Science topics: MicrobiologyAssays
Science topic

Assays - Science topic

Explore the latest questions and answers in Assays, and find Assays experts.
Questions related to Assays
  • asked a question related to Assays
Question
1 answer
Hi friends,
I am working with HSPC-colony formation and downstream WGS. I am looking for the more efficient assay to isolate DNA from smallest amount of cells (one colony is not more than 1000 cells). If you are experienced in this field, could you help me with choosing right approach?
Relevant answer
Answer
You can use QuickExtract DNA Extraction Solution (LUCIGEN) and follow manufacturer's instructions.
  • asked a question related to Assays
Question
2 answers
Hello everyone. I need some information about the use of CaCl2 in lipolytic assays. I often see the addition of CaCl2 in lipolytic activity assays, especially in probiotic screening, but there is no explanation about it, nor the reason for using the concentration. I hope you can help me.
Thank you.
Relevant answer
Answer
Thank you very much.
  • asked a question related to Assays
Question
1 answer
Can anyone suggest any kit for SOD activity in plant? I have the protocol for NBT reduction but don't have centrifuge for falcon as the protocol requires 3mL of homogenate to be centrifuged in falcon. Can someone provide any alternative?
Relevant answer
Answer
Instead of using falcon tubes, you can use microcentrifuge tubes that fit into a standard microcentrifuge. These tubes typically have smaller capacities (e.g., 1.5 mL or 2 mL), but they are suitable for processing smaller volumes of sample.
Here are a few examples of commercially available SOD assay kits:
Sigma-Aldrich SOD Assay Kit:
Cayman Chemical Superoxide Dismutase (SOD) Assay Kit:
BioVision SOD Activity Assay Kit:
Abcam Superoxide Dismutase (SOD) Activity Assay Kit:
These kits typically come with detailed protocols and all the necessary reagents for performing the assay.
  • asked a question related to Assays
Question
3 answers
I have cultured PBMC and some RBC contaminant for 2 weeks in suspension and low attachment plate. From the very first day, I noticed that there are many cells that have already turned all black-ish or grey-ish color. I don't know why and I keep continuing my culture process because the sample is very limited and I don't want to waste any samples. This is very confusing as because I will do viability assay using trypan blue, but the cells were already black. So I can't see the blue-ish color that supposed to appear. For additional information, my medium is DMEM F12, EGF, FGF, CoCl2, ITS, Penstrep, and Amphotericin.
Relevant answer
Answer
PBMC don't turn black. You have a very strange problem. Either your microscope is damaged, or a very weird molecule got into your culture somehow. How much cobalt is that?
PBMC do largely die over time, so it's difficult to estimate what portion might be alive based on other people's experiments. Can you access a Hoffman's contrast microscope? With that, you could discriminate live from dead PBMCs without any staining, by just looking at them. The live ones look nice and spherical, and the dead ones flat and kind of crinkled.
You could also use a dye like MTS to tell if there are any live cells at all in your strange culture.
  • asked a question related to Assays
Question
5 answers
Hi all!
I have to quantifiy some proteins in RBCs by ELISA. Since RBC number cannot be standardized and it obviously affect the results, there is the need of some normalization. I read that many groups normalize for total protein content measured by CBA assay. However, I was wondering whether I can normalize my values by using haemoglobin, which accounts for 98% of total proteins in RBCs. For me, it would be far less time and sample consuming, since I would use a spectophotometer and very few microliters of sample, but I would like to know if someone ever used this normalization and if can be considered as correct and reliable.
Many thanks in advice!
Relevant answer
Answer
thanks for your kind and thorough reply.
In RBC, I am actually looking for proteins to be used as biomarkers for diseasese not related to RBCs. Therefore, RBCs for me are just a biological matrix, and the proteins I am going to investigate should be not related to RBC function. I can consider my sample as sufficiently pure, since I careflly wash it many times to remove serum and carefully remove the white ring of leukocytes. The reason why I would like to normalize for haemoglobin content instead of total protein is because, as you mentioned, it is simpler, faster, more standardized and requires far less amount of sample.
  • asked a question related to Assays
Question
1 answer
Hi, I saw some paper used NaNO3 and some used NaNO2 in TFC assays. I seek opinion from experts for me to understand the difference between the usage of these 2 chemicals, is it possible to use either of them in TFC assays? Thank you in advance.
Relevant answer
Answer
You can refer to this paper. There is quite an extensive discussion regarding TFC assay.
  • asked a question related to Assays
Question
3 answers
Looking for biochemical assays for quantitative estimation of Albumin and Casein protein.
Relevant answer
Answer
If you have pure proteins in solution, you can use any of several protein assay kits that are commercially available (examples, BCA assay and Bradford assay). Each one requires a standard. The standard, in these cases, can be the same protein that you are measuring, since they can be purchased.
If the pure proteins are in a solution in which there is nothing else that absorbs in the far UV, you can use the method of Gill and von Hippel to calculate the molar extinction coefficient (or look it up somewhere), and measure the UV absorbance to get the concentration. This method requires a solution of 8 M guanidine-HCl to fully denature the protein.
Gill, S.C. and von Hippel, P.H. (1989). Calculation of protein extinction coefficients from amino acid sequence data. Anal. Biochem. 182:319-26
The refractive index of the solution of pure proteins can be used to measure the concentration of the protein, using the refractive index increment (dn/dc) for globular proteins (0.185 mL·g−1), if a refractometer is available.
For pure proteins, you can send them to a specialist lab to measure the concentrations by amino acid analysis.
If the proteins are present in mixtures, then you can use an ELISA kit with specificity to the protein of interest. These can be purchased, but are expensive. Standards of the same proteins of known concentration are required, and should be supplied with the kit.
If the protein of interest is a major component in a mixture, a semi-quantitative method would be to use SDS-PAGE, Coomassie Blue staining, and densitometry to measure the percentage of stain in the band corresponding to the protein of interest, and compare that to the total protein concentration measured by some other method, such as Bradford or BCA. This requires a densitometer or gel imager with this capability.
There are a variety of other possibilities, depending on the nature of the sample, the availability of a standard of comparison, and the available equipment.
  • asked a question related to Assays
Question
4 answers
Hi, I would like to perform the DHE assay to test antioxidant compounds on HT29. I am using antimycin A (100 microM) as a positive control (mitochondrial chain inhibitor). I have a good response with the positive control, but I am struggling to find a known antioxidant that works.
To specify, I'm using DHE as fluorescent probe and not MitoSox. Also, I use a confocal microscope.
Do you have any suggestions?
Thank you.
Relevant answer
Answer
I have used Quercetin and Ascorbic acid which effectively quench the relevant ROS release in DHE.
Hope it helps,
Thanks,
  • asked a question related to Assays
Question
2 answers
3D cell culture experiments and assays are often still carried out in the standard incubator at 5% CO2, which means around 19% oxygen. However, physiological values are different. In your opinion, how important is it to consider the physiological oxygen concentration when performing 3D cell culture assays?
We are currently working on a research project that focuses on these issues, including high-throughput and automation. I would appreciate a discussion and also participation in a short survey on this topic:
Relevant answer
Answer
It is important to also consider that the availability of oxygen to the outer layer of cells is different to what the inner cells in the 3D culture are receiving.
  • asked a question related to Assays
Question
1 answer
Method of exogenous control standardization in miRNA Taqman assay for qPCR
Relevant answer
Answer
In miRNA TaqMan assays, exogenous control standardization is crucial for accurate and reliable quantification of microRNA (miRNA) expression levels. Exogenous controls, also known as spike-in controls, are synthetic RNA molecules that are added to each sample at a known concentration before the RNA isolation step. These controls serve as reference points to normalize variations in sample preparation, RNA isolation efficiency, and reverse transcription efficiency across different samples.
  • asked a question related to Assays
Question
2 answers
I'm currently doing total cholesterol assay in SH-SY5Y cells treated with 0, 25, 50, 100ug cholesterol/mL (using cholesterol:mbcd, sigma)
Lipid extraction is done by chloroform and methanol. After taking chloroform sublayer it is dried in N2 gas, dissolved again in isopropanol and then absorbance is measured in 500nm.
The problem is that mg TC/mg protein is too high (almost 100 times bigger) compared to preceding studies.
The mg TC/mg protein of control(untreated) SH-SY5Y measured was about 0.1656.
Its value is similar(or even higher) to animal tissue rather than cell.
Can anyone help me with this problem, please?
Relevant answer
Answer
Thank you for the answer! I'll review my protocol again.
  • asked a question related to Assays
Question
2 answers
I am trying to perform Tunel assay for senescence. DNA fragmentation is one of the characteristics in senescence. I had extracted DNA from scenescent cells and performed Tunel assay. But the RFU obtained was very less. My question is, whether the low speed during centrifugation causes loss of DNA?
Relevant answer
Answer
Thank you
  • asked a question related to Assays
Question
3 answers
Hello! I´m developing a sandwich ELISA. My assay recognizes protein denaturalized very well, but there isn´t recognition when it is in its native form. So maybe if I heat the sample I could detect my protein.
Relevant answer
Answer
  • asked a question related to Assays
Question
2 answers
I have cells that produce GFP as a control, and I intend to perform the MTT and Caspase 3/7 Glo assays. The emission wavelength of GFP is 508 nm, and its absorption is 475 nm. In addition, I will provide a medication to the cells that emits light at 592 nm and is stimulated at 480 nm. I've never performed these assays before, so I'm not sure if that will have an impact on the outcomes. And how could I get beyond this obstacle if they have? Many thanks.
Relevant answer
Answer
Thank you for the explanation@ Kais Khudhair al Hadrawi.
  • asked a question related to Assays
Question
1 answer
I am working on the Primary neuron cell. I am trying to observe the stress granule in the cell. For that, I have done transfection with my gene of interest and treated the cell with Arsenite. I want to do Fluorescence recovery after photobleaching assay to know the mobility of the proteins. Can anyone tell you how you have performed the FRAP assay?
