reagents
Question
3 answers
I have access to simple reagents,LB, spectrophotometer etc. Can I determine the phosphate solubilization rate using these. Also please explain the procedure so that I can understand easily.
Relevant answer
Answer
I've never done this, but I can think of a pretty easy way to do it, if you are content to measure the concentration of phosphate ion (PO43-), or phosphate+pyrophosphate, in the medium.
At various time points, centrifuge the culture to pellet bacteria and remaining insoluble phosphate. Prepare serial dilutions of the supernatant in water and measure the concentration of phosphate ion using any of the many versions of colorimetric molybdate-based phosphate detection reagents (easily prepared or purchased).
To also measure the concentration of pyrophosphate, precede the measurement step by including the enzyme inorganic pyrophosphatase (commercially available) to convert pyrophosphate ion to 2 phosphate ions.
If you are also interested in measuring organic phosphate, it requires first treating the supernatant with hot sulfuric acid and peroxide to convert organic phosphates into inorganic phosphate ion. This is a rather nasty procedure, best avoided if unnecessary for your purpose.
  • asked a question related to Reagents
Question
3 answers
I want to measure the concentration of cobalt(II) and cadmium(II) with UV-visible.
Relevant answer
Answer
i suggest you use XRF for determination of cobalt and for cadmium you can use FTIR and raman az well. but depends on the condition of their bonds in molecule they need diferent methods
  • asked a question related to Reagents
Question
2 answers
I want to know what are the alternatives for Ficoll- paque plus. Can we use sucrose/percoll/Na metrizoate/Caesium chloride for Monocyte isolation
Relevant answer
Answer
Thank you
  • asked a question related to Reagents
Question
1 answer
Why sodium hydroxide can cause increasing the fluorescence intensity for a fluorogenic reagent however acetone completely diminish fluorescence peak of the same reagent?
Relevant answer
Answer
The effect of NaOH is probably due to alkaline hydrolysis of the bond linking the fluorophore to the rest of the fluorogenic reagent, generating the fluorescent product.
  • asked a question related to Reagents
Question
1 answer
We have to add specific molar concentration to the media (10^-6M). it is labelled as 1000X how can we convert it.
Relevant answer
Answer
If it is labeled as 1000X, it is intended to be added at 1 part per 1000 by volume. There is no way to convert to molarity unless the concentration in molarity or g/L is stated on the package or in the product information.
  • asked a question related to Reagents
Question
1 answer
Using Dimethyl formamide as dissolv reagent. The Eclipta prostrata powdered extract has been purchase.
Relevant answer
Answer
You can prepare the Liposomes with the active compound and the nanoparticles by ultrasonication or extrusion, and add the polymer during annealing.
Or if you first encapsulate the active compound with PGLA, form Liposomes by extrusion integrating np and PGLA
  • asked a question related to Reagents
Question
1 answer
I've been trying to replicate aqueous extractions using MTBSTFA and running on the GC-MS (I've done this before and it had worked). My solvent blanks are clear however it seems whenever I add MTBSTFA there is a lot of contamination peaks (both in a standard test and in PB). Could this possibly be a column issue? It's fairly old and has been used with other derivatization agents. I've tried 3 different MTBSTFA reagents and acetonitriles as I initially thought that might be contributing but all the results have been the same.
Relevant answer
Answer
Did you check or replace the liner and septa with the new ones? Is the liner inert and clean and septa is robust?
Since you indicated the solvent and system blanks are okay maybe interrogating the MS spectrum using peak annotation of each peak can indicate whether it is a column bleed derived, liner, or septa issue. Each mass (peak) gives clues about the problem...
On the other hand, BSTFA derivatizations are not very compatible with aqueous mediums. Hydrogen bonding affinity presented by alcohols and water can disrupt the desired reaction. Therefore it is advisable to perform a reaction in a hexane-like hydrophobic solvent environment. Otherwise inefficient reaction yield and unwanted impurities could be observed.
  • asked a question related to Reagents
Question
8 answers
I try to find out microstructure of 17-4 PH stainless steel. I have used two etchants, they are
1. Kalings reagent
2. Fry's reagent
Sample was immersed for 45 sec in each case.
I have added some pictures for reference. Picture 2 and 3 are images from Fry's reagent and pictures 4, 5, 14 and 15 are from Kalings reagent. These are the microstrctural images of solution treated specimen not aged. As 17-4 PH is a martensitic precipitation hardening stainless steel. kindly tell me whether it is correct or not and also recommend me any other etchant for microstructural study.
Relevant answer
Answer
Hello R. Saxena , I hope you got your microstructure.
I am also working on 17-4 PH SS could you please help me in suggesting etchant with time for microstructure revelation of 17-4 PH SS, I am also facing same problem.
  • asked a question related to Reagents
Question
10 answers
I use restriction enzyme cloning method, and I have been using new reagents e.g., competent cells, and ligase reagent because initially I thought the problem is that these reagents were outdated. Now with these reagents I attempted to clone my shRNA into the vector with 1:3 DNA:insert ratio, but I didn't even see a single colony.
Relevant answer
Answer
I've never tried to anneal oligos without salts. You can easily make appropriate buffer in-house by just searching for "annealing buffer". Good luck!
  • asked a question related to Reagents
Question
1 answer
Dear colleagues, I would like to load an aminoacid on Wang-Harz resin. Most protocols are using HOBt in order to activate the aminoacid. Would be possible to use PyBoP reagent for this purpose? Did someone try such coupling strategy? If yes, could you share a well-established protocol.
Thank you in advance,
Robert Gradinaru
Chemistry, UAIC, Romania
Relevant answer
Answer
Dear colleagues,
The question was trivial.
ByBoP is a form of HOBt.
We have try it and it is working.
I hope that will be helpful for other researchers in SPPS.
Kind regards,
Robert Gradinaru
Chemistry, UAIC, Romania
  • asked a question related to Reagents
Question
1 answer
Hello, I am working with the aforementioned reagent, but my problem arises that the reagent does not indicate the quantity of particles that come per vial, that is, the number of particles, it only gives me the concentration, which is 1 mg/ml in 2 ml. . My question is if anyone has worked with this reagent and can tell me the number of particles.
Thank you so much
im really sorry for my grammar, english is not my first language.
Relevant answer
Answer
Paulina Troncoso Hola! You need to know or measure the particle size distribution to be able to calculate the number of particles/mL in your system.
  • asked a question related to Reagents
Question
3 answers
What are the best chemical reagents (i.e. spray reagents) that can be used on TLC plates to detect the phytochemical constituents of each of the extracts mentioned in the previous question?
