Science topic

Enzymes - Science topic

Biological molecules that possess catalytic activity. They may occur naturally or be synthetically created. Enzymes are usually proteins, however CATALYTIC RNA and CATALYTIC DNA molecules have also been identified.
Questions related to Enzymes
  • asked a question related to Enzymes
Question
1 answer
Hello all,
I have a mini-prepped DNA that I am trying to digest with EagI. After doing the digestion it looks like the reaction with the restriction enzyme maybe linearizing my plasmid as it migrates at its expected weight when next to the DNA ladder. However the undigested sample runs very high, far above the 10kb band of the ladder. I am guessing this is the uncut is showing a nicked plasmid, which is why it's running so high? What do you think?
Thank you
MB
Relevant answer
Answer
dont worry what it is. do another digest that produces several typical fragments. when you see this patternon the gel the plasmid is definitely the one you are looking for.
  • asked a question related to Enzymes
Question
1 answer
i want to test the alpha amylase inhibitory activity of various pharmacuticals. is it possible to use alpha amylse from Aspergillus oryzae alpha (800EAU/Gram) to analyse in vitro alpha-amylase inhibitory activity using DNS Method. and what is the amount to enzyme (in ml) should i use?
Relevant answer
Answer
May 9, 2024
Dear Mukunda,
Yes, you should be able to use the A. oryzae enzyme to test for a-amylase inhibitory activity of your pharmaceutical agents. I would also suggest you try amylase from several other sources, e.g., porcine a-amylase, Bacillus, sp., a-amylase, plant amylases, etc. The protein structure of amylase from widely selparated biological sources will probably be different. The pharmaceutical agent may not inhibit A. oryzae amylase, but it might inhibit amylase from another biological source. That in itself would be an interesting observation, and perhaps give a clue about the mechanism of inhibition.
The last part of your question is confusing. Are you asking how much enzyme to use in the assays or what dosage of agent to use to inhibit the amylase? If you are asking how much enzyme to use in the assay, this you should be able to calculate from the potency of the enzyme, specifically the # of activity units per gram (or per ml for liquid enzyme). For the A. oryzae amylase this is specified (800 EAU/gram). You need to check to identify how the manufacturer defines a unit. DNS measures reducing sugar, so you want to use enough amylase to release an amount of reducing sugar that you can conveniently measure after a suitable reaction period (e.g., 10-15 min). You may have to determine this experimentally.
As far as how much of the pharmaceutical agent to use to inhibit the enzyme, this can be determined by adding increasing amounts of the agent to a fixed amount of the amylase. Again, some experimentation (trial and error) may be required.
I hope this information helps you.
Bill Colonna Dept. Food Science & Human Nutrition, Iowa State University, Ames, Iowa, USA [email protected]
  • asked a question related to Enzymes
Question
2 answers
The biocatalysts can be more available than traditional catalysts? The quation is related to the sustainablity by conversion of carbon using biocatalysts.
Relevant answer
Answer
Biocatalysts, which are typically enzymes derived from living organisms, have gained significant attention for their potential in catalyzing various chemical reactions. One area of interest is their ability to convert carbon sources into valuable products, such as biofuels, pharmaceuticals, and fine chemicals.
Compared to traditional chemical catalysts, biocatalysts offer several advantages:
1. Specificity: Enzymes often exhibit high specificity for their substrates, leading to fewer side reactions and higher product purity compared to chemical catalysts. This specificity can also lead to higher yields of the desired product.
2. Mild Reaction Conditions: Enzymes usually operate under mild conditions (e.g., ambient temperature and pressure, neutral pH), which can reduce energy consumption and minimize the need for costly equipment.
3. Renewable and Sustainable: Biocatalysts are derived from living organisms and can be produced from renewable resources, making them more environmentally friendly compared to chemical catalysts, which may rely on non-renewable resources and generate hazardous waste.
4. Compatibility with Aqueous Systems: Many biocatalysts are water-soluble and function well in aqueous environments, which simplifies reaction conditions and downstream processing.
5. Biodegradability: Enzymes are generally biodegradable, which can simplify purification and reduce environmental impact.
However, biocatalysts also have some limitations compared to traditional chemical catalysts:
1. Stability: Enzymes can be sensitive to temperature, pH, and other environmental factors, which may limit their stability and reusability in industrial processes.
2. Cost: Production and purification of enzymes can be expensive, especially for complex or low-yield reactions, which can impact the overall cost-effectiveness of biocatalytic processes.
3. Substrate Limitations: Enzymes may have specific substrate requirements, limiting their applicability to certain reactions or substrates.
4. Reaction Rate: While enzymes can exhibit high specificity, they may also have slower reaction rates compared to chemical catalysts, especially for large-scale industrial processes.
Overall, the efficiency of biocatalysts in converting carbon sources into valuable products depends on various factors, including the specific reaction, substrate, enzyme properties, and process conditions. In many cases, biocatalysis offers significant advantages in terms of selectivity, sustainability, and compatibility with aqueous systems, making it an attractive option for certain applications despite its limitations.
  • asked a question related to Enzymes
Question
1 answer
Dear Colleagues,
I working on evaluation of choline esterase inhibitors activity,the problem that I don't have the enzyme.
so i am trying to extract it from red blood cells of mice , rats ,rabbits or human .
can any one help me.
I'll appreciate every answer even if it is not suitable for me.
thank you in advance.
Relevant answer
Answer
Dear Jamal
I think this paper will be useful
Appl Biochem Biotechnol (2013) 170:198–209 DOI 10.1007/s12010-013-0177-3
Purification of acetylcholinesterase by 9-amino-1,2,3,4-tetrahydroacridine from human erythrocytes
Habibe Budak Kaya 1, Bilge Özcan, Melda Şişecioğlu, Hasan Ozdemir
  • asked a question related to Enzymes
Question
1 answer
I would like to understand the calculation of % enzyme inhibition
Relevant answer
Answer
One equation that can be used to plot a curve of % inhibition versus inhibitor concentration is the Hill equation. Assuming zero % inhibition in the absence of inhibitor,
% inhibition = MAX[Inh]n/(IC50n+[Inh]n) (Note: % inhibition is 100-% activity)
MAX is the maximal % inhibition, which is usually 100%, although it doesn't have to be.
IC50 is the concentration of inhibitor, [Inh], at which there is 50% inhibition.
n is the Hill coefficient of cooperativity. In the simplest case, it has the value 1.
It is not a good idea to try to extrapolate, especially since you have only 3% inhibition at 0.1 µM. What you need to do is measure the % inhibition at a range of concentrations of inhibitor, then use a nonlinear regression computer program to fit the % inhibition versus inhibitor concentration data set to the above equation (or an equivalent equation). From this you will obtain best-fit values for the parameters MAX, IC50 and n, and you will have a clear idea of the relationship between inhibitor concentration and extent of inhibition.
  • asked a question related to Enzymes
Question
2 answers
Say I have synthesized a new compound that gives great results against e. coli, how to know the best enzyme to target in e. coli? or how to predict the most possible target?
Relevant answer
Answer
If the question is how to figure out the target of the compound with antibacterial activity, a common approach is to select for resistance by gradually increasing the concentration of the drug to a culture, then sequencing the genomes of the parental and resistant strains to see what has changed. This does not always work, since the resistance may be due to changes in efflux pumps or other changes resulting in non-specific resistance, but it is worth trying.
Another method is bacterial cytological profiling. This method can classify the pathway in which an antibacterial compound acts by its effect on the way certain dyes stain the bacteria.