Relevant answer
Answer
Fluorescence Recovery After Photobleaching (FRAP) is a technique used in cell biology and microscopy to study the dynamics of molecules within living cells. Here's how it works:
  1. Principle: FRAP involves selectively bleaching a small region of fluorescently labeled molecules within a cell using a high-intensity laser. This bleaching process irreversibly destroys the fluorescence of the molecules in the targeted area.
  2. Observation: After bleaching, the recovery of fluorescence intensity in the bleached region is monitored over time. The recovery occurs as fluorescent molecules from the surrounding unbleached regions diffuse into the bleached area.
  3. Analysis: The rate and extent of fluorescence recovery provide information about the mobility, turnover, and interactions of the labeled molecules within the cell. Different models can be used to analyze FRAP data, such as diffusion, binding, or compartmentalization models, depending on the biological context.
  4. Applications: FRAP is used to study various cellular processes, including protein diffusion, membrane dynamics, cytoskeletal dynamics, and protein-protein interactions. It can also be used to assess the effects of drugs or genetic manipulations on molecular mobility within cells.
FRAP is a powerful tool for understanding the spatiotemporal dynamics of molecules in living cells, providing insights into their behavior and function in complex biological systems.
  • asked a question related to Assays
Question
2 answers
I am working on an cyanobacteriochrome protein, similar to other phytochromes. I have cloned, overexpressed and purified the protein. In order to confirm the chromophore binding ability of protein I have to perform the zink blot assay. There are various protocols available for this assay but I have to perform it with the purified protein. So, Please suggest the correct protocol.
Relevant answer
Answer
The zinc blot assay is a technique used to detect proteins that bind zinc ions. If you have purified protein and want to perform a zinc blot assay, here's a general protocol you can follow:
  1. Preparation of Samples: Prepare your purified protein samples in appropriate buffers. Ensure that your protein samples are denatured if required by your assay protocol.
  2. SDS-PAGE Electrophoresis: Load your protein samples onto an SDS-PAGE gel and perform electrophoresis under denaturing conditions to separate the proteins based on their molecular weights.
  3. Transfer to Membrane: After electrophoresis, transfer the proteins from the gel onto a nitrocellulose or PVDF membrane using a semi-dry or wet transfer apparatus.
  4. Blocking: Block the membrane with a blocking buffer (e.g., 5% non-fat dry milk or BSA in TBST) to prevent the non-specific binding of antibodies.
  5. Incubation with Zinc Probe: Prepare a solution of a zinc probe, such as FluoZin™-3, which binds specifically to zinc ions. Incubate the membrane with the zinc probe solution according to the manufacturer's instructions.
  6. Washing: Wash the membrane with TBST (Tris-buffered saline with Tween-20) to remove the unbound zinc probe and reduce the background signal.
  7. Detection: Detect the bound zinc probe on the membrane using an appropriate detection method. This can involve fluorescence detection if your zinc probe is fluorescent, or you may need to use a secondary antibody conjugated to an enzyme (e.g., HRP) followed by chemiluminescent or colorimetric detection.
  8. Analysis: Analyze the results, typically by visualizing the bands corresponding to zinc-bound proteins. Quantify the signal intensity using appropriate software if necessary.
  9. Controls: Include appropriate positive and negative controls in your assay to validate the results.
  10. Optional: If you're interested in determining the specific zinc-binding sites on your protein, you may perform additional experiments such as mutagenesis or zinc competition assays.
.
  • asked a question related to Assays
Question
4 answers
I am looking for a compound called Tryptoquivaline F/J (or Fumitremorgin F/J) for some validation assays. Either to purchase or synthesize. Thanks!
Relevant answer
Answer
Are you still looking for tryptoquivaline F/J (or fumitremorgin F/J)? Please contact me, if you are still interested in these.
  • asked a question related to Assays
Question
1 answer
According to Sundareshan and Khardori (2019), "The expectation is that antibiotic resistance can be prevented through the genotypic identification of bacteria and their antibiotic-resistance genes in the diagnostic microbiology laboratory. Clinical studies are still required to validate the genotypic approach to antimicrobial susceptibility testing." How do we validate DNA-based assays for antimicrobial susceptibility testing? How do we know if the approach is effective for the detection of bacteria and bacterial resistance genes?
Relevant answer
Answer
Validating the genotypic approach to antimicrobial susceptibility testing (AST) involves several steps to ensure its accuracy, reliability, and clinical relevance. Here's an overview of the process: Identification of Target Genes or Mutations, Development of Molecular Assays, Validation of Assay Performance, Comparative Studies, Clinical Evaluation, Quality Control Measures, Regulatory Approval, Continuous Monitoring, and Improvement, By following these steps, the genotypic approach to antimicrobial susceptibility testing can be thoroughly validated and integrated into routine clinical practice, providing valuable information for guiding antimicrobial therapy and combating antimicrobial resistance.
  • asked a question related to Assays
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 Assays
Question
3 answers
For H202 radical scavenging assay. How to prepare 75 uM hydrogen peroxide from 30% H2O2 solution ? Thanks in advance.
Relevant answer
Answer
Hello,
How will I calculate?
First of all I need to know the molarity of my H2O2 solution. For this I will need the density of H2O2 which is 1.11kg/L or 1111 g/L. this means that m = 1111* 0.3 (30%) = 333.3 g in 1 liter of solution.
n = m/M.
M of H2O2= 34.01g/mol
so n = 333.3/34.01 = 9.80 mol/L. So your solution is 9.80 M.
Then, to prepare a 75 µM solution you dilute the solution which is 9.80 M using the rule CiVi = CfVf
  • asked a question related to Assays
Question
1 answer
I wish to perform transwell migration assay for checking the effect of a drug on the migratory properties of Triple Negative Breast Cancer cell line (MDA MB 231). Would a difference in FBS concentration in the upper and lower chambers of the inserts act as sufficient cue to promote cell migration?
Relevant answer
Answer
Hello Titas Raha
Yes, serum is often utilised in transwell migration protocols to provide chemotactic stimuli. The cells will tend to move towards nutrient-rich media which is at a higher concentration of FBS. For this purpose, a chemoattractant gradient may be created by placing cells in the transwell insert with lower serum concentration supplemented media. Cells will tend to migrate towards the source of higher FBS concentration added to the lower chamber.
You may want to refer to the article attached below.
Best.
  • asked a question related to Assays
Question
2 answers
Hello everyone,
I want to do a chemotaxis migration assay with PBMCs. I have gone through the literature to find the optimum pore size of the transwell membrane but got confused. Some people have used 8 µm pore size while some others have used 5 µm pore size. Could somebody help me in this regard if working on a similar aspect?
Relevant answer
Answer
Transwell pore size should be chosen based on cell type and size. Generally, the selected membrane pore size should be smaller than the cell diameter when in suspension. You may perform a pilot experiment to determine the appropriate membrane pore size, which allows sufficient cell migration toward a chemoattractant while minimizing random cell migration without chemoattractant.
Generally, most leukocyte cell lines have a diameter of 10–15μm and will require a 5 or 8μm pore diameter. However, smaller cell types, such as primary lymphocytes, range between 7μm and 10μm in diameter and so may require a smaller pore size of 3μm to prevent high levels of cell migration in the absence of chemoattractant. Other cell types like fibroblast cells (for instance, NIH3T3) or cancer cells (for example, MDA-MAB 231) would require 5μm pore size.
So, I would suggest for PBMCs use a smaller pore size of 3μm since the average mean cell diameters range between 7μm - 10μm.
Best.
  • asked a question related to Assays
Question
1 answer
Hi, I've done a plaque reduction assay to analyze my possible plant activity, I used a HSV 2 virus at 6,5x10^3 as control, and I count (added file) the PFU... do I need to calculate something or I can make a graph with PFU results?
Relevant answer
Answer
That's not a great graph, because it uses a thick bar to show means and a detonator to show standard errors. It does not give any actual information about how much data you had or how they were distributed. The two strange values for group D are not evident. And the asterisk is a bit of a mystery.
What is your hypothesis? Then maybe someone can help with a recommendation for testing. Since your groups differ by dosage, it would make more sense to see if the observed values are a function of dose, rather than treating each group as a distinct treatment. This gives you a directional hypothesis, and a lot more statistial power.
  • asked a question related to Assays
Question
1 answer
I am performing fluorogenic assay with trypsin and fluorogenic assay Z-RR-AMC, my readings are decreasing with increasing time. What could be the reason for this strange results. Please guide.
Thanks in advance
Relevant answer
Answer
Excessive illumination of the the AMC fluorophore could result in photobleaching, which results in a loss of fluorescence over time. If you are using a continuous source of illumination, reduce the beam intensity by reducing the slit width on the excitation side. If you are using a flash lamp-type system, reduce the number of flashes per reading, the frequency of readings, and/or limit the duration of the measurement.
  • asked a question related to Assays
Question
2 answers
We conducted SRB assay to survey the cytotoxicity of a herbal extract with the concentration from 0.5 to 18 mg/mL. However, the inhibition percent dramatically decrease at 15 mg/mL and 18 mg/mL while it still increased at concentrations being from 0.5 to 12 mg/mL. What can lead to this phenomenon?
Relevant answer
Answer
Hi,
The phenomenon of decreased cytotoxicity at higher concentrations of herbal extracts, as observed through the Sulforhodamine B (SRB) assay, presents a paradox within the realm of pharmacology and toxicology. This observation suggests a complex interplay between the biological activity of phytochemicals and the physiological mechanisms of the cells exposed to them. To understand this phenomenon, it is necessary to integrate knowledge from various scientific disciplines, such as pharmacology, chemistry, and cell biology.
The increase in cytotoxic effects with increasing concentrations of a herbal extract can be explained by the effective interaction of the extract's active components with their target receptors or enzymes within the cell. This interaction usually follows a dose-dependent manner until a certain point. However, the decrease in cytotoxicity at higher concentrations suggests that saturation has occurred. Once the targets are fully engaged, additional molecules of the extract cannot further enhance the biological response, leading to a plateau or even a decrease in the observed effect.