Relevant answer
Answer
There are several chemical reagents commonly used to detect phytochemical constituents on TLC (Thin Layer Chromatography) plates. The choice of reagent depends on the type of compounds being analyzed and the specific functional groups or classes of compounds being targeted. Here are some commonly used spray reagents for TLC analysis of phytochemical constituents:
  1. Vanillin-Sulfuric Acid: This reagent is used to detect a wide range of compounds, including flavonoids, phenolic compounds, and alkaloids. The TLC plate is sprayed with a solution of vanillin in sulfuric acid, followed by heating. The presence of compounds results in the development of colored spots.
  2. Anisaldehyde-Sulfuric Acid: This reagent is particularly useful for detecting terpenoids and steroids. The TLC plate is sprayed with a solution of anisaldehyde in sulfuric acid and then heated. The presence of compounds leads to the formation of colored spots.
  3. Natural Products-Specific Reagents: Various reagents are specifically designed to detect certain classes of phytochemicals. For example:Dragendorff's reagent: Used to detect alkaloids; it forms orange to brown spots. Liebermann-Burchard reagent: Used to detect steroids; it produces a range of colors, including green, blue, or purple. Bornträger's reaction: Used to detect flavonoids; it forms yellow or orange spots.
  4. Ninhydrin: Ninhydrin is commonly used to detect amino acids and amines. The TLC plate is sprayed with a solution of ninhydrin, and upon heating, the presence of compounds results in the formation of purple or blue spots.
  5. Iodine Vapor: Iodine vapor is often used as a general indicator for the presence of compounds on TLC plates. The TLC plate is placed in a chamber with iodine crystals, and the iodine vapor reacts with compounds to produce visible spots. Different compounds exhibit varying degrees of color development.
It's important to note that the choice of spray reagent depends on the specific analytes being targeted and the desired detection sensitivity. Additionally, it's recommended to use appropriate safety measures and handle these reagents with care as some of them can be hazardous. It's always a good practice to consult
  • asked a question related to Reagents
Question
2 answers
Hi Everyone,
I have difficulties with my current work analyzing S-Methylmethionine(SMM).
According to several reports, the analysis of SMM can be done by HPLC with a UV detector (338nm) and OPA reagent. Meanwhile, there is another report showing SMM still can be analyzed without OPA. Later on, I tried to follow the report but I failed to obtain the SMM peak chromatogram.
FYI, the system that I used in my lab:
- HPLC (THermo vanquish)
- UV-DAD detector
- Zorbax AAA column
- 40mM NaH2PO4 (mobile phase A)
- MeOH:ACN:Water (45%:45%:10% mobile phase B)
Please let me know If you guys have any information or experience about analyzing SMM without OPA.
Thank you
Relevant answer
Answer
Since my lab have no OPA and the order is still in process, thus, I came up with the idea to find another possibility to analyze SMM without OPA.
I found several references that analyzed SMM without OPA, I followed those papers but I'm still failed.
  • asked a question related to Reagents
Question
1 answer
Hello, I hope you are well, I have been searching and I cannot find the components to make an RNase decontamination reagent in the laboratory, without having to buy the RNase away.
Thanks
Relevant answer
Answer
I am keeping quite well. Hope you are well too.
You could succeed in making RNase decontamination reagent in the laboratory. Follow the link provided below.
Best.
  • asked a question related to Reagents
Question
1 answer
Hello!
I am relatively new to what I'm currently working with and as the title suggests, I am currently having some issues with my experiment (viral titration).
So, what I have been doing is the following:
1) Seeding cells on a black grenedier 96-well-plate, by extracting cells from culture flasks with Trypsin, counting and diluting them to a sufficient concentration for seeding.
2) The day after, a virus dilution series was performed in infection medium, and the cells in the grenedier well plate were infected in different concentrations, including negative controls.
3) After incubation, all the cells were fixed using the reagents: PFA, Methanol, PBS.
4) After fixing, the plates were stained using a primary, and a secondary antibody attached to a fleurophore. The primary binds to a viral protein of interest. The antibodies were diluted in PBS, and the second one also with Hoechst. The staining was performed with standard conditions, including incubation and washing 3 times with PBS after each antibody. The plate was stored in a fridge over night before analyzing it.
5) The infection was visualized using a Cytation 5 plate reader.
Now, the issue Im facing is that I have kept seeing some infection in my negative controls, (which should have un-infected cells only), according to the images which showed the flourescent dye in these wells. I am trying to figure out what I am doing wrong.
I have been pipetting from lowest to highest concentration, when adding media or PBS, and used separate liquid plastic containers for the control-wells and the infected ones, to avoid contamination. I am going to redo it once again using new infection media, in case the old flask had contamination.
But apart from this, what other factors could possibly help explain why I keep seeing flourescence from the secondary antibody in my negative controls? ANY type of mistake I could possibly have been making I would love to have pointed out. Or other factors which could cause contamination in the lab.
Could PBS/the other reagents possibly be contaminated as well?
Could it be I am not washing away secondary antibodies sufficiently?
Any tips would be much appreciated!!
Relevant answer
Answer
There several possibilities:
  • Contaminated reagents/solutions - As you mentioned, if the infection media or PBS was contaminated previously, this could lead to fluorescence even in uninfected cells. Always best to use fresh solutions when possible.
  • Antibody binding non-specifically - The secondary antibody may be binding non-specifically to cells, even without the primary antibody present. Try using a higher concentration of serum (e.g. 5-10%) in your antibody diluents to block more effectively.
  • Incomplete washing - Residual antibodies left over after washing could lead to background fluorescence. Be very thorough when washing, using plenty of wash buffer each time.
  • Cross-contamination - As you noted, be very careful pipetting from low to high dilution to avoid carryover. Also change pipette tips between samples.
  • Autofluorescence - Some cells have natural autofluorescence. Make sure you have proper negative staining controls with no primary antibody to distinguish real signal.
  • Plate reader settings - Background fluorescence could be exaggerated by certain gain/exposure settings on the plate reader. Optimize the settings using your negative controls.
  • Cell density - Higher cell densities can increase background. Try plating fewer cells if possible.
  • asked a question related to Reagents
Question
1 answer
Hello!
I am relatively new to what I'm currently working with and as the title suggests, I am currently having some issues with my experiment (viral titration).
So, what I have been doing is the following:
1) Seeding cells on a black grenedier 96-well-plate, by extracting cells from culture flasks with Trypsin, counting and diluting them to a sufficient concentration for seeding.
2) The day after, a virus dilution series was performed in infection medium, and the cells in the grenedier well plate were infected in different concentrations, including negative controls.