This assumes that there is a specific binding target of this compound. You should also consider the possibility that this compound inhibits the bacteria by a non-specific mechanism, such as membrane disruption.
I suggest you watch the bacteria under a microscope when you apply the compound to see whether there are any obvious morphological changes that would provide a clue to the mechanism of action. For example, inhibitors of some processes result in prevention of cell separation after growth, leading to longer-than-normal bacteria. Membrane blebbing would be an indication that the compound acts on the cell membrane.
  • asked a question related to Enzymes
Question
3 answers
I'm working with recombinant protein, it's a protease enzyme that we use E. coli to synthesize, and usually, it's stable for up to six months in the -80 freezer. Recently, the protein activity has been acting weird or showing a sudden drop after just two months, knowing that this enzyme is not expected to digest itself. Does anyone have an explanation, or even know how to solve this problem?
Thanks
Relevant answer
Answer
Hi,
I think that it is possible with the following reasons:
1, Protein degradation
The enzyme may not digest itself but the presence of proteases or/and contaminants in the E.coli lysate could degrade the protein over the time. As a consequence, the activity of the enzyme could decline.
In case of the presence of proteases, you could consider adding protease inhibitors during protein purification to prevent proteolytic degradation.
2, Thawing
The loss of protein activity as well as denaturation can occur if the protein is frequently freeze-thawed. I normally aliquot the protein into many tubes with smaller volume before freezing.
3, Variability between batches
It is possible that protein expression after purification may be variable between batches, as a result, protein activity will be different over the time.
You can minimize the variability by maintaining consistency in protein purification procedures.
4, Aggregation of protein
Protein activity can be influenced by its aggregation occurring over the time because of several factors such as pH levels, salt-buffed solutions, and high protein concentrations. To minimize this, you can maintain the protein at low concentrations, use appropriate buffers, or introduce chaperones to prevent protein misfolding and aggregation.
I hope this information is useful for you.
Good luck!
  • asked a question related to Enzymes
Question
1 answer
I did as follow. I mix 1ml of starch with 1ml of crude enzyme. incubated for 15 minutes. After that I added 1ml of DNSA and bioled for 10 moinutes. after boiling i added 5ml of distiled water. thes measured absorbance at 540nm. my maltose standard curve equation is y=0.0009x + 0.01. how can I calculate Enzyme activity?
Relevant answer
Answer
Hi there,
The first issue would be to get a much more accurate equation for you standard curve. The slope has only one significant number (what are at least the 2 first numbers after the 9 ?)... Then reporting your readings onto the curve would give you the corresponding maltose amount/concentration produced in 15 minutes per mL of crude enzyme.
  • asked a question related to Enzymes
Question
1 answer
I am trying to express a FAD containing enzyme of mycobacteria in E.coli. I am able to purify the protein which is slightly yellow in color but it seems that my FAD is all unbound to the protein. How can I express the protein which has tightly bound FAD?
Relevant answer
Answer
You can try simply adding FAD back to the purified enzyme. It is commercially available.
  • asked a question related to Enzymes
Question
4 answers
Dear Research Community,
I am encountering a significant hurdle in my research involving enzyme inhibition testing. The inhibitor I am investigating exhibits solubility exclusively in DMSO, rendering it insoluble in aqueous environments such as the 100mM phosphate buffer I am utilizing for enzyme kinetics studies. Upon attempting to incorporate the DMSO-dissolved inhibitor into the reaction mix, it precipitates out, leading to haze formation in the solution and hindering accurate data collection.
I am seeking insights and suggestions on how to effectively address this challenge. Specifically, I am interested in methodologies , or alternative solvents that could facilitate the integration of the inhibitor into the reaction mix without inducing precipitation. Additionally, any advice on modifying experimental conditions or buffer compositions to mitigate this issue would be greatly appreciated.
Thank you in advance for your expertise and assistance.
Relevant answer
Answer
In addition, the suggestion of Adam Shapiro, to lower the phosphate buffer concentration (e.g. from 100 mM to 50 mM ) can be helpful. So, revise your assay conditions.
  • asked a question related to Enzymes
Question
3 answers
I need to develop an enzyme assay to evaluate the degradation of gluten using proteases. Previously, I conducted tests with finished products, which was straightforward. However, I now have to work with actual enzymes, which are in limited quantity. Consequently, I'm facing challenges in devising an assay method. The total enzyme quantity available is 100ml, and I need to conduct multiple tests to assess the enzyme's activity in degrading gluten. Could you please provide assistance in formulating an appropriate assay method for this task?
Relevant answer
Answer
You have been working on a scale that I consider to be enormous. I'm used to micro-scale assays of enzyme activity, conducted in volumes on the order of 50 microliters. Also, the gluten and enzyme concentrations you use seem enormous to me at 1 g/mL and 0.4 g/mL, respectively.
You haven't described the method of measuring gluten hydrolysis. The products will be smaller peptides. A straightforward way to follow the reaction would be to run the sample through an HPLC column that separates peptides on the bases of either size or hydrophobicity (size exclusion or reverse phase, respectively). The samples would probably have to be diluted and centrifuged prior to analysis, and you would need a blank of enzyme without gluten to identify which peaks were from the enzyme. Detection would be by UV absorbance at 214 nm.
I found a method in this paper
in which the hydrolysis of gluten was followed by the creation of new N-termini using the o-phthaldialdehyde (OPA) reaction, with detection at 340 nm with a spectrophotometer or absorbance plate reader. No chromatography would be needed, which would speed up the experimental testing of different conditions.
Both of these ideas could be performed in very small volumes, such as in 1.5-ml polypropylene microcentrifuge tubes containing no more than a few hundred microliters of solution. This would also simplify the centrifugation step, since the samples would already be in centrifuge tubes. The tubes could be incubated at various temperatures in temp-blocks designed to hold that kind of tube. The supernatants would be transferred to separate containers and either mixed with OPA for detection, or analyzed by chromatography.
  • asked a question related to Enzymes
Question
4 answers
This is supposed to be extremely efficient, but somehow, I've never gotten it to work.  I ordered completely new enzymes because I thought my enzymes may be thawing (I re-ordered BsmBI, BlgIII, and Sal1HF), and my negative control had a greatly decreased number of colonies compared to my guide+plasmid plates for the first time; hence, I thought my enzymes were the problem.  However, upon sequencing, I still got empty vector.  Should the next step be to order new ligase? I'm not sure what's going wrong!
Relevant answer
Answer
Dear [Recipient],
I hope this message finds you well. It's clear that you are encountering challenges with your CRISPR/Cas9 cloning protocol, specifically in the insertion of your guide RNA (gRNA) sequence into the vector. This step is crucial for the successful application of CRISPR/Cas9 technology, and encountering difficulties can be frustrating. Below, I outline a series of considerations and troubleshooting tips to assist you in resolving this issue.
Assessing the Cloning Strategy:
  1. Vector and Insert Compatibility:Ensure that the vector and insert have compatible ends for ligation. This could be cohesive ends generated by restriction enzymes or blunt ends for blunt-end cloning. Verify that the enzymes used do not cut within your insert sequence.
  2. Restriction Enzyme Efficiency:Confirm the complete digestion of the vector by the restriction enzymes. Incomplete digestion can lead to low cloning efficiency. It might be helpful to use a dephosphorylation step to prevent vector self-ligation.
  3. Insert Preparation:Verify the purity and concentration of the insert. Contaminants from PCR amplification or previous cloning steps can inhibit ligation. Consider running a gel extraction to purify the insert.