Beyond receptor saturation, cells have evolved various mechanisms to protect against excessive damage from xenobiotics, including herbal extracts. These mechanisms involve upregulation of detoxification pathways, such as those involving cytochrome P450 enzymes, and activation of efflux pumps that remove toxic substances from the cell's interior. At higher concentrations of the herbal extract, protective mechanisms may be activated more robustly, reducing the cytotoxic impact of the extract by lowering the intracellular concentration of active compounds.
Another important consideration is the stability of the phytochemicals in the extract. At higher concentrations, some compounds may become unstable, aggregate, or undergo chemical modifications that reduce their bioactivity. This instability can be influenced by assay conditions, such as changes in pH, solvent interactions, or the ionic strength of the medium, which could potentially lead to misleading interpretations of cytotoxicity.
It is important to consider the impact of high concentrations of herbal extracts on cell density and nutrient availability within the assay system. This can cause added stress from nutrient depletion and altered metabolic states, leading to non-specific effects that may reduce the apparent cytotoxicity and complicate the interpretation of results.
Furthermore, the high concentrations can lead to non-specific binding of extract components to proteins and other macromolecules in the assay medium, or even to precipitation of some compounds. These phenomena can significantly reduce the bioavailable concentration of active phytochemicals, thereby diminishing the observed cytotoxic effect.
  • asked a question related to Assays
Question
2 answers
Hello,
I'm conducting a beta galactosidase staining assay for my fibroblasts using X-Gal. I dissolve X-Gal (stock 50mg/ml prepared in DMSO) in the staining buffer at a final concentration of 1mg/ml. However, even after 24-48 hours of incubation at 37°C (w/o CO2), I'm unable to observe any staining. Additionally, X-Gal precipitates in the dish with prolonged incubation, forming crystals as shown in the picture.
I have also tried heating the staining solution to 65°C before adding X-Gal, but nothing seems to work. Kindly help me resolve this issue.
Composition of my staining solution: 5mM potassium ferrocyanide, 5mM potassium ferricyanide, and 2mM MgCl2 in 1x PBS (pH 6).
For fixation, I use 4% PFA in 1x PBS for 5-10 minutes followed by PBS washes twice before adding the staining solution with x-gal.
Relevant answer
Answer
Dear Akash Mali, Thank you for your suggestion. Due to the unavailability of glutaraldehyde, I've resorted to using PFA. However, I wonder if it would make much of a difference. Kindly let me know.
Thank you
  • asked a question related to Assays
Question
1 answer
I am doing a carbonic anhydrase activity assay using p-nitrophenyl acetate.
reaction conditions: 37 °C for 10 minutes
Blank: 40ul substrate + 160 ul 50mM Tris buffer pH 7.4
crude: 40 ul substarte+ + 60 ul 50 mM Tris buffer pH 7.4 + 100ul carbonic anhydrase crude
After 10 minutes of reactions, I noticed a yellow color in all the reaction tubes except the blank, which remained colorless. To stop the enzyme reaction, I added 800 ul of 2M Na2CO3 to all the tubes. Surprisingly, even the blank changed color to yellow. As a next step, I examined the absorbance at 400 nm. However, the results obtained turned out to be inaccurate. Upon rechecking the values, I observed either lower values or, at times, even negative values. I am consistently receiving a lot of unreliable results. What steps should I take to address this issue?
Relevant answer
Answer
The pH value of your sodium carbonate solution is likely around 11.5 and I would expect your p-nitrophenyl acetate to hydrolyze rapidly at this pH value to give yellow ionized p-nitrophenol.
  • asked a question related to Assays
Question
1 answer
Hi! I was going through the literature on Phospho-Molybdate Assay (Total Anti-oxidant Capacity) and I was puzzled by the formula for the percentage anti-oxidant capacity which is given below
%Antioxidant effect =(control absorbance - sample absorbance/ control absorbance)*100
To the best of my understanding, in this assay the anti-oxidant capacity increases as the absorbance increases. This should mean the control sample should have the lowest absorbance and hence the lowest anti-oxidant capacity. It would also mean that if I were to use the formula given above, the numerator would come out as negative. Can we use negative values? Is the formula correct? Or is my understanding of the basic principle of the assay flawed?
Relevant answer
Answer
%Antioxidant effect =(((absorbance at time t) - (absorbance at time =0))/ (absorbance at time =0)))*100
  • asked a question related to Assays
Question
1 answer
Would it be appropriate to compare this value with the IC50 value obtained from the MTT assay? How can I calculate it?
Relevant answer
Answer
Yes, you can calculate the IC50 (half-maximal inhibitory concentration) value from the Ellman assay data. The Ellman assay, also known as the 5,5'-dithiobis-(2-nitrobenzoic acid) (DTNB) assay, is commonly used to measure the inhibitory activity of compounds on enzymes, particularly cholinesterases. Here's how you can calculate the IC50 value from the Ellman assay data:
  1. Perform the Ellman Assay:Conduct the Ellman assay according to the established protocol for your specific enzyme and inhibitor. Typically, the Ellman assay involves incubating the enzyme with its substrate and varying concentrations of the inhibitor. After a suitable incubation period, add the DTNB reagent to develop colorimetric signal, which is then measured spectrophotometrically at a specific wavelength.
  2. Plot the Data:Plot the absorbance or activity (enzyme activity vs. inhibitor concentration) on a graph. Use a logarithmic scale for the inhibitor concentration (x-axis) if the inhibitor concentrations are varied over a wide range.
  3. Calculate the Percent Inhibition:Calculate the percent inhibition for each inhibitor concentration using the formula: % Inhibition=(1−sample/control)×100% Inhibition=(1−Acontrol​Asample​​)×100Where A sample A sample​ is the absorbance or activity of the sample containing the inhibitor, and A controlAcontrol​ is the absorbance or activity of the control sample without the inhibitor.
  4. Determine the IC50:Use the percent inhibition values to construct a dose-response curve. Fit a sigmoidal dose-response curve to the data using a nonlinear regression analysis method (e.g., sigmoidal dose-response with variable slope). The IC50 value is the concentration of the inhibitor that causes a 50% reduction in enzyme activity or absorbance, which corresponds to the midpoint of the dose-response curve.
  5. Interpretation:The IC50 value represents the potency of the inhibitor, with lower IC50 values indicating higher potency (i.e., the inhibitor is more effective at inhibiting the enzyme). It's important to perform appropriate statistical analyses to determine the confidence intervals and assess the reliability of the IC50 value estimation.
By following these steps, you can calculate the IC50 value from Ellman assay data and assess the inhibitory potency of compounds on the enzyme of interest.
  • asked a question related to Assays
Question
1 answer
Hellow, I have been conducting an alpha-amylase inhibitory activity assay using the DNSA (3,5-di-nitrosalicylic acid) method with Acarbose as the standard inhibitor. The absorbance values obtained for Acarbose exhibit the expected trend of decreasing absorbance with increasing concentrations, indicating inhibition of alpha-amylase activity. However, when testing sample extracts, the absorbance values show an unexpected trend of decreasing absorbance from low to high concentrations. We tried passing the extract through charcoal as well, yet the trend is not changing. could anyone please suggest improvements or possibly a different working protocol to carry out the experiment?
Relevant answer
Answer
Are you saying that the sample extracts are reducing the absorbance signal when added to a reaction to which you have added alpha-amylase and starch, in a similar fashion to the alpha-amylase inhibitor acarbose? This would suggest that the sample extracts are inhibiting the activity of alpha-amylase, although some sort of interference with detection of the product is also a possibility (see below).
Is the detection of glucose by DNSA reduced by the sample extracts? In other words, if you add sample extracts to the standard curve, is the slope of the standard curve reduced? This would be a clear indication that the extracts interfere with product detection.
  • asked a question related to Assays
Question
3 answers
I am been working for a couple of months without success on setting up an assay based on GTPases loading with bodipy gdp and then measure the exchange to GTP in presence of various GEFs.
Reading on literature, the idea is that once the Bodipy GDP is loaded onto the GTPase, there is a significant increase in fluorescence (compared to Bodipy GDP alone) , which decreases when adding GEFs, which exchange it to GTP and thus releasing the Bodipy GDP.
I have been stuck on the first step, because after incubating my GTPase with Bodipy GDP I saw that there was no difference in fluorescence compared to bodipy gdp alone.
Among different protocols that didnt work, here is one:
I store my GTPases in a simple Tris based buffer, tried to buffer exchange them into HEPES buffer that the paper uses but nothing. There are other assays that instead of Hepes use Tris, I have tried those too but nothing.
i think my GTPases cannot load the GDP for some reason and i dont understand why.
If anybody had been through this assay and would like to share any tip in protein storage handling or the assay i would be grateful.
thank you!
Relevant answer
Answer
I have had success in some cases using polarization/anisotropy to measure binding of Bodipy-labeled ligands to proteins. Other fluorescent dyes can also be useful, such as tetramethylcarboxyrhodamine (TAMRA).
Attaching a large fluorescent dye like Bodipy can alter the ability of the ligand to bind, or its affinity. You should also try other fluorescent probes, if they are available.
Finally, I suggest you analyze the purity of your supply of Bodipy-GDP. Commercially supplied probes are not always of the highest quality. You may be able to find a TLC or HPLC method in the literature. It may be possible to repurify the supply using some sort of chromatography. I did this with Bodipy-penicillin using a column of Sephadex LH-20.
  • asked a question related to Assays
Question
3 answers
I am using MDA kit to measure Lipid Peroxidation in tissues. Did anyone ever used it?
I have 2 questions to ask:
Q1) Shall I use fresh or frozen tissue, which is better? I already have frozen ones.
Q2) Which is recommended to use fluorometric or colorimetric assay, since both are described in the protocol.
Thanks.
Relevant answer
Answer
Use fresh tissues as frozen samples will undergo lipid oxidation (freezer burn) which will generate MDA unless purged with argon.
The fluorometric variant can detect smaller quantities. Depends what equipment you have and how much sample.