3) After incubation, all the cells were fixed using the reagents: PFA, Methanol, PBS.
4) After fixing, the plates were stained using a primary, and a secondary antibody attached to a fleurophore. The primary binds to a viral protein of interest. The antibodies were diluted in PBS, and the second one also with Hoechst. The staining was performed with standard conditions, including incubation and washing 3 times with PBS after each antibody. The plate was stored in a fridge over night before analyzing it.
5) The infection was visualized using a Cytation 5 plate reader.
Now, the issue Im facing is that I have kept seeing some infection in my negative controls, (which should have un-infected cells only), according to the images which showed the flourescent dye in these wells. I am trying to figure out what I am doing wrong.
I have been pipetting from lowest to highest concentration, when adding media or PBS, and used separate liquid plastic containers for the control-wells and the infected ones, to avoid contamination. I am going to redo it once again using new infection media, in case the old flask had contamination.
But apart from this, what other factors could possibly help explain why I keep seeing flourescence from the secondary antibody in my negative controls? ANY type of mistake I could possibly have been making I would love to have pointed out. Or other factors which could cause contamination in the lab.
Could PBS/the other reagents possibly be contaminated as well?
Could it be I am not washing away secondary antibodies sufficiently?
Any tips would be much appreciated!!
Relevant answer
Answer
There are a few potential sources of contamination to consider in your viral titration experiment:
  • Cross-contamination during pipetting - Make sure to always use fresh tips between different samples, and go from low to high concentration when pipetting samples and reagents. Also, avoid passing pipettes over uninfected wells.
  • Contaminated reagents - Try using fresh aliquots of reagents like PBS, antibodies, etc. Old aliquots may have become contaminated.
  • Incomplete washing - Be very thorough when washing between antibody incubations. Increase wash volume and number of washes if needed.
  • Antibody cross-reactivity - Ensure your secondary antibody is specific for your primary and is not binding non-specifically. Include secondary only controls.
  • Cell culture contamination - Check your cells under a microscope before infection to ensure no contamination. Discard and start fresh if uncertain.
  • Viral stock contamination - Make sure viral stock aliquots remain frozen and only thaw immediately before use. Avoid freeze-thaw cycles.
  • Lab environment - Decontaminate workspace thoroughly before starting. Ensure proper sterile technique. Use dedicated equipment for infected vs uninfected samples.
  • Plate reader contamination - Clean plate reader thoroughly between uses, especially if used for both infected and uninfected plates.
  • asked a question related to Reagents
Question
1 answer
I have tried PEI and lipofectamine 2000 to deliver a GFP plasmid into HEp-2 cells, both showed low transfection efficiency. Does anyone have good experiences in HEp-2 transfection?
Relevant answer
Answer
Hi Huang,
Lipofectamine 3000 reagent certainly worth trying if you're facing low transfection efficiency with HEp-2 cells. Always perform control experiments and assess cell viability and transfection efficiency to ensure that the new reagent is working effectively for your specific application. Additionally, consider running parallel experiments with your previous reagents for comparison.
Transfection can be a trial-and-error process, and the right reagent and conditions can vary depending on the cell type and the specific plasmid being used.
  • asked a question related to Reagents
Question
1 answer
I would like to simulate a process, I know T,p, and the selectivity. Can I calculate the reagent conversion in Aspen Plus?
In that case, what kind of reactor should I use?
Thank you in advance.
Relevant answer
Answer
of course you can calculate conversion of yor process. Related of your second question, you have to be in mind that software respond to a set of conditions made by THE OPERATOR, At final of the simulation software is not responsible by the results. This responsability is of the operator, that, at the end has to check if the response is resonable ( for example, if there is a strange current composition, etc). So the type of reactor you should use depends of your reaction and the set of conditions you choose. I recommend to visit software manuals and some literature to see what kind of reactor people are choosing for the type of reaction you intend to use. Also I recommend you to check in the literature what kind of themodynamic model you will choose: it may influence your results.
in other words, simulation is a very and fast tool to study process, but you have to be sure what you want to sudy and set the themodynamical model and in the case the type of reactor to be helped by software.
  • asked a question related to Reagents
Question
3 answers
I'm encountering a PCR issue in our experiments. We employ a master mix as control and utilize 3 or 4 primers in a single reaction. Since our samples are typically heterozygous (containing both wild-type and mutant alleles), it's expected to observe two bands as results. In one particular PCR, I'm consistently observing a faint wild-type band in the master mix. I've repeated the experiment using all-new reagents, but the outcome remains consistent. This light band consistently aligns with the length of the wild-type. Is it common to observe such bands depending on the choice of primers? What are the places I can improve.
Relevant answer
Answer
Run a PCR with 2 vials each of no primer, no-template, and master mix alone. And in case you are using strips, better do it in vials so that contamination can be avoided.
  • asked a question related to Reagents
Question
1 answer
How l can measure the presence of micronucleated red blood cells among normochromatic red blood cells by using the flow cytometry technique.
I need the details protocol with what the reagents should be used
Relevant answer
Answer
  • asked a question related to Reagents
Question
4 answers
I want to estimate phenol in methanol crude extract and I dissolve 1 mg/ml of crude extract as a sample then i took 500 μL of it then add 100 μL of FC (50%) reagent then add 500 μ of 20% Na2CO3 then incubate it for one h then i took 200 μL of it to 96 plat then i read absorbance at 750 nm ( Same i did for gilic acid i prepared in deffrent conc ( 5, 10, 15, 20, 25 ,30) μg/ml and i add DW to make volume 500μL then i add 100μL FC then 500μL of NaCO3 then incubation for 1 h then i took 200μ of every conc to 96 plate and i read absorbance How can we calculate the total phenols in the methanol extract ?
Relevant answer
Answer
Niti Sharma Thank you so much ma'am
I got it ma'am and i got by this method same what i got from form this excel f (=TREND(G158:G163,H158:H163,E170)) which is 20.3 ug/ml But ma'am How can i got TheTotal Phenolic Content TPC mg/g.
The conc of crude extract was 1mg/ml and the volume of it i took 0.5ml
  • asked a question related to Reagents
Question
3 answers
What concentrations of reagents are recommended to start standardizing an RT-PCR primer set?
Relevant answer
Answer
The only components you can vary are the amount of cDNA and primer concentration.
For everything else, follow the instructions in the kit since those are already optimized (e.g. temps to use for polymerase).
Amount of cDNA is going to depend on expression of your gene-of-interest so you'll have to figure it out by trial and error.
Good luck!