Ligation and Transformation Efficiency:
  1. Ligation Conditions:Optimize the molar ratio of vector to insert. A common starting point is a 1:3 vector-to-insert ratio, but this can be adjusted depending on the sizes of the vector and insert and the specifics of your ligation kit. Ensure that the ligation reaction is incubated under optimal conditions recommended by the enzyme supplier.
  2. Competent Cells:The efficiency of transformation can greatly affect cloning success. Use high-efficiency competent cells and ensure they are properly thawed and kept on ice before transformation.
  3. Recovery Phase:After transformation, a proper recovery phase in rich medium allows for the expression of antibiotic resistance genes before plating on selective media.
Verification of Cloning Success:
  1. Colony PCR:Screen transformants by colony PCR to quickly identify colonies that may contain the insert. Design primers that anneal to vector sequences flanking the cloning site.
  2. Restriction Analysis:Perform a restriction digest of plasmid DNA from positive colonies to confirm the presence and orientation of the insert.
  3. Sequencing:Sanger sequencing of plasmid DNA from potential positive clones can confirm the correct insertion and sequence integrity of the insert.
Additional Considerations:
  • Gel Purification: If your insert and vector are of similar sizes and difficult to separate by gel electrophoresis, consider using an alternative method for purification or a different strategy for cloning.
  • Alternative Cloning Methods: If traditional cloning continues to fail, consider using a site-specific recombination system (e.g., Gateway cloning) or a seamless cloning kit (e.g., Gibson Assembly or Golden Gate cloning) that might offer more flexibility and efficiency.
Conclusion:
Troubleshooting cloning protocols requires patience and systematic optimization of each step. By carefully reviewing your protocol and considering each of the points mentioned above, you can identify and rectify the issue hindering your cloning success.
Should you require further assistance or have specific questions at any step of your protocol, please do not hesitate to reach out. I am here to support you in advancing your research projects.
Best regards,
With this protocol list, we might find more ways to solve this problem.
  • asked a question related to Enzymes
Question
8 answers
hi everyone,
i am trying to figure out why the gene inserted is not amplifying after cloning. I have inserted a mycobacterium-specific gene into pET 32a plasmid and transformed the ligation product with BL21 cells. after transformation, I have isolated the plasmid from the obtained clones and done restriction digestion with the desired restriction enzyme. i have got a positive result for this experiment by running the restriction digestion product in 1% agarose gel. hence i kind of confirmed that the cloning has worked. ionrder to re confirm it, i have done a colony PCR with the obtained transformant colony.but i have got no amplicon for the gene. i have also tried to amplify the gene from the isolated plasmid using the gene-specific primers,but that also gave a negative result. i have repeated these experiments for multiple time but each time am getting the same pattern of result. that is a positive result for restriction digestion of the isolated plasmid and negative result for the colony PCR as well as the plasmid PCR. what can be the possible reason behind this. also i tried to express the protein using iptg induction, that also resulted in negative result.
Relevant answer
Answer
The PCR did not work (on the plasmid and on the colony), the expression also did not give any protein.
Potentially, the DNA fragment you cloned is not your target gene, but another DNA fragment you cloned.
If you want to confirm your result 100%, you would need to carry out sequencing.
  • asked a question related to Enzymes
Question
9 answers
An enzyme protein overexpressed in bacteria needs to be purified to show its activity. Why doesn't the cell lysate show activity when added to the substrate?
Relevant answer
Answer
In general, a cell extract is a mess... It may contain inhibitors of your enzyme, or other enzymes that consume the substrate and prevent detection of your enzyme's activity. Artifacts may also arise depending on the type of reaction you're trying to monitor and the assay you're using. If, say, your enzyme generates hydrogen peroxide you may not be able to detect this compound because some substances (thiols) in the cell extract readily react with it. All these problems, you don't have them with a purified enzyme. As the late Arthur Kornberg used to say "Never waste clean thinking on a dirty enzyme".
  • asked a question related to Enzymes
Question
1 answer
Hello everyone,
I am interested to know how I have to calculate the concentration of a specific analyte during the digestion steps (salivary, gastric, intestinal...).
During the method, the sample is dissolved by half between steps, for example: salivary (50%); in gastric step, the salivary step is dissolved by half with the gastric enzyme mix, so the sample is dissolved at 25%... etc etc.
When I obtain my quantification data after LC/MS analysis, how should I express my results? Do I have to take into account the dilution factor for each step? (Saliva x2, Gastric x4 ....)
Kind regards,
José Ángel
Relevant answer
Answer
Yes, you should account for each dilution step when expressing the concentration of analyte in the original source.
  • asked a question related to Enzymes
Question
3 answers
Hello,
I am using Trypsin to determine its Km and Vmax values against casein from bovine milk. I know higher enzyme concentrations increase the Vmax value, but does more trypsin also affect the Km value?
Thank you in advance
Jordan
Relevant answer
Answer
No, enzyme concentration does not influence Km, as long as the measurements are done under proper conditions for Michaelis-Menten kinetic analysis. That means that the enzyme concentration is far below the substrate concentration and the initial rate is measured.
If the enzyme concentration is too high, it becomes difficult to measure the initial rate because the reaction goes too fast. If the enzyme concentration is not much lower than the substrate concentration, the free substrate concentration may be significantly reduced due some of it binding to the enzyme, so the free substrate concentration does not match the total substrate concentration.
  • asked a question related to Enzymes
Question
3 answers
I need help analyzing enzyme kinetic data.
I have data from the Octet K2 system. In my experiment, I load the sensor with our protein of interest (6XHis tag on my recombinant protein to Ni-NTA sensors) and then expose this sensor to increasing concentrations of the candidate binding protein (five concentrations per experiment and each experiment is replicated four times). Each association step is followed by a dissociation step in buffer. A control sensor is used in each experiment where a sensor is loaded with the protein of interest but only exposed to buffer. (See picture, Part 1)
I have separate data where I loaded smaller recombinant domains of the protein of interest to the sensor and exposed it to the candidate binding protein. I would like to combine this data (the binding of the full-length protein and the binding of the domains) on the same graph.
My problem: In trying to analyze the data with the software provided with the Octet system (HT 11.1), the data misaligns. (See picture, Part 2)
My goal is to determine kinetic constants (KD) of the full-length protein and its separate domains to the protein of interest.
Suggestions for correctly aligning the data in the Octet software HT11.1? (I think the misalignment is because the program is trying to align the y axis to baseline 1 instead of baseline 2, which is the baseline right before the association step. If so, can you change this label after the fact?)
If the glitch with the Octet software cannot be fixed, then is there a manual/tutorial for the enzyme kinetic module for Sigma Plot?
I found I can extract the raw data from the Octet system. I can remove the background from the control sensor and manually assign concentrations. I uploaded this into Sigma plot 15, which has an enzyme kinetic module. I found the embedded help guide, but I have specific questions. For example:
*My candidate binding protein does not change, but how do you take into account the change in the kilodaltons of the proteins that are loaded to the sensor, full length vs. the smaller domain proteins? This is automatically taken care of in the Octet software.
*How do I differentiate between the association and dissociation phases?
I am new to Octet biolayer analysis and the Enzyme Kinetic Module analysis in Sigma Plot.
Any help will be greatly appreciated! I am happy to provide any more information.
Relevant answer
Answer
Awaiting for the analysis results' success 😊
  • asked a question related to Enzymes
Question
6 answers
I am working on an enzyme and checking its stability in different solvents. One of the solvents is DMSO. I am interested in checking how much active or correctly folded protein is still in the reaction mixture after exposure to DMSO or any other solvents after a certain amount of time.