  • asked a question related to Assays
Question
5 answers
i have recently done a disk diffusion assay where by i measure the zone of inhibition of 5 antibiotics on 7 different pathogens, i want to establish if there is any significant difference between the zones of inhibition acquired by each antibiotics to the pathogens overall. as it is a large data set i am confused as to what the appropriate statistical test would be, i have attached the excel file of my data and also my prism file. Any advice welcome , thank you!
Relevant answer
Answer
I am just curious of why you need to statistically analyze the values you got instead of categorizing them as resistant or susceptible? and then maybe calculating the percentages for each pathogen and so.
  • asked a question related to Assays
Question
1 answer
I need to order an ATP Assay Kit for my work, and I found this kit on the BT Lab website. Has anyone tried the kits from this site? Because this is the first time I will order from them.
Relevant answer
Answer
no
  • asked a question related to Assays
Question
1 answer
Hello everyone,
I'm currently conducting the BCA assay for a membrane protein present in a solution containing 1XTBS buffer, 10% glycerol, and 0.2% DDM/CHS (in a 10:1 ratio). I have a couple of questions regarding this setup:
  1. Does the detergent DDM/CHS interfere with the BCA assay in any way? I could not find them in the list of chemicals provided.
  2. Does the BCA assay measure the native state of the protein, denatured protein (treated with SDS), or both? If it measures both, would the protein assay yield different results when the protein is in its native state compared to when it's denatured?
I would appreciate any insights or experiences you may have on these matters. Thank you in advance for your help!
Relevant answer
Answer
Detergents should not interfere with the BCA assay. The conditions of the assay are moderately strong base, which should denature most proteins. So, in general, the denatured state is measured.
  • asked a question related to Assays
Question
1 answer
Hi,
I'm in the process of developing a hydrolysis probe qPCR assay to quantify gene copy numbers of Microcystis 16S rDNA. In troubleshooting my cycling parameters, I'm getting resulting plots such as the attached photo, where a plateau phase is not reached but rather a secondary jump in fluorescence is observed in the late stages. I ran NTC samples on this run (they're labeled "Blank" in the photo), and no fluorescence was detected at all, indicating this is not due to primer dimers.
Does anyone have knowledge of certain assay parameters that may lead to a plot such as this? Possibly related to annealing or extension times/temperatures? Could inhibitors in my template cause something like this?
Relevant answer
Answer
hi mate, this link may help you and may arise from high template concentration just dilute samples 10x - 100x and check again
  • asked a question related to Assays
Question
9 answers
Hi
I am currently working on lateral flow assay (LFA). I found that my negative control gave false-positive results in my test. I have checked using ELISA and did not face this problem. I blocked my LFA assay with BSA, but the false-positive still occurred. I am using PBS as my negative control.
Has anyone experienced the same issue?
Relevant answer
Answer
Ahmed Jehanli thank you for your suggestion. I treated the nitrocellulose with 1% BSA after lining the capture antibodies. My conjugated quantum dot-detection antibody contained 1% BSA too.
Is it because the concentration of the
detection antibody is too high that caused non-specific happened?
  • asked a question related to Assays
Question
2 answers
I have ordered the SOD colorimetric kit (invitrogen) and a GSH/GSSG ratio assay (abcam, ab205811) and was hoping to use the same brain tissue for both kits. In the methods provided the by the manufactures, the GSH/GSSG assay requires PBS/0.5% NP-40 and the SOD assay just requires PBS. Does anyone have experience with these kits and can advise? Could NP-40 be left out if cells are throughly homogenised with a ultrasonic homogeniser since this is just a detergent?
Really appreciate any advice.
Relevant answer
Answer
Be careful. Although you may achieve sufficient cell lysis to release cell's glutathione and enzymes into the medium, glutathione oxidizes very rapidly in neutral pH. I am not sure about the specific kit you mentioned, but a common practice in measuring glutathione with colorimetric methods is to prepare the cell lysate in acid (e.g., trichloroacetic acid or sulfosalicylic acid). The use of acid serves two purposes, 1. to keep the pH and avoid oxidation, and 2. to precipitate proteins that might interfere in the assays.
  • asked a question related to Assays
Question
1 answer
Hello
I need suggestions, As I wanted to estimate sugars from bacteria(pellets/biomass) sample by phenol-sulfuric assay but not sure how to extract carbohydrates from the bacteria source.
Thanks
Relevant answer
Answer
Dear Nishitha Landgey ! It is necessary to destroy bacterial cells with a tissue shredder or ultrasound, and then carry out centrifugation. You will get a solution of carbohydrates in water.
  • asked a question related to Assays
Question
3 answers
We will be conducting an enzymatic assay using a-glucosidase and a-amylase using acarbose as the positive control. What should be our negative control?
Relevant answer
Answer
Add stopping reagent to assay buffer (generally shifting pH to inactive the enzymes). Next, add the enzyme to mixture to generate a negative control...
  • asked a question related to Assays
Question
1 answer
Hello! I am not sure whether or not my protein samples are reduced or not. Based on the information below, can someone please let me know if they are reduced?
Sample setup:
__ uL total cell lysate (exact amount calculated from BCA assay)
1 uL Thermo Fischer 10X Sample Reducing Agent
2.5 uL Thermo Fischer 4X LDS Sample Buffer
1X RIPA buffer to dilute sample to 10 uL
Relevant answer
Answer
Dear Kristen,
Protein samples are typically reduced before loading onto SDS-PAGE using a loading buffer containing a reducing agent such as DTT or b-mercaptoethanol (and the sample is also often boiled to further denature the proteins).
If you dilute your protein sample first with RIPA buffer and then add Sample Reducing Agent (and perhaps boil it for 60 - 90 seconds) all of the proteins should be reduced. You can then add some of the Sample buffer and load directly onto your SDS-PA gel for electrophoresis. (I am sure that Thermo Fisher would have supplied a standard protocol for this!).
You should take care to keep all of the buffers at the correct temperature, and always check that the Reducing Agent always smells a bit like rotting eggs as DTT and b-ME go off quickly (I always store these in the fridge but Thermo Fisher may recommend freezing the Reducing agent).
Andrew
  • asked a question related to Assays
Question
1 answer
Both TTC and NBT yield formazan when interacting with oxides. The superoxide dismutase assay requires NBT, which reacts with oxide to form formazan. TTC also produces Formazan when it interacts with oxygen. So, can I use triphenyl tetrazolium chloride (TTC) instead of nitroblue tetrazolium (NBT) to estimate SOD activity?
  • asked a question related to Assays
Question
4 answers
Hello everyone
I am trying to design an MTT experiment for an immortal adherent cell line with doubling time of 20 hours. The goal is to evaluate effects of certain growth factors and their combination on cell proliferation and determine the optimal dose.
Many papers suggested seeding 10000 cells in 96 well plates and performing MTT at 24 h intervals or at days 1, 3, 7.
I am not sure about some technical issues:
1. should I proceed with 24 h interval assay and for how many days? or the 1, 3, 7 days evaluation is good enough?
2. can I seed 5000 cells/ well in 96 well plates instead of 10000?
3. I expect my cells to become confluent after 72 hours (starting from 10000 cells), and they definitely die without medium change, so should I change the medium every 24 hours for all plates? or just change the medium after 72 hours and wouldn't this affect my results?
Thanks a lot!
Relevant answer
Answer
Thank you very much
I learned a lot
  • asked a question related to Assays
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 Assays
Question
2 answers
I am doing a resazurin assay to determine the MIC of plant extracts against Mycobacterium tuberculosis. The problem is the MIC is coming at a very high value. I read in one paper that even if there are 80 cells the color changes from blue to pink. I am diluting the inoculum also (1:20 dilution). How to solve this problem. Kindly give suggestions. Should I dilute the inoculum more?
Relevant answer
Answer
Yes I have ..1st line anti-TB drug..Isoniazid
  • asked a question related to Assays
Question
2 answers
BSA assay and Fluorometric peptide assay do not work for the quantification of the Pepstatin A (aspartic protease/cathepsin D inhibitor/peptide). Does anybody have an idea to quantify it besides using LC-MS?
Relevant answer
Answer
Somewhat less complicated than LC_MS I would suggest reversed-phase LC in combination with UV detection at 218 nm employing a proper (thus not to steep!) acetonitril gradient using 0,1% TFA.
  • asked a question related to Assays
Question
2 answers
We developed a rapid viability test for plant seeds in which yeasts metabolize organic compounds that leach from seeds (dependent on the physiological age of the seed) and thereby reduce our redox-indicator resazurin, accompanied by a color change (5). Unfortunately, seeds of some species acidify our test solution causing a pH-dependent colour change of the resazurin (abiotic reaction) which distorts our test results (6). We are looking to replace our redox indicator dye for these cases!
We already researched on several dyes from the tetrazolium-family that unfortunately also change their colour upon acidification, i.e., MTT (1), TTC (2), MTS (3) or the solubility is severely limited, e.g. MTT (3), XTT (4).
We are grateful for any advice!
1: Plumb et al. (1989) Effects of the pH Dependence of 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium Bromide-Formazan Absorption on Chemosensitivity Determined by a Novel Tetrazolium-based Assay. Cancer Res 49: 4435–4440.
2: Lopez Del Egido et al. (2017) A Spectrophotometric Assay for Robust Viability Test of Seed Batches Using 2,3,5-Triphenyl Tetrazolium Chloride: Using Hordeum vulgare L. as a Model. Front. Plant Sci. 8: 747. doi: 10.3389/fpls.2017.00747
3: Riss et al. (2013) Cell Viability Assays. https://www.ncbi.nlm.nih.gov/books/NBK144065/
4: Goodwin et al. (1995) Microculture tetrazolium assays: a comparison between two new tetrazolium salts, XTT and MTS. J Immunol Methods 13: 95-103. doi: 10.1016/0022-1759(94)00277-4
5: Mohammed et al. (2019) Dead or Alive: Simple, Non-destructive, and Predictive Monitoring of Seedbanks. Trends in Plant Science 24: 783-784. DOI: 10.1016/j.tplants.2019.05.014
6: Wellmann et al. (2023) Maize Grain Germination Is Accompanied by Acidification of the Environment. Agronomy 13: 1819. doi.org/10.3390/agronomy13071819
Relevant answer
Answer
@ Zardar Khan
Dear Zardar,
thank you for your response, but Alamar Blue is a trade name of resazurin, which we are already using.