  • asked a question related to Reagents
Question
4 answers
Hello,
I've been having issues with my cDNA PCR which used to work previously. I'm working with cDNA from velvet worms, amplifying Cox1. Earlier, I had successfully amplified Cox1 with specific parameters that I managed to standardize. However, suddenly it stopped working when I changed the cDNA samples, and even when using the previous sample, I couldn't get amplification. I tried using new reagents at the same concentrations and only got smears. I attempted a new cDNA synthesis, considering potential degradation, but again, only smears. I experimented with different concentrations of MgSO4 (1.2 mM, 1.5 mM, 1.8 mM, 2.0 mM, and 2.2 mM) and still only got smears. I tested various cDNA concentrations and got smears as well.
I'm unsure what else to try, and I would appreciate your guidance and recommendations on this issue.
Relevant answer
Answer
Hi, these are the primers
OniCox1F TGTGACTGGTCATGCATTTGT
OniCox1R ACCACCTGCAGGGTCAAAAA
  • asked a question related to Reagents
Question
7 answers
I would like to measure the protein concentration using the Bradford assay. To do this I have to resuspend the isolated protein pellet in the sample buffer. However, at this stage, I do not have ampholyte reagent to make the rehydration buffer (I do have Urea, DTT, CHAPS and Bromophenol Blue). After this, I want to rehydrate the IEF gel strips as the first dimension gel and then run 2nd dimension gel. I am wondering if missing ampholyte in the rehydration buffer will considerably affect the result. How important is the role of ampholyte?
Any suggestions and comments would be greatly appreciated.
Relevant answer
Answer
Hi there, this is the reason this kit (see below) was invested. It is compatible with almost any buffer.
I hope it helps!
  • asked a question related to Reagents
Question
1 answer
Use of anti-FcR-antibody is popular among reserchers to study PBMCs. But is it realy neccessary to use them? Since people also use blocking reagents which seems to be working fine with most of the cases.
Please share your experiences in this regard and explain in detail.
Relevant answer
Answer
The Fc part of the antibody molecules interact with a plethora of immune cells such as B lymphocytes, monocytes, macrophages, including granulocytes. These cells express a variety of Fc receptors on their surface and can therefore generate increased background or even false positive signals in flow cytometry analysis.
This unwanted antibody binding to Fc receptors can be avoided by blocking, which is done by saturating the receptors prior to staining the cells with labeled antibodies. The potential blocking reagents that could be used include specific anti-Fc receptor antibodies or excess purified IgG, or excess (unpurified) IgG in the form of adult serum.
In theory, using antibodies against Fc receptors is the best way to eliminate unwanted signals mediated by Fc receptors binding, but you may not always have a well-optimized antibody pool against Fc receptors.
I would recommend the use of purified IgG to specifically block Fc receptors.
You could use 1 μg of IgG per 1 x 10^5 cells for 15 minutes at room temperature prior to staining. This will reduce unwanted Fc receptor interactions. When performing Fc receptor blocking with IgG while using unconjugated antibodies, it is imperative that the IgG for Fc receptor blocking not be of the same species of origin as the primary antibody to prevent the secondary antibody from recognizing any Fc receptor bound IgG.
You could also use a blocking solution prepared by adding healthy human serum to PBS to a final concentration of 10%.
Best.
  • asked a question related to Reagents
Question
4 answers
I'm using a phosphate assay kit from a company, which doesn't divulge the specifics of what's in their reagents. To prepare it, we mix a yellow-colored solution with a very small volume of clear solution. I'm trying to understand which solution has the molybdate or the malachite green, or whether they're already mixed together beforehand. Can anyone help me based on this information?
Relevant answer
Answer
Try this reference if you want to DIY the method ( ). In general you add 10x the mass of PVA than your malachite green.
  • asked a question related to Reagents
Question
3 answers
What's your experience with this conjugation method? I want to try a proximity ligation assay and I know there's kits out there that seem to be quite pricey and there are methods to simply bind Abs-oligo. I'm wondering how ppl get around the clean up step prior to adding to the target if they don't use a kit? Does anyone know what is in the Clean up reagent in the kits? Do people use columns.
Any help would be great.
Relevant answer
Answer
Thanks for your suggestions
  • asked a question related to Reagents
Question
3 answers
Hi,
I have a problem regarding Bradford protein assay. When I dissolve Coomassie Brilliant blue G-250 (C.I. 42655) in 96% ethanol, the solution is blue. Upon adding phosphoric acid, it turns into red-brown. But, when I add water to make up to the final volume, the final solution is blue. Shouldn't that be a red-brown solution?
Many thanks for your comments.
Siavoush
Relevant answer
Answer
Dear Manuele,
Many thanks for your kind suggestions and the link.
Yours,
Siavoush
  • asked a question related to Reagents
Question
2 answers
My electroactive substance is only soluble in chloroform, I have tried mixing it with Triton x 100 but it still doesn't dissolve in PBS. Is there any reagent I can use to facilitate the dissolution of chloroform in PBS?
Relevant answer
Answer
Dear Collince Omondi Awere,
Various grades of BioPBS were dissolved in one of the three solutions: chloroform, a cosolvent system of chloroform/N,N-dimethylformamide (DMF), or chloroform/dimethyl sulfoxide (DMSO).
See: Electrospinning Process and Structure Relationship of Biobased Poly(butylene succinate) for Nanoporous Fibers, 2018, DOI: 10.1021/acsomega.8b00332
Best wishes,
Ali
  • asked a question related to Reagents
Question
3 answers
I'm trying to assay different lipases and their kinetic parameters using Ellman's reagent and DMPTB as a substrate.
I've tried to measure the rate of reaction using a microwell plate in a spectrophotometer.
My protocol consists of making a premix with 0.5M Tris, 0.1M EDTA, 5% Triton, MQ water and 40mM DTNB. I'll add specific volumes of the DMPTB, mix it with the lipase and then assay it using the microwell plate. I have found that there is not colour change in the assay unless the DMPTB concentration above 5mM, so I have tried assaying different concentrations of DMPTB between 5-20mM.
However, at increasing concentrations of DMPTB (above 10mM) the rate of reaction does not increase with DMPTB (i.e. not following M-M laws)
I have also noticed precipitation in DMPTB stock solution (dissolved in Triton and 50mM Tris). But I vortex the solution prior to pipetting to offset this effect. Could this be contributing to the problem I'm seeing?
Any help would be great !
Relevant answer
Answer
Dominique Liger Nick Gee Thank you both! I replaced the premix with one at pH 8 and removed the EDTA, and now can see colour change within in the 250uM - 5mM range.
  • asked a question related to Reagents
Question
2 answers
I would like to isolate RNA from plants, is an low cost RNA isolation protocol available? Preferable using Chloroform as main reagent.