Relevant answer
Answer
As pointed out by Jeremy Pronchik , it is not necessarily true that a given protein molecule is either fully active and in its native conformation or fully inactive and unfolded. A protein molecule's structure might be mildly perturbed by a low concentration of DMSO and retain partial activity. High solvent concentrations, on the other hand, would probably result in major loss of native structure of most proteins.
If a solution of a protein at a reasonably high protein concentration is substantially unfolded, it will almost certainly result in aggregation of protein molecules due to exposure of the hydrophobic interior. In this extreme case, a simple measurement by dynamic light scattering will reveal the presence of aggregated protein. This requires a special instrument, but if the aggregation is severe enough, it can even be detected with a UV-VIS spectrophotometer, which most labs have.
The effect of a solvent or other chaotrope on the stability of a protein can be monitored by measuring the melting temperature of the protein in solution, using either differential scanning calorimetry, or differential scanning fluorescence. Each measurement requires a special instrument.
Many proteins contain tryptophan residues whose fluorescence is sensitive to their environment, specifically the hydrophobicity of their environment. Using a spectrofluorimeter, you can measure the tryptophan emission spectrum to follow major and minor structural changes in the protein. This can also be done using far UV circular dichroism (CD).
Far UV CD, mentioned by Jeremy Pronchik is used to measure changes in the secondary structure elements of the protein. Loss of secondary structure would be a sign of a major loss of native structure. I think the presence of DMSO would interfere with this measurement, however.
  • asked a question related to Enzymes
Question
2 answers
I've selected protease as the optimal enzyme for eliminating gluten formed from flour. Could you please provide insights on the best enzyme for removing gluten, dosing methods, and how to identify the suitable enzyme variant given that proteases have diverse types? This information is essential for my project aimed at resolving pipeline blockages induced by gluten from flour in the food industry using enzymes.
Relevant answer
Answer
Gluten is responsible for elasticity, thus breaking down gluten will change the texture/flavor. Broken amino acids can also change the chemistry of the fermentation, and thus may affect the taste as well. Most people would go for enzyme supplements as an aid in the digestion of gluten. However such ideas have been explored in the past and if you can overcome such hurdles, it can be a good project.
Have a look at these papers
  • asked a question related to Enzymes
Question
6 answers
I'm trying to establish the Km for different substrates for a specific enzyme, however, at every attempt a new doubt arises. This is a new field of knowledge to me, so any suggestion will be helpful.
So far I am following these premisses:
- at least 6 substrate concentrations, sometimes 7 (from 1 mM to ~ 0.015 mM) ;
- the substrate consumption should be less than 10% ( so I am testing several amounts of enzyme for each substrate and using the amount that is enough to be measured and also consumes less than 10% of the substrate, even in the lowest concentrations);
- Results of the Km and Vmax are obtained by nonlinear regression of the Michaelis-Menten curve using GraphPad Prism software.
The analysis is performed by the formation of the phosphate in the reaction (molybdate/malachite green-based assay).
Some of the problems I'm facing:
1) for some substrates I couldn't get an M-M-like curve, even when trying different amounts of enzyme or substrate. Then, I believe that the data are not adequate to perform Km and Vmax calculations. I've noticed that with these substrates the consumption is lower than than with the other ones. Any suggestion on how to improve the assay in this case?
2) for the substrates that I do get a proper M-M-like curve, many times I am not able to obtain good independent replicates. Once the amount of enzyme is not supposed to affect the Km, but only the Vmax, what else could be affecting changes in Km?
3) Also, I've noticed that for some substrates (even if the replicates are consistent) if I consider 7 or 6 different substrates concentrations the Km changes significantly. For example: using substrate from 1 mM to ~ 0.015 mM (7 concentrations) it was obtained a Km of ~ 0.30 mM; while considering just substrate from 0.5 mM to ~ 0.015 mM (6 concentrations) the Km obtained was ~0.17 mM. Someone could explain that or indicate some bibliography?
Thank you in advance!
Relevant answer
Answer
If you could send me your measurements, I could try with my software.
  • asked a question related to Enzymes
Question
3 answers
I tried to isolate pr-FMN from UbiD like enzyme and verify it via UPLC and Mass spectrometry. The results obtained from MS shows detection of right mass but UPLC spectrum tell another story. How can I identify compound just with mass if it’s not prFMN?
your guidance will be highly appreciated.
Relevant answer
Answer
UPLC spectrum?... Did you mean UPLC chromatogram or LC-UV spectrum/spectral view?
If you mean chromatogram and resulting different retention times with the same masses, these may be the isomeric variants of the same molecules. This can be clarified by the use of a high-resolution power of the mass spec. High-resolution m/z values (say 40.000 and above FWHMs) can confirm if these are probably the isomeric species.
If you are meaning UV spectral shift differences (distinct UV max absorption wavelengths) but the same masses, probably they are different molecules. You can use the abovementioned second option if you did not apply before.
Alternatively MS/MS and MS to the n experiments would be more beneficial to discriminate the molecules. EAD, CID, HCD, UVPD, and ETD are the orthogonal approaches that can also be used for molecular structure elucidation.
Good luck,
İEA
  • asked a question related to Enzymes
Question
2 answers
I work in a investigation about hydrolysis of protein of a andean grain and is very important for continue with my work what method is more effective for the activation or maybe is possible not to do it?
Relevant answer
Answer
Reducing conditions are sometimes necessary to activate/reactivate cysteine proteases. This way you may get a higher activity.
  • asked a question related to Enzymes
Question
2 answers
  • In the enzyme reaction with PFAS, how to stop the reaction after a certain time? (I plan to add 1 mL 0.5% methanolic ammonium hydroxide in 0.5 mL reaction solution, will it work?)
  • Then I want to dissociate the enzyme from PFAS, and remove the enzyme because I need to measure the PFAS. How to do that? (I plan to freeze (-20 ℃) the sample overnight and then centrifuge it (at 4 ℃) to get the supernatant, will it work?)
Relevant answer
Answer
Add the organic solvent at a volume ratio of 1:4, this time the enzyme will denature, and then centrifuge at high speed to take the supernatant, this is how the serum sample was treated in metabolomics experiments :-)
  • asked a question related to Enzymes
Question
2 answers
As a classical biocatalyst, what is the normal range of the catalytic constant (Kcat) value of the typical enzymes? What does it mean if it's less than 1/s?
Relevant answer
Answer
What's the point of an enzyme-catalyzed reaction being only three times faster than an enzyme-free one?
  • asked a question related to Enzymes
Question
1 answer
I will extract and purify elastase from the pancreatic tissues, and then calculate the optimum pH, temperature, and substrate concentration. After, I will apply different plant extracts to the enzyme to identify the inhibition effect.
my question is:
1. what is the chemical inhibitor of serine elastase?
2. what is the perfect method for extraction?
3. what is the substrate
4. what is the better method to add the plant extracts ?
Relevant answer
Answer
Serine proteases are often inhibited by phenylmethylsulfonyl fluoride (PMSF) and 3,4-dichloroisocoumarin.
Chromogenic and fluorogenic artificial substrates for neutrophil elastase are available, consisting of short peptides with a C-terminal nitroaniline or aminomethylcoumarin group. An example is:
Methoxysuccinyl-Ala-Ala-Pro-Val-p-nitroanilide
I don't know whether these will work with a plant enzyme.
Adding plant extracts may be tricky if you are measuring a color or fluorescence change because the extracts may interfere with detection. In that case, you may want to use a protein substrate and follow the cleavage by SDS-PAGE.