Klaus
  • asked a question related to Assays
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 Assays
Question
1 answer
I am conducting a series of assays in which I incubate intact human keratinocytes in culture with a variety of metabolic modulator compounds, then lyse with a gentle buffer and use the lysate for a kinetic plate reader assay for lactate dehydrogenase activity (abcam ab102526). Typically, I immediately proceed to the LDH activity assay after lysing the cells and assessing protein concentration, but for longer incubations I am wondering if there is a safe stop after the lysis step which would be more convenient timing wise. Could I store the lysates at this point and perform the LDH activity assay the next day without losing significant enzymatic activity? If so what storage conditions would be optimal?
Relevant answer
Answer
You could freeze the lysate at -80 degree C. Do not forget to add a cocktail of protease inhibitors to the cell lysis/extraction buffer before lysing the cells. I would recommend the addition of phosphatase inhibitors as well to the lysis buffer since enzymes are activated by phosphorylation and dephosphorylation may cause a loss of enzyme activity.
Another important point to note is that you should aliquot your sample lysate for future use as multiple freeze-thaw cycles will damage your protein of interest. Storage of protein lysate under such conditions may help for a few months before losing significant activity.
Best.
  • asked a question related to Assays
Question
4 answers
I'm doing mic and mbc following the disc diffusion assay. I want to calculate the cfu value for the microbial growth in the plate. Can you please share any relevant article or the exact calculation?
Relevant answer
Answer
Your enumeration of microbes as Colony Forming Unit ( CFU), you should be very perfect what type of microbes you are dealing with. Accordingly you should proceed serial dilution pour plate or spread plate technique. For instance, if you are dealing with bacteria, your accurate dilution will be 10‐⁴, 10-⁵ and 10-⁶ . Suppose you get 30 colonies in 10-⁵ by inoculating 0.1 ml, Then your CFU will be average count x dilution factor and find out as CFU per ml or per gram as follows: 30 x 10⁵= 3x 10⁶ for 0.1 ml So CFU per ml will be 3 x 10⁷. It is better to express as log value. Your answer is 7. 477. Why log value? Because exponential growth of bacteria is always in log phase. If you deal with Actinobacteria , go one step lower dilution I.e., your sampling will be 19³, 19⁴ and 10⁵ ( because Actinobacterial population is less than bacteria in normal soil). Similarly, when you deal with fungi, choose the dilution 19², 19³ and 10⁴. Hope, I could clear your doubt.
  • asked a question related to Assays
Question
4 answers
I have dried leaves from my past experiment and need to conduct SOD activity assay. Can it be done in dried leaves?
Relevant answer
Answer
Shafuq Abbas Can you please send me a paper for the protocol?
  • asked a question related to Assays
Question
1 answer
Hello everyone,
After exposing HeLa cells to phthalate, I will detect various antioxidant levels with an ELISA kit. Before ELISA, I will determine the protein concentration of the cells with BCA assay. During BCA, after lysing my cells with RIPA buffer, I will dilute them 2x, 5x and 10x with distilled water. My question is, what is the number of cells that need to be seeded per well for BCA assay? I'm open to your suggestions. Thank you!
Relevant answer
Answer
Hello Naile Merve Guven,
It would not be proper to give you the cell number because it will vary with cell type. So, you will have to perform a pilot experiment to decide the optimum cell density for a multi- well plate format which you will be using for the actual experiment.
In the pilot experiment, you may seed different cell densities and choose the optimum cell density that gives you 80% confluency in 24 hours. For instance, for HeLa cells in a 96-well plate format you may seed cells in the range of 5000 to 25000 cells per well in 200ul, and choose that cell density which gives you 80% confluency after 24 hours.
After having obtained the optimum seeding density from the pilot experiment, you may seed cells at that optimum cell density in whichever plate format you decide, and after 24 hours you may carry out the treatment.
After treatment, you may lyse the cells with RIPA buffer and determine the total protein of the lysate using BCA assay with different dilutions of the lysate.
Best.
  • asked a question related to Assays
Question
2 answers
We tried to do activation-tagging in Arabidopsis using the plasmid pSKi015. Nevertheless, we failed to obtain positive clones in GV3101? The recommended Agrobacterium strain for pSKI015 is GV3101 pMP90RK. Does GV3101 also work? Can carbenicillin be used for screen the positive clone instead of Ampicillin?Thanks!
Relevant answer
Answer
Thanks, Dr. Stracke! @ Ralf Stracke
  • asked a question related to Assays
Question
1 answer
This is from the oncomine comprehensive assay v3 protocol. Does "gDNA (10 ng, ≥0.67 ng/μL) " mean that the concentration range of gDNA should be from 10 ng to ≥0.67 ng/μL, and If I already have 10 ng/ul, do I set up a reaction without Nuclease-free Water?
Relevant answer
Answer
I think you mast have ≥0.67 ng/μL concentration in one well. You can get it from adding 10ng gDNA in 15 ul water . (10/15=0.67).
  • asked a question related to Assays
Question
3 answers
hello everyone
my samples are HeLa cells. i‘ll use them for ELISA assay. before elisa, i’ll perform BCA assay. during bca, i’ll dilute my samples.
what do you recommend for dilution solution and dilution ratio?
thank you in advance!
Relevant answer
Answer
Sometimes you can use samples without dilution. Usually depends how much material you used for extraction.
  • asked a question related to Assays
Question
6 answers
Hi,
I performed a competition assay of serially diluted concentrations of the unlabeled peptide by adding a constant concentration of the FITC-labeled peptide. I have observed higher fluorescence intensity in higher concentrations of unlabeled peptide compared to the lower concentrations of the unlabeled peptide. As per literature we should have lower intensity in higher concentration so of unlabeled peptide. Could you kindly clarify why this is happening or should it be the opposite.
Relevant answer
Answer
Thank you for your suggestions.
  • asked a question related to Assays
Question
1 answer
I have been working on antioxidant activity using FRAP assay. But, the correct way for calculate FRAP value is difficult to find, Can anyone help?
Relevant answer
Answer
You read the original paper for the method? ( ).
  • asked a question related to Assays
Question
2 answers
Hello, I am currently isolating nuclei from adipose tissue and I am interested in obtaining high-quality RNA for further assays. What extraction methods do you recommend?
I´m currently using the TRIzol protocol, without modifications but in addition to obtaining very little amount of RNA, the ​​280/260 and 280/230 values are very low, any suggestions?
Relevant answer
Answer
i agree with Emilee Bargoma RNA from tissue can be tough. are you getting good nuclear isolation? if so then use a lysis buffer with sodium dexoycholate (RIPA) to help punch holds in the membrane and lyse on ice for up to 30 minutes then perform RNA isolation. if you are not using a column then be very conservative when you are transferring phases and err on the side of less but clean RNA rather than trying to get every single ul of aqueous phase
  • asked a question related to Assays
Question
1 answer
Hi guys,
Can anyone recommend a good protein extraction buffer for enzymes? After extraction I would like to test the enzyme for its activity in an assay, so it should still be active in the buffer.
Many thanks in advance!!!
Relevant answer
Answer
There is no such best protein extraction buffer for enzyme, when i work with new protein, typically I use 50 mM Tris.Cl, 10% Glycerol, pH 7.1. and if there is a possibility of aggregation by the known sequence structure (such as any disulfide bond, etc) I will add BME or DTT in the buffer
  • asked a question related to Assays
Question
1 answer
Catalase assay for bacteria - not screening test with hydrogen peroxide
The complete procedure using spectrophotometer
Relevant answer
Answer
This published protocol may help you:
Rapid Kinetic Microassay for Catalase Activity
Yi Li and Herb E. Schellhorn
J Biomol Tech. 2007 Sep; 18(4): 185–187. PMCID: PMC2062561 PMID: 17916790
  • asked a question related to Assays
Question
1 answer
I want to conduct haemolyis assay on mouse blood (I need 0,5 - 1 ml of blood), I know that it is generally hard to collect good sample without inducing haemolysis. As a anticoagulant do you prefer EDTA or heparin? Syrgine must be coated with anitcoagulant? And need I special tubes that are coated with anticoagulant or can I just prepare solution of anticoagulant in a tube? As I understand blood should be collected from heart or junglar vein? Do you have any recommendations?
Relevant answer
Answer
Hi Alicja,
We collected blood from mice after euthanasia from the portal vein. It was a very long time ago, but I will try to remember the stages of blood sampling. First, 50-100 μl of heparin was drawn into the syringe, so that the solution was mainly in the needle. Immediately after cutting the abdominal cavity, the intestines were moved away, the vein that led to the liver was found, and a needle was carefully inserted into it. The needle should not be too thin to avoid hemolysis, the syringe plunger should move smoothly. After the blood had collected, the needle was removed and the blood was squeezed into the test tube.
  • asked a question related to Assays
Question
3 answers
If I am testing the antimicrobial activity of a bacteria in a specific agar (like Nutrient Agar), shouldn't I use the same media for other tests such as biochemical analysis?
I am a bit lost as to why my superior would choose another agar (ISP 2-7 agar). Although that agar is specific for that microorganism, shouldn't I check the biochemical activity also present during the antimicrobial activity assay done in NA?
EDIT: I am using Mueller Hinton Agar. I just wrote NA since that was the initial agar they chose during the proposal.
Relevant answer
Answer
"NA" presumably is nutrient agar. This is not ideal for antimicrobial activity testing by the disc method, and a dedicated susceptibility test agar should be used.