Thank you in advance,
Kiriakos Athanasiadis
Relevant answer
Answer
Hi Kiriakos,
I have done RNA isolation from different types of tissue i(n the time when no RNA isolation kits were available) with the method described by Chomczynski and Sacchi, 1987 (attached). I am not sure if the purchase of all components required is cheeper than a comercial kit but this might be dependent on the number of samples you need to process.
Best,
Murat
  • asked a question related to Reagents
Question
1 answer
I need to make a standart curve for quantitative the ammonia produced by bacteria Bacillus. I want to make standart curve ammonium chloride with the various standart 0-20mg/L. And then I was take 25uL of each standart added to 850uL H2O and 125uL of Nessler reagent was added. Please help me to solve how to make various standart.
Thanks
Relevant answer
  • asked a question related to Reagents
Question
3 answers
I am using 100 and 120 nm latex particles and monoclonal and poly-clonal anti-crp antibody. i tried with Tris, Glycien, PBS, and MES buffer with different molarity (10 to 100 mmol) but not getting linearity more then 100 mg/L.
Relevant answer
Answer
Wolfgang Schechinger Finally, i got the 150 linearity with single point calibration. Thanks for your reply.
  • asked a question related to Reagents
Question
5 answers
I would like to add BG-azide to cells for SNAP tag pulldown however I don't know whether it is going to be cell permeable. My instinct is to day yes it will be but neither me nor the supplier know whether that is true. I thought I would ask if anyone has tried something similar, even though that is unlikely... I have attached the structures in case that is informative
Relevant answer
Answer
Thanks Wolfgang, I appreciate the detailed answer!
  • asked a question related to Reagents
Question
1 answer
Looking to perform TEM on tissues that have already been processed via RNAScope. Are there concerns with viral and/or cellular degradation from RNAScope reagents that may prevent TEM from detecting the molecularly-confirmed virus?
Relevant answer
Answer
I have gotten somewhat useful results out of this once, in that I could at least identify viral capsids in infected cells, but it obviously depends on what information you exactly need for your analysis.
The treatment is harsh and thus the ultrastructural preservation is ... bad. Expect the lipids to be pretty much gone, and the protease treatment doesn't help either ;)
  • asked a question related to Reagents
Question
2 answers
All parameter is ok but hemoglobin is 4% higher than control. the formula is SDS + Triton x100+ phosphate buffer.
Relevant answer
Answer
Jukka Salminen Hello dear Salminen calibration is not standard solution for this issue because the difference is high (4%).
  • asked a question related to Reagents
Question
2 answers
Hi, I'm trying to block FcR on human macrophages in some in vitro cocultures with human T cells. Is there any FcR blocking reagents used in vitro culture?
Thanks,
Kun
Relevant answer
Answer
Yes, there are several FcR blocking reagents that can be used in vitro to block Fc receptors on human macrophages. Fc receptors are involved in the binding and uptake of immune complexes and play a role in various immune responses.
One commonly used FcR blocking reagent is human immunoglobulin G (IgG). IgG can saturate the Fc receptors on macrophages, preventing them from binding to other antibodies or immune complexes. Human IgG can be obtained commercially and used at a high concentration in the culture medium.
Another option is the use of monoclonal antibodies specifically targeting Fc receptors. For example, anti-CD16 (FcγRIII) antibodies can block the FcγRIII receptor on macrophages. Similarly, anti-CD32 (FcγRII) antibodies can block the FcγRII receptor. These antibodies can be obtained from commercial sources and used at the appropriate concentrations for blocking.
It's important to note that the choice of FcR blocking reagent may depend on the specific Fc receptors involved and the experimental setup.
All the best with your experiment
  • asked a question related to Reagents
Question
2 answers
I am a novice in chemical experiments,and my teacher wants me to use steglich esterification reaction(add DCC and DMAP) to make PEG and RAFT reagents(DDMAT) react into a macromolecular reagent(PEG DMATmacroCTA), my method is as follows:
Under stirring conditions of 0 ℃, the dodecyl S'-( α,α'- Dimethyl- α″- Acetic acid) tri Thiocarbonic acid (DDMAT) (0.5485mmol, 200mg), PEG-400 (0.2493mmol, 99.72mg) and 4-Dimethylaminopyridine (DMAP) (0.0997mmol, 12.18mg) were mixed in 15mL chloroform (super dehydrated grade), and dicyclohexyl Diimide (DCC) (0.5485mmol, 113.17mg) dissolved in 5mL chloroform was dropped into it, further stirred at 0 ℃ for 1h, and then stirred at room temperature for 72h, Concentrate the filtrate and separate it by Column chromatography.And this method is derived from my first linked literature, which was used by the author to synthesize T2 using ethylene glycol and RAFT reagent.
However, I repeated the experiment twice but failed. The points seen through thin layer chromatography cannot be separated through column chromatography, I am also unable to determine whether the newly generated point is my product point, and the substance seems to decompose easily. It always disappear during the post-processing process.
I want to use the classic acyl chloride method for synthesis(The references are in the second and third links), but my teacher doesn't seem to want me to switch methods.
So I really want to know which step went wrong and why I couldn't get the product. If you need more specific experimental details, please contact me. I write a lot to describe my difficulties, and thank you for spending your time to read it. If you can provide suggestions, I would be very grateful.
Relevant answer
Answer
Wolfgang Schechinger Thank you for your suggestion. I will consider the method you mentioned and carefully learn from it. Thank you again for taking your time to reply to me. And the research you have done is really great! I really hope to become someone as powerful as you in the future.(๑•̀ㅂ•́)و✧
  • asked a question related to Reagents
Question
1 answer
I would like to know your opinion on kits for separation of nuclear and cytoplasmic fraction. I have used NE-PER™ Nuclear and Cytoplasmic Extraction Reagents, but I was not satisfy with the results even though I did some optimizations. Now I'm planning to order Cell Fractionation Kit #9038 and I need some helpful information if I made a good chose. I'm open for any suggestions. Thank you in advance!
Relevant answer
Answer
Hi Elena,
Could you please share your feedback on why you were not satisfied with the "NE-PER Nuclear and Cytoplasmic Extraction Reagents" kit? I'm particularly interested in knowing about the quality of fractionation you obtained. I am planning to conduct studies on transcription factors and it is crucial for me to obtain a clear fraction from the nucleus. I appreciate your time and any insights you can provide. Thank you!
  • asked a question related to Reagents
Question
9 answers
Hi there!