  • asked a question related to Enzymes
Question
3 answers
For enzyme assays using cell supernatant which are slight yellow and using pnpp substrate which will also produce the yellow pnp substrate will predictably have a distortion on the absorbance. to what extent will this affect enzyme rate calculation and any papers on this?
Relevant answer
Answer
You can subtract a blank, as already stated, but it isn't really necessary as long as you are sure that the background absorbance due to (1) the supernatant without pNPP and (2) pNPP without the supernatant does not change over the time course of the full reaction. That is because you only need to know the rate of change of the absorbance caused by the enzyme-catalyzed reaction to measure the activity.
  • asked a question related to Enzymes
Question
10 answers
Hello everyone,
I need a citrate buffer with an optimal pH of 4.8 for the enzymes I'm working with. I'm using citric acid monohydrate (molecular weight: 210.14 g/mol) and adjusting the pH with NaOH. I'm preparing it as a 10x concentration and diluting it to 1x in the final volume (15-200 µl).
However, I've come across recipes for citrate buffer that use both sodium citrate and citric acid.
My question is whether the buffer I'm making will be strong enough to maintain a pH of 4.8 when I dilute it to 1x in my sample. Is the recipe with sodium citrate and citric acid a better option for buffering at pH 4.8?
Relevant answer
Answer
We used sodium citrate and citric acid to prepare citrate buffer . There is a Table for buffer preparations in Methods in Enzymology Vol 1 .
  • asked a question related to Enzymes
Question
6 answers
We are using some enzymes to test in the laboratory. Trying to figure out the best method treat the waste water. The actual chemical we using are testing with enzyme based drain cleaners. Need guidance and suggestions..
Thank you.
  • asked a question related to Enzymes
Question
1 answer
I'm working in a project where I need to test the enzyme activities of an insect's gut fluid. I've already tested it and found some good activity. Now I want to know whether the enzyme, showing high activity, is endogenous to the insect, or just produced by gut microbiota. There is no genome sequence or any other information available about that insect. How can I test it? How can I prepare enzyme solution, containing only endogenous enzyme of the insect?
I've read about the antibiotic feeding process. Anything else?
I found a paper where they incubated the insects for 24 hours, dissected, collected the gut fluid and then filtrated the gut fluid through 0.22 micrometer syringe filter, then demanded that this filtered solution contain no bacteria, neither their enzymes. But how can they demand there is no bacterial enzyme? If it is for starvation, then isn't it obvious for the insect's enzyme to reduce too? Can anyone provide any reference on support of this method, or concept? Please let me know.
I'm struggling in this issue and can't find any solution. I can't use the antibiotic method for a reason. Please help me out. Thank you.
Relevant answer
Answer
If you can't kill off the bacteria in the insect gut by the use of antibiotics, can you somehow raise the insects in such a way that they do not have any gut bacteria, the way it is done with mice? This would involve first eliminating the gut bacteria of some parental insects with a combination of antibiotics, maintaining them in a sterile environment with sterile food, then breeding those insects to produce offspring that could be studied without antibiotic treatment. I don't know if anyone has ever done this.
Even if you had the entire genome sequence of the aseptic insect, it would not necessarily help you with your experiment if you are measuring the enzyme activity level with an enzyme assay, since that does not necessarily uniquely identify the enzyme(s) responsible for the activity. You would have to do a proteomics study to see what proteins were present in the gut fluid. You might be able to identify the enzymes that way by comparison with genomic sequences of related insects. You might also consider obtaining the full genome (or at least the full exome) sequence of your insect to inform your proteomics work. It's not that expensive nowadays.
You should also consider that there might be an interaction between the insect's gut lining cells and the gut microbiome that could affect the insect's secretion of enzymes. In other words, the insect and its microbiome may not be independent, so you would get a different amount and/or set of enzymes secreted by aseptic insects compared to insets with a normal microbiome.
  • asked a question related to Enzymes
Question
1 answer
What is the particle size of the cross-linked enzyme aggregates? Can the prepared cross-linked enzyme aggregates be dissolved in water again?
Relevant answer
Answer
Particle size distributions of cross-linked enzymes can be measured by a variety of techniques, including SDS-PAGE, size exclusion chromatography (SEC), SEC coupled with multiangle light scattering (SEC-MALS), analytical ultracentrifugation techniques, dynamic light scattering and other light scattering techniques. The choice of technique will be influenced by the size of the particles and equipment availability.
Covalent crosslinking is usually not reversible. However, there are reversible crosslinking agents, such as those containing disulfide bonds that can be split by reducing agents.
  • asked a question related to Enzymes
Question
1 answer
functionalized mesoporous silica unable to immobilise alpha amalyse enzyme
Relevant answer
Answer
Dear B.s. Bini ! Are the pores in the silica gel large enough? If the enzyme and substrate molecules are placed in pairs, then you can safely proceed to immobilization.
There are several ways to immobilize amylase on silica gel. To do this, it is necessary to fix the amylase with chemical bonds. Covalent bonds and ionic bonds are suitable. Ionic bonds are more convenient. Check out my post.
  • asked a question related to Enzymes
Question
1 answer
Hello,
I want to use a commercial preparation of recombinant PNGase, but I do not have the buffer. What cofactors or buffer conditions does PNGase F require? I will add the enzyme to cell lysate and then run western.
Thanks!
Relevant answer
Answer
Here is the protocol from New England Biolabs.
file:///Users/adam/Downloads/PNGase%20F,%209PIV483.pdf
  • asked a question related to Enzymes
Question
2 answers
I completed the experiment for one of our colaborators (I measured inhibition of an enzyme by 3 compounds). Unfortunatelly compounds were not active in the range of dilutions that I had used (up to 10 micromol). Higher concentrations caused the paradoxically high signal due to (probably) interactions with the detection reagent. Therefore concentrations higher than 10 micromol were excluded and, in the end, I could not observe the full dose-dependent curves (and did not calculate IC50). If it was my publication I would left the results like this with the explanation why higher concentrations were not included. But the colaborator asked If I could "complete the curve" on the graph for one of the compounds. I would rather not do that but I consulted one of my supervisors and added dotted line in place where could be the curve (the highest concentration of compound reached ~ 50% of the maximal inhibition, rest of the curve was predicted by the program). I though, that the request was made just to make graph more pleasant to the eye. But later the same collaborator made another request - to calculate IC50 from that curve (where half was from the experiment and half was "predicted" by the program with the assumption that the compound will reach 100% of inhibition - which is not certain). The collaborator also asked to "complete" another curve (where experimental data reached only 25% of the maximal inhibition).
If it was my publication I would never do that. I do not want to suggest that I tested concentrations that I did not test and I do not want to suggest that the compounds were able to fully inhibit enzyme (which I did not prove). Also I do not want to create "artificial IC50" which could be used to compare the compound with different compounds creating false conclusions. But maybe I am wrong? Therefeore I want to ask:
Is it correct practice to "complete the curve" (to elongate the curve to reach 100% of expected effect) even if you did not test higher concentrations (because higher concentrations were to high for the assay). Is it correct to calculate IC50 from that kind of curve?
Relevant answer
Answer
Robert Adolf Brinzer is right. 15-85 is the part of the sigmoid curve that most resembles a straight line. We can't quantitate reliably outside that region of a calibration curve.
Although we may "predict" results, we must never fabricate data. You might try to publish a curve with points indicating your data and dotted lines suggesting how you think the system may behave at other concentrations. You might get that published in a predatory journal.