  • asked a question related to Assays
Question
10 answers
Hello everyone, I'm working with predicted antimicrobial peptide using machine learning. Recently, we use an antimicrobial peptide from DRAMP which had been verified to have antimicrobial activity. We synthesized the peptide exactly as what provided in DRAMP. so we did test using disc diffusion assay but no activity. is there any possible issue like it must be tested in microdilution method or due to the synthesis parameter? Thanks in advance for the help and idea.
Relevant answer
Answer
Did you characterize the peptide, both with bioinformatics and experimental? I mean:
-Using for example Heliquest (https://heliquest.ipmc.cnrs.fr ) you get a proper idea about the peptide (its hydrophobicity, net charge, etc.). See for classification for example:
-Before checking in your assay, I think it is good to check the peptide in some way. If for example bioinformatics predicts membrane activity you can check quickly by using a monolayer technique or depending on what you have available in the lab check by circular dichroism (with and without lipids), see for example:
If this seems fine, then do realise that sometimes a pretty insoluble peptide (as described in the link above) that requires something like DMSO or TFE is all of a sudden reasonably soluble in aqua dest. as described here:
However, in your medium there will be a buffer containing salt (and a certain pH) and the ionic strength in the medium can cause aggregation (this can be checked with the peptide and the use of for example circular dichroism).
Obviously, something might have gone wrong with the synthesis, or the prediction is simply wrong, and the peptide has no antimicrobial activity but a lot can be checked before one have to draw this conclusion.
Best regards.
  • asked a question related to Assays
Question
2 answers
We have developed HPLC method for estimation of Sodium metabisulfite in oral suspension. Method validation completed with Spiked samples.
Question is when we analyse actual test product, assay values obtained are less than 10% of label claim. When confirmed that input in product is correct.
Need suggestions.
Thanks
Relevant answer
Answer
Typically, concentrations of Sodium Metabisulphite are determined by titration with Potassium Thiosulfate using a starch endpoint. In this case it sounds more like the metabisulphite has been transformed by the sample matrix into another ionic form, thus we need more information (like sample matrix, chromatographic conditions, column, mobile phases, detector...)!
  • asked a question related to Assays
Question
4 answers
Hello,
I have to quantitate free amino acid in my sample using LC-MS/MS. My sample mainly is an assay for checking the enzyme activity of phenylalanine hydroxylase in converting phenylalanine to tyrosine. Mostly researchers are using ez-faast kit but I want to know about AccQ tag from waters or any other alternative.
Can you please help me find a kit that suits my assay? Iam completely new to this field..
Thank you in advance
Relevant answer
Answer
Yes, Chromservis can supply MetAmino kit for both, LC/MS and GC/MS. See https://www.chromservis.eu/en/metamino-kit-info. If you are interested in a specific matrix, let me know. There is an option to test various matrices.
  • asked a question related to Assays
Question
1 answer
I want the procedure for Nitric oxide assay in plant sample.. Please kindly help in this regard at the earliest .. send an email to [email protected].
Relevant answer
Answer
Hey there Veluchamy Balakrishnan! Nitric oxide assay in plant samples, huh? Well, buckle up, because I got you Veluchamy Balakrishnan covered! First things first, you'll need to gather your gear and prepare for some serious plant chemistry.
1. **Materials:**
- Plant samples (obviously!)
- Griess reagent (sulfanilamide and N-1-naphthylethylenediamine dihydrochloride)
- Phosphate buffer
- Sodium nitrite standard
- Microplate reader
2. **Procedure:**
a. **Sample Preparation:**
- Grind your plant samples into a fine powder.
- Extract the nitric oxide using a suitable buffer.
b. **Reaction Time:**
- Mix the extracted solution with Griess reagent.
- Allow it to react in the dark for about 10-15 minutes.
c. **Measurement:**
- Measure the absorbance at a specific wavelength (around 540 nm) using the microplate reader.
- Compare against a sodium nitrite standard curve to quantify the nitric oxide levels.
3. **Analysis:**
- Calculate nitric oxide concentrations in your samples based on the standard curve.
- Don't forget statistical analysis for that extra scientific flair.
Now, my friend Veluchamy Balakrishnan, you're armed and ready for some serious nitric oxide analysis in your plant samples. Go forth and conquer the world of plant biochemistry! If you Veluchamy Balakrishnan run into any hurdles, you know where to find me for more wisdom.
  • asked a question related to Assays
Question
1 answer
Is Transition Pore Assay a good assay for assessment of cardiac mitochondrial function in large animal experiments?
Relevant answer
Answer
Depends on what you are looking for. If you want to study mitochondrial permeability transition, processing the cells as soon as possible after euthanising the animal is recommended.
  • asked a question related to Assays
Question
3 answers
I have been facing the issue of False negative results in both Dengue IgG & IgM assays. I have tried various buffer compositions with commonly known blockers to resolve sample interference but unable to resolve the false negative issue.
Relevant answer
Answer
The occurrence of false negatives and false positives in your results is intricately linked to the sensitivity and specificity of the diagnostic protocol. One effective strategy for optimization involves utilizing known positives and known negatives during the experimental procedure. Subsequently, the development of a Receiver Operating Characteristic (ROC) curve can aid in identifying an optimal cutoff point. This is the point at which both sensitivity and specificity approach 100%. We trust this suggestion proves beneficial to your work.
  • asked a question related to Assays
Question
1 answer
"The reactivity of peptides was confirmed using western blotting and enzyme-linked immunosorbent assay (ELISA) assays"
Relevant answer
Answer
Assuming the peptide functions by binding. The target protein could be immobilized on a plate or transferred to a blot. Then the peptide (likely also with fluorescent / chemiluminescent tags) could be applied to bind to the protein for detection.
  • asked a question related to Assays
Question
2 answers
I would like to differentiate the presence of Escherichia coli K-12 in my samples by qPCR. In particular I would like to discriminate between K-12 and HB101. The problem is that these two strains are quite related.
Is there a qPCR that can specifically target K-12?
Is there an assay that can quantify K-12 over HB101?
Thanks
Relevant answer
Answer
Thank you I am already using this PCR but does not help me differentiate K-12 from HB101...
  • asked a question related to Assays
Question
2 answers
can I use the cox activity assay kit with fish liver lyophilization samples??
Relevant answer
Answer
Dear Dur. Thanks a lot for your answer. however, the kit's manual doesn't provide specific guidance about this option or a protocol for reconstitution...do you have extra info?
  • asked a question related to Assays
Question
3 answers
According to the Kit instruction, I'm using 10 to the power 6 cells (platelets). But the no. of events while recording the reading doesn't even cross 10. Kindly suggest me a solution .
Thankyou
Nilanjana Ghosh
Relevant answer
Answer
The concentration you mentioned usually works well. Check cytometer settings for proper gating of platelets and volume/time/events the software collects/acquires for analysis. Also make sure that you are taking enough sample volume.
  • asked a question related to Assays
Question
3 answers
What is the role of PVP (Polyvinylpyrrolidone), when we use it in phosphate buffer for antioxidant enzymes activity assays? such as assay of SOD or POD activity in plant cell....
Relevant answer
Answer
Many Thanks for your complete and useful answer. Mohammed Danouche
  • asked a question related to Assays
Question
1 answer
For an assay result of a particular sample that was repeatedly tested, I would like to know whether is it ok to average the result of the different times the assay was performed? The sample here is a plant-based extract. It would be great if you could provide an example of a publication that shows this situation and one that is related to assay-based fractionation.
Relevant answer
Answer
You use the repeated-measures t-test, also known as the paired samples t-test, to assess the change in a continuous outcome across time or within-subjects across two observations :)
  • asked a question related to Assays
Question
1 answer
I am studying precision for my assay/kit with the following parameters followed:
Panel members to be tested:
1. Weak positive
2. Medium positive
3. Strong positive
4. Negative
All these were panel members were tested in triplicates in three lots of the kit tested on 3 days, at 3 different sites by 3 persons.
Now, I have to calculate inter assay variation, intra assay variation, person-to-person variation, day-to-day variation and site-to-site variation.
Please help me if this assay design is correct to calculate the precision of the assay and how to calculate the variation mentioned above?
Thank you in advance.
Relevant answer
Answer
Hi
You need to calculate standard deviation and coeffficient of variation.
  • asked a question related to Assays
Question
3 answers
I am working with NOVATECH ELISA KIT for Brucella. In the leaflet, it is mentioned that it is a qualitative assay with a cutoff starting at 11. When I enrolled the assay through the DS2 automatic ELISA machine, the layout results showed values as numbers, not only positive and negative. What does it mean? Are those numbers the real titre value? Are they the real concentrations of measured antibodies? If yes, why has the company not highlighted the kit as a quantitative ELISA?
Relevant answer
Answer
ELISA may be run in a qualitative or quantitative format.
Qualitative results provide a simple positive or negative result (yes or no) for a sample. The cut-off between positive and negative is determined by the analyst and may be statistical.
For diagnostic tests that produce a numerical result, the point above which test results are classified as positive is called the cut-off. These are usually established by testing a large number of infected and non-infected animals and selecting the value that maximizes the sensitivity and specificity of the test. For diagnostic kits the cut-off is established by the kit manufacturer. You may want to refer to the article attached below.
In quantitative ELISA, the optical density of the sample is compared to a standard curve, which is typically a serial dilution of a known concentration solution of the target molecule. For instance, if a test sample shows an absorbance of 1.0, the point on the standard curve that gives an OD = 1.0 will be of the same analyte concentration as the sample.
So in this case, the samples showing values 11 and above are considered positive.
Best.
  • asked a question related to Assays
Question
3 answers
Hello everybody,
I want to perform a zymography assay to evaluation of MMP2- and MMP-9 activity. In my case after the treatment of cells, cell supernatant was gathered and centrifuged to remove the cells' debrides. Now I want to save cell supernatant, but I don't know what is the best temperature for cryopreservation of cell supernatant.
Relevant answer
Answer
Vatamanu Sergiu-Marian Hi, I am trying to do zymography and use a lysis buffer with Tris, NaCl, and NP-40, but the sample stays very viscous making it very difficult to load on the gel. How should I process it to use so that the enzyme activity is also retained? Do you have any suggestions?