I got amplification of my genes in the negative control in Real-time PCR experiment although the Ct values were above 30. I setup my reaction again after making new dilutions of PCR reagents, cleaning the workbenches, using unopened autoclaved tips, and cleaning my pipettes with 70% ethanol. I tried to remove every possible source of contamination but still I am getting the same Ct values in my negative control. It is a TaqMan probe-based multiplex PCR reaction where I am amplifying three genes in the same reaction and I am getting amplification of two of these genes in my negative control. Can anyone guide me how to get rid of this amplification?
Relevant answer
Answer
I recommend verifying these steps:
· Cross-contamination: Take care to prevent cross-contamination between samples by using separate equipment and workspace for the negative control. Double-check that pipettes and consumables are not shared.
· Contaminated reagents: To minimize the risk of contamination, handle reagents with caution. Use fresh reagent stocks from a different batch, store them properly, and ensure they are not compromised.
· Aerosol contamination: Employ filtered pipette tips, work in a clean environment, and keep PCR tubes or plates covered while performing actions like opening tubes, vertexing, or pipetting to minimize the potential for airborne DNA fragment contamination.
· Carryover contamination: Thoroughly clean and decontaminate laboratory equipment, including pipettes, tubes, and the thermal cycler, to eliminate any residual DNA from previous experiments that could contribute to carryover contamination.
My suggestions:
1. Repeat the experiment using new reagents, preferably from a different batch, to eliminate the possibility of contaminated reagents.
2. Set up a fresh negative control in a separate area using different pipettes and consumables to reduce the risk of cross-contamination.
3. Conduct additional negative controls using sterile water instead of template DNA to determine if the issue persists. This will help identify if the contamination originates from the reagents or the laboratory environment.
4. Ensure that PCR tubes or plates are properly sealed to prevent aerosol contamination during the experiment.
  • asked a question related to Reagents
Question
1 answer
in the experiment to quantify the ability to produce siderophore in rice roots by CAS reagent. We have performed 3 times at different levels, but the OD measurement result is still negative. Tell me the reason
First time: 600ul bacterial epidemicin, 600ul CAS reagent. Measured at 630nm
Second time: 600ul bacterial epidemicin, 600ul CAS reagent. Measured at 530nm
3rd time: 900ul of bacterial epidemicin, 100ul of CAS reagent. Measured at 630nmm
Relevant answer
Answer
If OD measurement results for quantifying siderophore production in rice roots are consistently negative, there are several potential reasons. These include insufficient siderophore production, an ineffective CAS reagent, incorrect measurement parameters, experimental conditions, and technical errors. Insufficient siderophore production can occur due to the bacteria's inability to produce siderophores, while ineffective CAS reagents may not be suitable for detecting specific siderophores. The optimal wavelength for measuring OD should be appropriate for the CAS reagent and validated for siderophore quantification. Experimental conditions, such as pH, temperature, incubation time, and bacterial growth stage, can also impact siderophore production.
Technical errors, such as inaccurate pipetting or contamination, can lead to inconsistent or inaccurate results. To troubleshoot the issue, verify the bacterial strain, check the compatibility and specificity of the CAS reagent with the siderophores produced by the strain, validate measurement parameters, evaluate and optimize experimental conditions, review the experimental procedure, and consult relevant literature or experts for guidance. By systematically assessing these factors, potential causes for negative results can be identified and adjustments can be made to improve the experiment.
Some references:
Layer plate CAS assay for the quantitation of siderophore production and determination of exudation patterns for fungi Megan Y Andrews et al. J Microbiol Methods. 2016 Feb.
https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3577196/ TITLE: Use of Blue Agar CAS Assay for Siderophore Detection - PMC CONTENT: Journal List J Microbiol Biol Educ v.12(1); 2011 PMC3577196 J Microbiol Biol Educ. 2011; 12(1): 51–53.
  • asked a question related to Reagents
Question
2 answers
Looking for Protocol of GRIESS reagent test on brain.
many thanks
Relevant answer
Answer
Yes, the Griess reagent test can be used to detect nitric oxide (NO) in brain samples. The Griess reagent test is a colorimetric assay that detects the presence of nitrite (NO2-), which is a stable metabolite of NO. The test is based on the reaction between nitrite and the Griess reagent, resulting in the formation of a deep purple azo compound.
Here is a general protocol for conducting the Griess reagent test on brain samples:
1. Tissue preparation: Collect brain tissue samples and homogenize them in a suitable buffer or solution. Ensure that the samples are properly stored and handled to prevent degradation of NO.
2. Sample preparation: Take an aliquot of the brain homogenate or extract and transfer it to a clean tube. If necessary, perform any required sample dilutions to ensure that the nitrite concentration falls within the linear range of the assay.
3. Griess reagent preparation: Prepare the Griess reagent by mixing equal volumes of 1% sulfanilamide solution (in 5% phosphoric acid) and 0.1% N-(1-naphthyl)ethylenediamine dihydrochloride solution. Mix the reagents well and allow the Griess reagent to develop a deep purple color.
4. Assay procedure:
a. Take a volume of the sample or diluted sample and transfer it to a clean tube.
b. Add an equal volume of the Griess reagent to the sample and mix thoroughly.
c. Incubate the mixture at room temperature for a specified time (e.g., 10-30 minutes) to allow the color reaction to occur.
d. Measure the absorbance of the resulting colored solution at a suitable wavelength (e.g., 540 nm) using a spectrophotometer.
e. Compare the absorbance values of the sample to a standard curve generated using known concentrations of nitrite to determine the nitrite concentration in the sample.
It's important to note that the Griess reagent test specifically detects nitrite (NO2-) rather than NO itself. Nitrite is a stable metabolite of NO and can serve as an indicator of NO production. Therefore, the Griess reagent test indirectly provides information about NO levels in the brain samples.
Make sure to follow safety guidelines and proper laboratory practices while conducting the assay. Additionally, consult relevant literature or protocols specific to your research needs for more detailed instructions and considerations.
  • asked a question related to Reagents
Question
3 answers
Kindly tell the electrolytic polishing reagent as well as chemical and electrolytic etchant for 304H austenitic steel.
Relevant answer
Answer
304H Stainless Steel, I am generally use 10% aqueous solution of oxalic acid at a voltage of about 6 volts and a duration of 40-60 seconds. if not etching properly increase voltage up to 12 v and time to 120 seconds
  • asked a question related to Reagents
Question
3 answers
Hello everyone. I want to perform TPC assay for phenolic content estimation of my honey samples.
However, the only FC reagent available in my lab is from Sigma Aldrich that got 'suitable for determination of total protein content by lowry method' on the bottle label.