  • asked a question related to Enzymes
Question
2 answers
Hello everyone,
I need to use a final concentration of 90ng/uL of BSU polymerase in a final volume of 50uL. The NEB BSU polymerase specification sheet indicates 5ng/uL as the stock concentration. How do I convert units/uL to ng/uL knowing that one unit is defined as the amount of enzyme that incorporates 10 nmol of dNTP into acid insoluble material in 30 minutes at 37°C.
I am not 100% sure but I know that:
Specific activity (units/ng) = 5 U/μl / 10 nmol
To convert nmol to ng, we need to know the molecular weight of dNTP. Assuming an average molecular weight of 330 g/mol for a single nucleotide, we can calculate the specific activity in units/ng as follows:
Specific activity (units/ng) = 5 U/μl / (10 nmol x 330 g/mol)
and... if this is correct... then what?
Thanks a lot :)
Relevant answer
Answer
I believe you meant to write that the stock concentration is 5 U/µL. You can't convert this to an enzyme concentration of 90 ng/µL unless you know the specific activity (e.g., in U/mg) of the stock solution. 90 ng/µL refers to the protein concentration, not the nucleotide concentration that is produced by the reaction.
The specific activity may be written on the label or in the product information sheet or certificate analysis for the particular lot of the product that you received. This information may be available on-line from NEB. Once you know the specific activity, the conversion is simple.
For example, suppose the specific activity is 1,000 U/mg.
You need 90 ng/µL = 90 x 10-6 mg/µL
90 x 10-6 mg/µL x 1,000 U/mg = 0.09 U/µL
Since the stock solution is 5 U/µL you must dilute by a factor of 5/0.09 = 55.6-fold into the reaction.
Or, you could do it this way, using the same example specific activity of 1000 U/mg:
(5 U/µL) / (1000 U/mg) = 0.005 mg/µL = 5 µg/µL = 5000 ng/µL
The required dilution factor is 5000/90 = 55.6
  • asked a question related to Enzymes
Question
1 answer
I am conducting an on-going research study and I am evaluating the performance of consortium of bacteria as a biocatalyst in a microbial fuel cell. There's some misunderstanding between me and the panelist about what to call the subject the microbial fuel cell that we are assessing. Prior to our consultation, we called the MFC's as our participants. We were called out about this because according to the panelist, we cannot call the microbial fuel cell as participants because they are inanimate objects, thus, won't be able to answer the questionnaire in the pre-test and post test.
If that's the case, what is the right term to call the microbial fuel cell? And if it's not a questionnaire, what's the type of question should we ask to evaluate the performance of the MFC's?
Relevant answer
Answer
The microbial fuel cell in its entirety might be called the test apparatus. The consortium of bacteria should just be called "the consortium of bacteria." There should be one or more objective, numerical measurements of performance of the apparatus and its bacterial contents. There is no need for a survey.
  • asked a question related to Enzymes
Question
1 answer
Crosslinking on surfaces makes the enzyme reusable multiple times, I wish to know whether this is possible for a chemical reagent. Once fixed or coated on a surface, can the reagent be used multiple times for a microassay? And if this can be done, please direct me to a source. A research paper/any kind of literature would be helpful.
Relevant answer
Answer
Industrial chemistry often uses immobilized enzymes for synthesis. See https://en.wikipedia.org/wiki/Immobilized_enzyme.
  • asked a question related to Enzymes
Question
2 answers
Hi,
I have Michaelis-Menten curves for 2 conditions (control VS disease) to look at the difference in activity of my enzyme of interest. I'd like to see the best way to quantify the difference in activity statistically. I have attached my curves. The blue + purple curves are biological replicates of control and the green + red curves are disease samples. I'd like to quantify the change in the enzyme activity.
Any help would be appreciated!
Relevant answer
Answer
If you use a nonlinear regression program to fit the data for each curve to the M-M equation, it will give you best-fit values of Km and Vmax. It will also give you values for the standard error of the best-fit value of each fitted parameter. You should be able to use a statistical test of some sort (e.g. t-test, but I'm not sure about the best way) to compare the values of each parameter between pairs of curves to see if any differences are statistically significant.
However, I think a better way would be to prepare 3 independent M-M plots for each condition, and use nonlinear regression to fit each plot individually to obtain 3 Kms and 3 Vmaxs for each condition. Then you will have 3 independent measurements of each parameter, allowing you to calculate averages and standard deviation. Then you can do t-tests on pairs averages to test for statistical significance of the differences.
  • asked a question related to Enzymes
Question
3 answers
I have just started my Phd , I have to work on enzyme engineering and my previos background was quite different, for my research have studied literature and selected an enzyme santalene synthase for engineering this enzyme has the PDB structure available but when I blast the same sequence on NCBI results are showing synthetic construct with its name . what does it mean and can I use this enzyme for my research ? Your kind guidance will be appreciated. Thanks
Relevant answer
Answer
Hello Amna,
I don't have a concrete answer for you, but, I have the same problem with PETase and MHETase of Ideonella sakaiensis. I would assume the reason for #1 is just an error in the nucleotide blast database or search algorithm. I would not worry about it; there probably is an uninteresting reason (syntax, or something funny with the parameters) that it doesn't match.
In my case, 16SrRNA data confirms it is the correct bacterium.
  • asked a question related to Enzymes
Question
4 answers
Greetings
I am working with a plasmid encoding the restriction enzyme EcoRV.
Do you know of E. coli strain resistance to EcoRV restriction?
Relevant answer
Answer
It has certainly been cloned and lots of studies done on it. My suggestion would be to see if someone like New England Biolab could share it (they have always provided strains and clones freely) or in this day and age it might be easiest to just have it synthesized into a plasmid backbone appropriate for your needs.
  • asked a question related to Enzymes
Question
8 answers
I am trying to carryout diagnostic digest with AatII and DraIII (Adei) but the two did not cut as expected. Could it be because of CpG sensitivity?
The tests are the paired lanes separated by blank from left to right. The first lane on the left is treated with enzyme (Bam HI, AatII, Xbal, Dra III). The second lane contains the uncut plasmid in each case.
These are not PcR products, they are plasmids from miniprep.
Relevant answer
Answer
If a restriction enzyme is CpG methylation sensitive, you need to consider the following:
  1. Methylation Status: Check the methylation status of the DNA you want to digest. If it's highly methylated at the enzyme's recognition site, it may not work effectively.
  2. Digestion Conditions: Optimize digestion conditions, including enzyme concentration, buffer composition, and reaction temperature, as CpG methylation sensitivity can vary between enzymes.
  3. Use Alternative Enzymes: Consider using alternative restriction enzymes that are not CpG methylation sensitive or using a combination of enzymes to achieve the desired DNA cleavage.
  4. Enzyme Inhibition: Some enzymes can be inhibited by CpG methylation. In such cases, you may need to perform a demethylation step before digestion.
  5. Bisulfite Treatment: If your goal is to study DNA methylation, consider bisulfite treatment to convert methylated cytosines to uracils, allowing subsequent analysis without restriction enzyme issues.
  6. Consult the Literature: Consult enzyme-specific literature and databases for information on CpG methylation sensitivity and recommended conditions.
Overall, optimizing conditions and exploring alternative enzymes are key to making CpG methylation-sensitive restriction enzymes work effectively for your experiments.
l Take a look at this protocol list; it could assist in understanding and solving the problem.