Thank you
  • asked a question related to Assays
Question
2 answers
Dear all,
A very good day!
I need to quantify the amount of poly-gamma-glutamic acid levels in my samples, but I do not want to use chromatography methods or western blot. Are there any easy-to-use kits for colorimetric absorbance assays to recommend to me? Thank you all for your kind assistance and have a great day!
Relevant answer
Answer
CTAB method
  • asked a question related to Assays
Question
1 answer
Hi , My over all experimental goal is to define Zika virus-specific T cell responses in macaques infected with Zika virus. We also have control macaques that were not infected with Zika. We used PBMCs obtained from 14-28 days after Zika infection. PBMCs and stimulants were arranged in the Elispot plate as shown below.
Viability - determined with trypan blue assay
PBMC preservation - frozen in DMEM with 10% FBS, antibiotic, L-glutamine+10% DMSO
Elispot assay: stimulant and PBMCs mixed together prior to adding to well, incubated ~17 hours overnight>
Expected results: should see lots of spots in all CD3 wells, and spots in all the wells from a Zika animal stimulation with UV-inactivated Zika virus or Zika peptides
Actual results: only see spots in PBMCs from 1 Control animals stimulated with CD3.
No spots in Zika PBMCs stimulated with CD3 Ab. No spots with Zika stimulants.
Any suggestions?
Do you think changing the format to Cultured Elispot help?
Thanks for your response.
Saswati
Relevant answer
Answer
Dear Sawati,
try to rest your cells for 24h in the appropriate medium, no stimulant. after 24h of resting in the CO2 incubator, wash them and count them again.
PBMCs tend to not react shortly after thawing. Try this! And, maybe it makes no difference, but I would recommend to use no antibiotic for Cryopreservation.
Good luck!
  • asked a question related to Assays
Question
7 answers
Respected expert, as I have done antioxidant assay for zinc oxide nanoparticle using medicinal plant Serriphidium kurramense. In this assay I have used Ascorbic acid as standard and DPPH as free radical. In the assay for control 2ml methanol and 1ml DPPH is used, and different concentrations of nanoparticles and plant extract are used as test samples against DPPH. The question is, my test sample that is nanoparticle always give higher absorption value then control due which I get wrong result and my sample showing no antioxidant activity while the plant used in the synthesis show antioxidant activity. Is it possible that the plant showed antioxidant activity but the nanoparticles synthesized from that plant show no antioxidant activity. The change from pinkish color of DPPH to yellow or colorless is completely done in case of Plant Extract and standard Ascorbic acid but show antioxidant activity not more than 80% when calculated although color change is 100% in both plant extract and standard and in case of NPs no color change occurred.
File is attached for reference. Please help what should I do in this case?
I have repeated the assay many time but got no positive result.
Your precious time and comments will be highly appreciated.
Relevant answer
Answer
I would like to express my sincere gratitude for the time and attention you have generously given me. Your expertise and guidance have been truly invaluable. I have shared the detail via email, if you kindly check it Mohanprasanth Aruchamy
  • asked a question related to Assays
Question
1 answer
Name of Assay: Albumin Denaturation Assay
Relevant answer
Answer
There is no trend in the data, so there must be some technical problems with the assay. One possibility is that the inhibitor is interfering with the measurement due to effects such as insolubility or optical interference.
  • asked a question related to Assays
Question
4 answers
Hello everyone ,
I am following a protocol where I treat my cells with 1 .. 2.. or 3 compounds
and I do an atp cell titer glo assay
I noticed that the more compounds my cells receive . the lower their viability is .. which messes up everything
did anyone face this issue before? does it make sense ?
Thanks!
Relevant answer
Answer
The compounds themselves may be mildly cytotoxic. Adding up the mild cytotoxicity of multiple compounds may lead to the loss of viability you are seeing. It is also possible for there to be synergy between compounds in terms of their cytotoxicity, which means that the effect of the combination is greater than the sum of their individual effects.
  • asked a question related to Assays
Question
10 answers
What are the solutes used to dilute essential oil for DPPH, TPC, FRAP and Beta/Carotene Bleaching Assay?
Can I simply dilute the essential oil using
1 mg/mL in methanol/dH2O for all assay?
and can I use the solutes listed in the article included
Relevant answer
Answer
@Farah Fatihah Yes, you can start with the concentration of your choice, it is up to you. Then, according to the results you obtain you'll adapt.
The first thing to do is to review the works related to your plant, to have an approximative estimation of where to start.
For the dosages it is simple, we don't need dilutions since in the protocoles we always take from the 1mg/ml sample concentration and perform it in triplicates (three repetitions)
For the ATox' activities, let's assume you started with 1mg/ml, prepared 6 dilutions out of it and analysed it. There are 3 possibilities:
1 : The absorbance values of your dilutions contain an interval where 1/2 initial absorbance of reagent is reached, which means you reached the IC50. You're good, no need to do anything else, and you have a good or very good ATox' activity
2: Even for your most diluted sample, the absorbance values are lower than 1/2 the initial absorbance of reagent, which means all the reagent was inhibited in the solution, you don't have the IC50 because your sample is too strong, you need to dilute it more and you have a very good to excellent ATox' activity.
3: Even for your most concentrated solution, your absorbance values are higher than 1/2 reagent absorbance, your sample is too weak and couldn't inhibit 50% of reagent. You'll need to start with more concentrated solutions.
Why do we look for 1/2 Initial absorbance of reagent? Simply because it represents the inhibition of 50% of it, which is the IC50. If I start with an ABTS adjusted to 0.700, my IC50 will be somewhere in the interval of absorbances that include 0.350
If you have any other questions or concerns, please feel free to DM me and we'll organise a visio meeting where we will discuss whatever you want in this area
  • asked a question related to Assays
Question
1 answer
Something went wrong with our cell incubator over the weekend so to my knowledge the cells inside were deprived of a sufficient level of Carbon Dioxide for about 24 hours. The photo attached shows the state of the cells.
The cells are NTERA-D2 cells. Passage number 16.
Magnification is 10X.
Cells were passaged 3 days ago into a T-25 flask.
My question is; Can I salvage these cells so they can be used for rtq-PCR and colony formation assays? Or should I just thaw a new vial of NTERA cells?
Relevant answer
Answer
Without CO2 supply, cells are adversely affected within five minutes. Usually, when there is a technical fault in the CO2 incubator and CO2 supply is shut, one can minimize pH change by supplementing the media with 25 mM HEPES buffer. Cells can proliferate without CO2 if the medium is buffered with 25 mM HEPES at pH 7.4, but this environment is viable for no more than 10 hours and is highly dependent upon type of cell line and cell concentration.
Cells deprived of sufficient level of CO2 for about 24 hours may be stressful to the cells, and may result in irreversible damage. I would recommend that you should not waste time salvaging these cells as they would not be healthy as before and therefore unsuitable for any cell-based assay. Since you have NTERA-D2 cells in stock, you should go ahead and thaw a new vial.
Best.
  • asked a question related to Assays
Question
3 answers
Currently working on a USP assay method for HPLC that requires water-saturated butyl chloride as a mobile phase and water-saturated chloroform as a diluent.
What is the whole point of using water-saturated solutions like this?
Would there be alternative ways to substitute these solutions?
Relevant answer
Answer
Using water-saturated butyl chloride as a mobile phase and water-saturated chloroform as a diluent in HPLC maybe to enhance sensitivity, aid in phase separation, or reduce ionization. The specific reason for using such aqueous form of solution could differ based the type of assay and the APIs or analyte being analyzed. Alternatives can be considered, but method validation (still dependent on the analyte involved) is essential to ensure the reliability of results.
  • asked a question related to Assays
Question
5 answers
I have been struggling to obtain the concentration from microplate reader that match up with the Qubit reader. Approximately, they differ by factor of 10.
The Qubit is tolerable for few samples, but if we were to measure for the whole 96 plate, we want to use the microplate reader.
The example of discrepancy is attached in the picture.
The only factor I could think of that may be messing up the values are how I set up the dilution factor series.
Any suggestions/advice would be greatly appreciated!!
Relevant answer
Answer
Daniel Carneiro Moreira Thank you very much for your insight. I did not think about the differences in the pathlength. I will reassess the sample concentration.
  • asked a question related to Assays
Question
1 answer
The reviewer requested that the authors indicate the detection limit for the viral titers assay. However, the assay was performed using PFU, and I am unsure how to determine the assay's detection limit.
Can someone understand the reviewer's request?
Coul help me? Please?
Relevant answer
Answer
Dear Carolina,
The detection limit may refer to the minimum or the maximum number of viable phage you can detect using (e.g.) a soft-agar plating method, and both can be valuable for different reasons.
The maximum detection limit is simply the highest dilution of your suspension which produces one or more plaques. If you tested a 1/1,000 dilution (and saw one or more plaques) and a 1/10,000 dilution (and saw no plaques), the maximum detection level for an assay that only went up to a 1/10,000 dilution would be 9,999 PFU (or <10,000). The maximum is simply determined by the number of dilutions you were prepared to try. It is useful if you are testing the samples with the same dilution level where a sample with very high levels of virus will still have virus particles when the same dilution of the other samples have none.
The minimum detection limit is a bit more problematic, since you may not have any viruses in the suspension from the start. If you tested an undiluted sample and saw no plaques, you can argue that the minimum detection limit would be 1 PFU - because had there been at least one virus present, you would have seen it (!).
The minimum detection limit is more useful if you are undertaking some sort of survey in which you are expecting some samples to have lots of virus and other to have none at all. This is a problem if you planned which dilutions to test based on the maximum number of virus particles you expect to see in some samples and you choose to leave out the no-dilution and low-dilution samples (e.g., because you were using a 96 multi-well plate). If you don't find viruses in the dilutions you checked but did not test the no-dilution sample, you won't be able to say there were no virus present in that sample – all you can say is that there is less than 1 in the lowest dilution you tested. If that was the 1/100 dilution, the minimum detectable becomes <100 (or ≤99).