Is it still suitable to be used for my TPC assay or should I purchase another one?
Thanks a lot for your advice!
Relevant answer
Answer
Folin reagent ( FC) is famous use in TPC determination content. In every standard methods
  • asked a question related to Reagents
Question
1 answer
Do you need to wash with ethanol or another non-polar reagent in the washing step to obtain a HTC hydrocarbon with higher purity? in addition to washing with distilled water.
Relevant answer
Answer
Following acid can be used for removing such types of impurities
1. HCl,
2. Chromic acid (H2CrO4)
3. H2SO4
Hold for 15-25 min. and stirring and then rinse with DM water.
  • asked a question related to Reagents
Question
1 answer
hello, I have a major question that i would really appreciate if anyone could make me clear.
In a project I am going to transfect BMSCs with a plasmid by lipofectamine 2000 reagent.
then the transfected cells will be transplanted to the rat brain.
SINCE the rats are going to be tested for 10 days after cells transplantation, THE QUESTION IS THAT can plasmids remain in cells during this time and efficiently express the desired gene or not?
Relevant answer
Answer
The persistence of transient plasmids in cells after transfection can vary depending on multiple factors, including the cell type and the characteristics of the plasmid. In the case of bone marrow stem cells, the duration of transient plasmid expression can range from a few days to a few weeks.
Bone marrow stem cells are known to have a relatively short lifespan in culture, and their division rate can vary depending on the specific subtype of stem cells and culture conditions. Therefore, the duration of plasmid expression may be limited by the cell's proliferation and turnover rates.
Additionally, the choice of promoter in the plasmid can influence the duration of expression. Plasmids commonly used for transient transfection, such as pEGFP-N1, typically utilize strong constitutive promoters, like the CMV promoter, which can lead to robust protein expression shortly after transfection. However, the activity of the promoter may decline over time due to various factors, such as epigenetic modifications or dilution of the plasmid during cell division.
It's worth noting that transient plasmids are not maintained in the genome of the host cells. Therefore, if you need long-term or stable expression of your gene of interest in bone marrow stem cells, you may consider exploring alternative approaches such as viral vectors or gene editing techniques.
To assess the duration of transient plasmid expression in your specific bone marrow stem cell culture, you can monitor the expression of the transfected gene over time using techniques like fluorescence microscopy or flow cytometry. It is recommended to perform time-course experiments to observe the decay of expression and optimize the timing for downstream analyses or experiments based on your specific research goals.
  • asked a question related to Reagents
Question
3 answers
When i started my real time pcr experiment i was using applied biosystems power up sybr green qpcr master mix. i was getting my results with a proper melt curve of my target gene at 83.5 degree Celsius and housekeeping gene at 85 degree Celsius. but this reagent was limiting, so i had to shift to applied biosystems power sybr green, and after that my target gene did no show a melt curve. even for housekeeping gene the melt curve decreased to 83 degrees. in this entire experiment the only change was for sybr green reagent. i ran a control, where i freshly prepared rna, and set up real time experiment simultaneously with 2 sets of sybr green, i got the melt curve as well as band in agarose gel for power up sybr green, but not with the power sybr green.
Relevant answer
Answer
I set up a reaction volume of 10ul. But I am not getting a melt curve. The band for gapdh is of low intensity when compared to power up sybr green gapdh band
  • asked a question related to Reagents
Question
2 answers
I'm making 4 formyl carbaldehyde by cycling the hydrazone derivatives using vilsmeir hack reagent . But after 15-20 days my product is showing two spots in tlc. One non polar spot is generating extra.
I'm using 20% Ethyl acetate: Pet. Ether as eluent.
Can anyone explain why or how can I avoid it.
Relevant answer
Answer
"4 formyl carbaldehyde" is insufficient to indicate a structure. You might want to provide a drawing, or better yet a reaction scheme.
  • asked a question related to Reagents
Question
2 answers
As we are finding difficult to secure the Trypan Blue supplies in Colombia, i would like to know if there is any alternative reagent to Stain the PBMC cells.
Relevant answer
Answer
  • asked a question related to Reagents
Question
3 answers
Hello,
I need to ask about why DS value is decreasing when increasing molar ratio of reagent?
First I would like to tell that I performed 5 synthesis of cellulose with vinyl laurate. The result of DS is decreasing when I increased molar ratio of vinyl laurate. Kindly tell me why I am getting this decreasing trend and also my DS values are less than 1, least is 0.16 and highest is 0.40.
Kindly help me out.
Regards
Relevant answer
Answer
how to control degree of substitution (DS)?
  • asked a question related to Reagents
Question
2 answers
My graduate work was in C. elegans, and this is my very first time performing live cell imaging in human cell lines. (fibroblasts)
I would like to use live cell imaging to track two proteins of interest, but I am not sure where to start.
Does anyone have recommended reagents, plasmids, or protocols?
Relevant answer
Answer
To track two proteins of interest using live cell imaging, there are several plasmid and protocol options you can consider. Here's a suggestion:
  1. Plasmids:Fluorescent protein tags: Use plasmids that express your proteins of interest fused with different fluorescent protein tags. For example, you can use green fluorescent protein (GFP) for one protein and red fluorescent protein (RFP) for the other. This allows you to visualize the proteins separately using different fluorescence channels. Tandem dimeric fluorescent proteins (tdFPs): Another option is to use plasmids that express tdFPs, such as mCherry2, TagRFP-T, or mEos2. These proteins self-assemble into higher-order complexes, resulting in enhanced brightness and reduced photobleaching compared to single fluorescent protein tags.
  2. Imaging Techniques:Confocal microscopy: Confocal microscopy provides high-resolution, three-dimensional imaging. It allows you to capture detailed images of protein localization and dynamics within living cells. Time-lapse microscopy: Employ time-lapse imaging to monitor the dynamics of the two proteins over time. This technique can capture changes in protein localization, movement, and interactions within live cells. Super-resolution microscopy: If you require higher spatial resolution, consider using super-resolution techniques like structured illumination microscopy (SIM) or stochastic optical reconstruction microscopy (STORM). These methods can provide enhanced imaging of subcellular structures and protein distributions.
  3. Protocol considerations:Cell type and transfection: Choose a transfection method suitable for your specific cell type, such as lipid-based transfection reagents, electroporation, or viral transduction. Ensure efficient transfection to achieve robust expression of the tagged proteins. Fluorescent protein maturation: Take into account the maturation time required for the fluorescent proteins you are using. Some proteins need time to mature and reach their optimal fluorescence. Imaging conditions: Optimize imaging parameters such as exposure time, laser power, and acquisition frequency to minimize phototoxicity and photobleaching while maximizing the signal-to-noise ratio. Live cell imaging chamber: Use a specialized live cell imaging chamber to maintain the cells under physiological conditions, including temperature, humidity, and CO2 levels. This helps ensure cell viability during extended imaging sessions.