  • asked a question related to Enzymes
Question
1 answer
I'm writing a report on enzyme activity and characterisation and i need help with understanding this: we were told to report on our results for a 1/10 diluted acid phosphatase. I want to know why we need to dilute the enzyme in the first place and why not use the crude enzyme lysate?
Relevant answer
Answer
The rate of an enzyme-catalyzed reaction is directly proportional to the enzyme concentration. If the enzyme is too concentrated, the reaction may go too fast for the assay. For example, it might use up all the substrate, or the amount of product formed may be too high for the measurement technique. In such a situation, you might not be able to tell the difference between different levels of enzyme activity.
If you are trying to measure the initial rate of the reaction for the sake of studying the kinetics, it is important to keep the rate of product formation within a certain range at which the product concentration increases in direct proportion to the reaction time (i.e. the initial rate), so adjusting the enzyme concentration accordingly is necessary.
  • asked a question related to Enzymes
Question
1 answer
Dear researchers
I want to ask about molecular docking in research. I have a complete research in which I isolated an enzyme. Any researcher can complete my research with theoretical calculations of the isolated enzyme against bacteria. I am waiting for the answer in order to complete the research. Greetings to all.
Relevant answer
Answer
I am virtual docking serine/ threonine kinase and bcl2 apaptosis at the momment it can be useful for you ?!
  • asked a question related to Enzymes
Question
1 answer
we know that chitosan is not actually soluble in physiological body fluid.
Relevant answer
Answer
Biocompatibility, biodegradability, and low toxicity make chitosan a promising medication delivery agent. It has trouble crossing the BBB. Several methods can improve chitosan's BBB crossing. Nanoparticle formulation, surface modification, conjugation with lipid carrier systems, Focused ultrasound technique (FUS), intranasal delivery, Complexation with CPPs, and particle size and charge optimization are examples. Surface changes with targeted ligands and nanoparticles can improve BBB penetration. Through olfactory and trigeminal nerve pathways, intranasal administration bypasses the BBB.
  • asked a question related to Enzymes
Question
4 answers
Hi,
I am measuring the production of 4nitrophenol at 405nm for my enzyme assay.
In the neg control (buffer + substrate) and the enzyme reaction at pH 5.5, 6.5 I observe the absorbance initially increases,decreases and fluctuates . Why is this happening?
I have attached a copy of the pH6.5 timecourse.
Relevant answer
Answer
You need to measure the initial reaction rate. If the change in absorbance is constant over a time period and then declines it is due to substrate being limited or product inhibition.
Can decrease enzyme concentration and standardise by changing substrate concentration till you get a constant reaction rate .
Then monitor initIla reaction rate at constant enzyme concentration and saturating substrate concentration.
  • asked a question related to Enzymes
Question
3 answers
I have been trying to digest a vector pXMCS for the last two months but no luck so far. I have been using NdeI and KpnI (Both of them were present on the plasmid map of the vector) as restriction enzymes and incubating the reaction at 37 degrees for 5 to 6 hours. I am using buffer 2.1 which is compatible with both enzymes. Even overnight digestion does not seem to work out. I also tried single enzyme digestion reactions for 5 to 6 hours and overnight (just to check which one of the enzymes is not working) but this is what I got (2nd picture). I would love to get some suggestions on the same. Thank you.
PICTURE 1: Lane 1: 1 kb plus ladder, Lane 2: uncut vector, Lane 3 and 4: Digestion reactions (with 1ug concentration vector each), Lane 5: Uncut vector, Lane 6: Digestion reaction (3ug vector)
PICTURE 2: Lane 1: 1 kb plus ladder, Lane 2: uncut vector, Lane 3: KpnI, Lane 4: NdeI
Relevant answer
Answer
@Robert Adolf Brinzer - Purchased it 2 years ago I think. Not too sure about the number of freeze thaw cycles. Yes, they were put back in the freezer immediately after use. Both enzymes seem to linearize the vector individually but I dont see the second band after digestion in the presence of both vectors.
@David Lepetit - i am expecting two bands. One around 3kb and the second band between 700bp to 1kb. The vector was isolated using a kit (gbiosciences)
I tried purifying it several times with a different kit as well but I am getting the same result. Should I try isolating the vector manually without the kit?
  • asked a question related to Enzymes
Question
3 answers
Is there anything can be done if the insert gene contains an additional restriction site which recognized by restriction enzyme not only one restriction site at the end of gene sequence, but another at middle of the sequence. In the middle of the experiment when checking for digestion via gel electrophoresis I realize this is the case happened since I find extra band in the gel. (Insert is cut into two parts).
How can I solve this issue if I only have my designed primers for this sequence, one type of polymerase enzyme? (I dont have any other materials which are not needed for this experiment).
Relevant answer
Answer
if the two parts are fundamental for your gene , please purify the bands from the gel , use the ligase and add adaptors for a new cloning by PCR, then you can re-clone using new enzymes fit for the adaptors
  • asked a question related to Enzymes
Question
1 answer
Inhibition of alpha-glucosidase Enzyme in invitro
I read a few articles and saw that the enzyme concentration they usually use is 0.1U or 0.2U but some articles use 1U. So which one is better and based on what factors do we choose this concentration? Thank you very much.
Relevant answer
Answer
The rate of the reaction is directly proportional to the enzyme concentration, as long as initial rate conditions are met. Choose an enzyme concentration that provides a reaction rate that gives you sufficient time to accurately measure the initial rate. If the rate is too fast, you might have difficulty measuring the initial rate. In that case, lower the enzyme concentration to slow down the reaction.
  • asked a question related to Enzymes
Question
2 answers
I need to prepare a liquid culture medium for enzyme production. Kraft lignin will be the substrate in the medium. What would be the best solvent for this?
Relevant answer
Answer
lignin solubility could be increased by up to 70% roughly by the addition of 2% DMSO into the aqueous media...These levels of DMSO must not be problematic for most of the enzymes...
  • asked a question related to Enzymes
Question
5 answers
Hello,
I am measuring thermal stability of a small protein (131 aa) using circular dichroism following the loss of its secondary structure. The data obtained is normalized to be within 0 and 1 where 0 is folded protein and 1 is completely unfolded. The CD of the fully unfolded state was calculated from a different experiment on the same batch and taken as reference. Once plotting my data in Graphpad Prism 9, I am fitting a standard 4PL curve using non-linear regression, constraining the regression to use 0 as bottom value and 1 as top value (see attached file). The Tm is reported as IC50 in this screenshot because this formula is often use for calculating IC50 and EC50. However, the resulted fitted line seems to not being able to represent correctly my data. I performed this experiment twice and the replicate test is showing that the model is inadequately representing the data. Should I look for a different equation to model my data? Or am I making a mistake in performing this regression? Thank you for the help!
Relevant answer
Answer
To ascertain that your model is applicable to your data, plot the residuals (y-\hat{y}) against the independent variable, they should be randomly distributed. You can perform a “runs-test” as a non-parametric test, or (more work, but also more powerful) a t-test to compare the residuals with the standard deviation of the data. Both methods test for H0: The data are reasonably well described by the regression curve, against H1: The data significantly deviate from the regression curve.
Most commercial software performs non-linear curve fitting with the Marquardt-Levenberg algorithm. I had this fail me on occasions and found that Nelder-Mead's simplex-algorithm is more reliable (DOI:10.1093/comjnl/7.4.308). Disadvantage: you need to get the errors of the fitting parameters from bootstrapping (10.1016/0076-6879(92)10009-3), ML gives them directly.
  • asked a question related to Enzymes
Question
3 answers
Should I get certain 'fungi' or buy enzymes and use it directly for biological treatment ? Which enzymes can be used ? Where to buy from ?