Hope this helps!
Andrew
  • asked a question related to Assays
Question
4 answers
I have produced cellulase enzyme by solid state fermentation using rice straw as substrate and fungus Aspergillus flavus. I use CMCase assay method to determine enzyme activity. Now, how do i calculate enzyme activity in U/ml.
My assay method was 1% CMC as substrate in 0.05 M phosphate buffer. I used 1.8ml 1% CMC and 0.2ml Enzyme extract and incubated at 60°C for 30min. Then add 3ml of DNS and incubated at 100°C for 5min. and measured Optical density at 575nm. I made a standard curve. How do i calculate in U/ml from standard curve.
Relevant answer
Answer
Divide the rate of product formation in absorbance units per minute by the slope of the standard curve in absorbance units per micromole to get micromoles per minute.
Notice that the units are micromoles, not micromolar. You could use micromolar units instead, then multiple by the reaction volume in liters.
  • asked a question related to Assays
Question
2 answers
I have been considering the number of MGB probes I can use in my multiplex real-time PCR. The conventional advice suggests using up to two MGB probes, with the third probe being 'normal' - without MGB. However, in my experiments, I tested three sets of gene-specific primers and MGB probes, and surprisingly, it worked perfectly. I did not observe any decrease in efficiency compared to a singleplex assay. I'm curious if others have experience with using three MGB probes and if they observed any reduction in reaction efficiency.
Relevant answer
Answer
It depends on how any filters your instrument has and how well the deconvolution is programmed (the more filters the less different the detected wavelengths the larger possible crosstalk).
  • asked a question related to Assays
Question
1 answer
What is the suitable blank reading for ABTS assay at 734 nm by using 7.4mM ABTS solution mixed with 60 ml methanol ?
Relevant answer
Answer
Dear Ajay,
You can use methanol as a blank at 734 nm as you are using methanol as solvent system for the experiment.
  • asked a question related to Assays
Question
2 answers
Hello,
I am trying to decide a cut-off value (probably equally to "change-point") in an ELISA assay, using R package. To make our assay convinient, I do not set either negatve or positive control in every plate.
A reference paper (doi: 10.1590/0074-02760160119) indicates a package saying "the Pruned Exact Linear Time (PELT) algorithm was selected (Killick et al. 2012) with the CUSUM method as detection option (Page 1954). The PELT algorithm can therefore rapidly detect various change-points in a series. The CUSUM method
is based on cumulative sums and operates as follow: The absorbance values x are ordered in ascending values (x1,…xn) and sums (S) are computed sequentially as S0 = 0, Si+1 = max (0, Si + xi - Li), where Li is the likelihood function. When the value of S exceeds a threshold, a change-point has been detected."
I am not sure how to create such a code and could not find a package on the Internat. If someone knows or experienced the decision of "change-point" using R, please tell me hot to do that.
Thank you.
Relevant answer
Answer
example of how you can achieve this using the R programming language:
```R
# Install and load the required packages
install.packages("changepoint")
library(changepoint)
# Generate example ELISA assay data
# Replace with your actual ELISA assay data
data <- c(0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5)
# Perform change-point analysis
cpt <- cpt.mean(data, method = "BinSeg")
# Get the estimated change-point
change_point <- cpt@cpts[1]
# Print the estimated change-point
cat("Estimated Change-Point:", change_point, "\n")
```
`changepoint` package is used to perform change-point analysis on the ELISA assay data. First, you need to install the package using `install.packages("changepoint")`. Then, you can load the package with `library(changepoint)`.
Replace the `data` vector with your actual ELISA assay data. This vector should contain the measured values from your assay.
The `cpt.mean` function is used to perform the change-point analysis using the "BinSeg" method, which is a popular algorithm for detecting changepoints in data. You can explore other available methods in the `changepoint` package documentation.
The estimated change-point is extracted from the analysis result using `cpt@cpts[1]`, assuming there is a single change-point detected. If you expect multiple change-points, you can access them accordingly.
Hope it helps
  • asked a question related to Assays
Question
14 answers
It is a fact that enzymes affect by temperature. I assayed trypsin, papain and alpha amylase enzyme on insect crude extraction on different temperature I found that the increase of temperature (80 and 100 Celsius) make increase on the amount of these enzyme then repeat it again I obtained the same result how can I explain the results?
Relevant answer
Answer
Have you subtracted the absorbance signal from a blank reaction treated the same way, but without the enzyme, to correct for instability of the substrate at high temperature?
  • asked a question related to Assays
Question
2 answers
Alpha-Amylase Inhibitory Assay
This assay was carried out using a modified procedure of McCue and Shetty [17]. A total of 250 μL of extract (1.25–10 mg/mL) was placed in a tube and 250 μL of 0.02 M sodium phosphate buffer (pH 6.9) containing α-amylase solution (0.5 mg/mL) was added. This solution was preincubated at 25°C for 10 min, after which 250 μL of 1% starch solution in 0.02 M sodium phosphate buffer (pH 6.9) was added at timed intervals and then further incubated at 25°C for 10 min. The reaction was terminated by adding 500 μL of dinitrosalicylic acid (DNS) reagent. The tubes were then incubated in boiling water for 5 min and cooled to room temperature. The reaction mixture was diluted with 5 mL distilled water?? and the absorbance was measured at 540 nm using spectrophotometer.
( cre: Modes of Inhibition of α-Amylase and α-Glucosidase by Aqueous Extract of Morinda lucida Benth Leaf - PMC (nih.gov) )
Relevant answer
Answer
Could also be the volume of the cuvettes they used.
  • asked a question related to Assays
Question
1 answer
Hello Everyone,
I have one question about the concentration of LPC for ADA assay.
When we validation the confirmatory assay, we need the LPC concentration?
Thank you very much in advance!
QL
Relevant answer
Answer
Given antibodies will tend to bind in a 1:1 ratio and assuming 100% binding you would need the same concentration of drug as your antibody + the minimum concentration detectable by your assay. From that starting point do serial dilutions until you can no longer reliably detect your drug.
  • asked a question related to Assays
Question
1 answer
The binding of the sugar-dye compound with a lectin should facilitate a shift in the emitted light that enables monitoring of lectin activity. Or perhaps there are other ways to monitor lectin activity apart from a heamagglutination assay?
Relevant answer
Answer
Nicolai
If you have the desired structures of the sugar-dye compounds, you can search the are avalaible or they can be made just for you!
  • asked a question related to Assays
Question
2 answers
Hello, I would like to configure the spectrophotometer to 630 nm for a siderophore assay. The spectrophotometer we will be using is the Beckman Coulter DU730. Do you have any information about operating this machine?
Relevant answer
Answer
There is a user manual online
  • asked a question related to Assays
Question
1 answer
While performing a Fluorospot assay, generally there should not be a situation where non-specific spots are observed. However, after performing the experiment multiple times by multiple people in our lab we observe occurance of non-specific spots with the same cytokine kit. Where one could have numerous reasons why a spot occurs in wells with cells added to them, it is quite confusing to reason out why the spots appear in wells where there were no cells at all (Cell Control) but just the media. Therefore, for this particular cytokine kit both the Cell control and Stimulant Control with cells shows similar spots. Is there anyone who has experienced a similar problem and can explain what actually goes wrong here, what might be its probable reason and the measures one should take to avoid this?
Relevant answer
Answer
Would you mind sharing what kit you're using?
  • asked a question related to Assays
Question
4 answers
I am validating an ELISA measuring A1c (also called glycated hemoglobin; target for humans) for use in another species. It is a direct sandwich assay. The standard and my sample (whole blood) were both serially diluted twofold.
I am wondering if anybody can help diagnose what I am seeing in my parallelism graph. As you can see in my graph (attached), the curves are near inverse of each other (linearity R2 = -0.91). Since it is a direct assay, I would of course expect this correlation to be positive.
I have gone over my plate map and how I set up my dilution rack a hundred times in my head, and I am quite certain that I did load my plate in the correct sequence as my plate map - ie, having both standard and sample increase in concentration down their respective column. Is there any other possibility (besides me incorrectly loading my plate according to my map) that could cause this "perfectly incorrect" parallelism correlation?
I would love to be proved wrong and find out that it was indeed my error in loading - but I'm pretty sure I loaded it correctly...so I just wanted to check if there was any biological answer to this (matrix effect?)
I plan to run another plate tomorrow to check- but just wanted to ask.
Thank you!
Relevant answer
Answer
Robert Adolf Brinzer I will try that. Thank you!
  • asked a question related to Assays
Question
2 answers
I am trying to find out parasite burden of spleen in Leishmania donovani infected mice using limiting dilution assay. I have plated in serial dilution and in many replicates but now I am confused as to how I can analyze the data.
Can someone please help?
Relevant answer
Answer
I have tried 8 replicates of each of the 24 (1/5th) serial dilutions made from 1mg tissue/mL in 20% (FBS) Schneider medium. Initial infection in my animals was done with a low quantity of Leishmania donovani with only 1000 parasites and animals were dissected on the 6th week post-infection. Dilution culture was maintained until 21 days in continuous shaking at 22-24oC and yet no visual promastigotes were observed. only very tiny dot-like structures were observed with significant movements. However, the culture was not contaminated. I need to establish the limiting dilution method for a certain experiment and I don't know what else to do to make the transformation happen. Can someone please help?
  • asked a question related to Assays
Question
3 answers
I'm doing a targeted metabolite analysis using LCMS. I did a LCMS assay once using a set of serum samples processed for LCMS. However I wanted to repeat the LCMS run, using the same set of samples to optimize the protocol used for LCMS. The re-run was done using the initial processed samples which were stored in -80'C for about 2 months. The precision of the data of the repeated analysis was low and I didn't detect peaks for some of the metabolites which were detected in the initial run. Could you please let me know whether processed samples once used for LCMS couldn't be used again and if so, what is the reason for that?
Relevant answer
Answer
Periodic analysis of the sample can establish the stability of your sample. Most likely, your sample may have degraded.