Remember that the choice of plasmids and protocols may depend on your specific research goals, the characteristics of the proteins of interest, and the imaging equipment available in your laboratory. It's advisable to consult relevant literature, established protocols, or seek guidance from experienced researchers to tailor the plasmid and protocol choices to your specific experimental needs.
  • asked a question related to Reagents
Question
1 answer
Hi, I normally use Lipofectamine 2000 reagents for siRNA transfection, we adquiere a new lot, and in the second use, after taking it from the refrigerator, I put it on ice, and at the time of use I noticed that the tube appeared cloudy, I thought it was frozen, after a minute and shaving it looked normal but cloudy, I have made my transfections with out problem, but the transfection is in OptiMEM with out serum or antibiotics for 18 hours, after that, I harvest the cells for TRIzol RNA extractions, the cells and transfection process look perfectly normal after those hours in OptiMEM. We suspected the reagent to be contaminated, but we haven't made sequential experiments involving reseeding the cells in other media.
Relevant answer
Answer
Dear Alex
Lipofectamine is prone to form cationic liposomes at the concentrations above its critical micelle concentration (CMC). The resultant colloidal solution will be cloudy. Nevertheless you can observe few microliters of it by the microscope to prove/disprove any bacterial contamination.
  • asked a question related to Reagents
Question
1 answer
I want to know the composition of anti fluorescence quenching reagent,how can i make it .
Relevant answer
Answer
According to the Safety Data Sheet for the anti-fade product from Sigma-Aldrich (catalog number A7114), the solution contains 0.1% p-phenylenediamine dihydrochloride in glycerine.
  • asked a question related to Reagents
Question
1 answer
I am trying to use BriteVu casting reagent to perfuse the coronary vasculature and image using microCT. I have tried 1) perfusion via catheterization of the descending aorta with ligation of aortic branches and 2) injection directly into the LV without aortic ligation. Both methods of yielded incomplete and inconsistent filling of the vasculature. I am seeking protocols, tips, etc. to improve our perfusion protocol.
Relevant answer
Answer
You can take help of leading research institute across world they will help u to making protocol .
  • asked a question related to Reagents
Question
2 answers
Characterizing a GPCR-ligand interaction is critical to understanding the biology of the receptor. As GDP/GTP exchange is one of the earliest events that follows ligand binding, monitoring GTP binding can measure GPCR activation or inhibition. Assaying more downstream events in GPCR signaling is often not as quantitative or stoichiometric, may not distinguish full agonists from partial ones, and can require expensive reagents. Moreover, increased GTP binding to Gα proteins is an almost-universal event following GPCR activation, meaning that measuring GTP binding is a broadly applicable assay for monitoring the activity of most GPCRs. Measuring GTP binding is a simple and rapid approach to monitor GPCR signaling in cells overexpressing the receptor of interest or in native tissue. The present protocol details a functional GTP-binding assay using an archetypal GPCR, the µ-opioid receptor (MOR1), to quantitatively determine the activity of an agonist and antagonist on GPCR signaling.
But we are facing some logistical issues acquiring the ([35S]GTPγS), and due to shortage of time we need to measure the GPCR signaling without the radio-labeled ([35S]GTPγS).
Could you please help me out and suggest some alternate approach?
Relevant answer
Answer
There are several alternative methods available to measure G protein-coupled receptor (GPCR) signaling without relying on radio-labeled GTP binding. These methods provide valuable insights into GPCR activation and downstream signaling events. Here are some commonly used techniques:
1. Fluorescent GTP Analogs: Fluorescently labeled GTP analogs, such as mant-GTP or Bodipy-GTP, can be used to monitor GTP binding to G proteins. These analogs emit fluorescence upon binding to G proteins, allowing for real-time detection of GPCR activation. Fluorescence resonance energy transfer (FRET) techniques can also be employed to measure GTP binding indirectly.
2. Bioluminescence Resonance Energy Transfer (BRET): BRET is a proximity-based assay that measures protein-protein interactions. In the context of GPCR signaling, BRET can be used to detect interactions between GPCRs and downstream signaling molecules, such as G proteins or β-arrestins. This technique provides insights into GPCR activation and downstream signaling events.
3. Calcium Imaging: GPCR activation often leads to intracellular calcium mobilization. Calcium imaging techniques, such as fluorescence microscopy using calcium-sensitive dyes (e.g., Fluo-4), can be employed to measure changes in intracellular calcium levels upon GPCR activation. These measurements provide information about GPCR signaling and downstream calcium-dependent processes.
4. Phosphorylation Assays: GPCR activation often triggers phosphorylation events mediated by protein kinases. Phosphorylation assays, such as western blotting or immunofluorescence using phospho-specific antibodies, can be utilized to detect and quantify GPCR-induced phosphorylation of downstream signaling proteins. This approach provides insights into GPCR signaling pathway activation.
5. cAMP Assays: GPCRs can activate intracellular signaling pathways, such as the cyclic adenosine monophosphate (cAMP) pathway. cAMP assays, such as enzyme-linked immunosorbent assays (ELISAs) or fluorescence-based assays using cAMP-sensitive dyes, can be employed to measure changes in cAMP levels upon GPCR activation. These measurements provide information about GPCR signaling and downstream cAMP-dependent processes.
  • asked a question related to Reagents
Question
11 answers
Reagents
Relevant answer
Answer
AAS is recommedned for micro to mg/L concentrations in water and wastewater samples.
  • asked a question related to Reagents
Question
2 answers
Hi,
I'm looking to learn how to use RNAscope. I could have access to some of the reagents but they are expired. Since the reagents and probes are quite expensive, I was wondering if I could use them despite the fact that they are expired.
Thank you for your comments.
Mathieu
Relevant answer
Answer
Thank you Camille for your input
  • asked a question related to Reagents
Question
2 answers
I am using the Cell Biolabs Inc. AAV Purification Standard Kit to purify Adeno-Associated Virus particles, but unfortunately, I have lost the Reagent B. I have checked the product manual and website but cannot find any information about the specific function of this reagent. Does anyone who has used this kit before know what Reagent B is used for and what its composition is? Any help or suggestions would be greatly appreciated.
Relevant answer
Answer
Thank you for your answer. I have attached the protocol of my kit to show where Reagent A and B is used. Does it look similar to yoour PEG/NaCl protocol. Also can you suggest any protocol to purify AVV2?