Relevant answer
Answer
Muralishwara Kakunje check organic chemicals to reduce lignin content.
  • asked a question related to Enzymes
Question
3 answers
I am currently trying to compile a meta analysis looking at the influence of enzyme activity of xylanase on fiber digestibility.
However, much of the research I have compiled have different enzyme units reported. The main issue is that most articles do not define U. I know the standard definition of U is defined as the amount of enzyme required to liberate 1 umol of substrate in 1 minute under standard conditions but since articles do not report this and simply give enzyme activity as XX U/kg, I have no way to compare these values. The articles also do not confirm that U is defined as international U (IU).
Was there anything I can do to standardize the enzyme activity in order to analyze the reported data? Alternatively, was there any other type of analysis I could still do with enzyme activity data and fiber digestibility?
Thank you in advance
Relevant answer
Answer
I agree with Adam. However, depending on your question, you may not have to compare absolute activities. For example, if different labs report the maximal activity under their respective conditions, and the activity in the presence of an inhibitor under the same conditions, then the v/Vmax values may be comparable.
Just an aside: The correct unit of enzyme activity in the SI is not µmol/min (= U), but mol/s (= katal).
  • asked a question related to Enzymes
Question
1 answer
I need information on concentration of enzyme to be used for particular amount of collagen, the buffer solutions, temperature, time, pH required for the optimal enzyme activity.
According to the papers I referred, I used 0.05M Tris-HCl and 5mM CaCl2 solution for enzyme activity but didn't seem to workout.
Relevant answer
Answer
Hi Siddhi,
You should be more specific in describing what is your starting material, what type of analysis you want to do, what is your aim, otherwise it is difficult to suggest the correct procedure.
  • asked a question related to Enzymes
Question
1 answer
Hi all,
I have an anaerobic bacteria, which could utilize xylan as the sole carbon source. I tested and found that the xylan-degrading enzyme was cell-associated protein. I harvested the cell mass which growth on the xylan (incompletely soluble in broth culture) by centrifugation. The I did the sonication to break down the cell wall and release the cell-associcated protein. Then I centrifuged to collect the supernatant. I precipitated the supernatant by 40% (NH4)2SO4, centrifuge to harvest the pellet. Did dialysis overnight by 1kDa-cut-off dialysis membrane.
Then I had the crude extract enzyme. I incubate the crude enzyme with substrate were oligosaccharides of xylan, from xylose (X1) to hexose (X6). for 24h. But when I develop the hydrolysis product on TLC, the patern of spots were strange. In the hydrolysis of all sets, presence of spots at X3, X4 and X5, even in incubation of crude extract enzyme with Xylose.
Therefore, I think, the presence of X3, X4, X5 in the product enzyme reaction with xylose, might be the remaining oligosaccharide products from broth culture.
Could you give me some suggestion or recommendation to remove this remaining products from crude extract enzyme?
Thank you in advanced.
Relevant answer
Answer
11/23/23
Dear Tra Thi,
I will try to answer your questions and provide some suggestions.
After your initial recovery of the enzyme (i.e., ammonium sulfate (AS) precipitation and dialysis), did you first assay the enzyme on xylan oligosaccharides to see if the enzyme actually had any activity? A quick and easy way to do this would be to incubate the enzyme w/ xylan oligosaccharides, then test for an increase in reducing power. Even without enzyme treatment, xylan oligosaccharides are reducing sugars. However, if they are hydrolyzed by the enzyme, lower molecular weight sugars (e.g., xylose, X2) should be released, so the reducing power after enzymolysis should be higher. There are a number of methods for measuring reducing sugar, e.g., methods based on copper reduction, etc. They are quick and easy to do. One is the Nelson method (Nelson, N., J. Biol. Chem. 153:375-380). It's an OLD method, but it works well. There are other methods that will work. These can be found by doing a google search. This would tell you if your enzyme was active when you did your incubations.
In your incubation, you tested the enzyme w/ xylan oligosaccharides (X3, X4, X5). Even if the enzyme was active, it is possible that it did not completely hydrolyze these sugars, so some of them would remain following incubation, and would show up on TLC. Was xylose present in your incubation mixture along w/ X3, X4, X5? Xylose is one of the sugars you would be looking for by TLC of the hydrolysis products. If it was initially present in your reaction mixture, there's no way to tell if it came from hydrolysis of the larger sugars. Setting up an incubation mixture w/ enzyme plus only xylose might tell you if the enzyme contained other enzyme(s) that converted xylose into something else, although that is not very likely.
When you ran your TLC, did you include enzyme alone as one of the spots on the TLC plate, w/o X3, X4, X5? This should have told you if the enzyme contained any remaining oligosaccharide products from the broth culture. This seems unlikely. When you did your initial AS precipitation, these sugars should have been left in the AS supernatant after centrifugation of the enzyme pellet. Even if some of the sugars had remained, they should have been removed by the dialysis step.
If some of the sugars are still in your partially-purified enzyme, there are a couple of things you can do. One thing is to do a second AS precipitation as before to separate the enzyme from any soluble sugars. Another thing is to re-dialyze the enzyme. This is easier than AS precipitation. If I were doing this, I wouldn't use a 1kDa-cut-off dialysis membrane. I would use a dialysis membrane with a higher mol. wt. cut-off, e.g., 3500-5000 daltons.
I hope this information helps you. Good luck w/ your research!
Bill Colonna
Dept. of Food Science & Human Nutrition
Iowa State University, Ames, Iowa, USA
  • asked a question related to Enzymes
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 Enzymes
Question
3 answers
Attached picture has data obtained from activity (initial velocity) measurement of an enzyme. This enzyme is homodimeric, dimer is more active than monomer. Each subunit has two obligatory Mg2+ at its active site, maybe there are extra binding sites for Mg2+ we don't know of yet.
At [Mg2+] = 0.5 there is this inflection point, which is reproduced in experiments, and I want to make an appropriate equation to fit the data, but I couldn't find the model.
Considering all this, what kind of magnesium binding can describe enzyme behavior giving such activity curve?
Relevant answer
Answer
You could try a two-site equilibrium binding model. It might look like this:
Y = Ax/(B+x) + Cx/(D+x) where B and D are the dissociation constants of the two Mg2+ ions and A+C=100.
Alternatively, you could try a cooperative binding model (Hill equation):
Y = 100xn/(Kd + xn). In this case, the cooperativity looks like it will be negative, so n<1.
  • asked a question related to Enzymes
Question
2 answers
I have insert having flanking regions with XbaI site and and a vector digested with XbaI. How can I prevent self ligation in insert as well as vector before ligation?
Relevant answer
Answer
as Matthew Edward Thornton told it is not ideal since, even if you use CIP treatment of the vector to reduce the probability of self ligation, you will have random and no directional insertion of the fragment and therefore after you need to design and set up a good PCR colony screening that is able to identify the colony where the insert carry the right orientation.
Hovever is suggest to you to skip the use of restriction enzimes and use the PIPE cloning approach. You need just an high fidelity polimerase, to amplify both vector and insert, design primers with overhang extremities and a specific e.coli strain (mach 1 thermo work well)
If you are interested to know more details about it you can read the following papers of the inventors of PIPE:
and/or give a look to the following videos on my blog (ProteoCool)
good luck
Manuele
  • asked a question related to Enzymes
Question
3 answers
I have been trying to determine Km and Vmax of alpha amylase with the MM plot but I cannot reach the saturation point.
My substrate concentration (starch) goes from 0.1 to 3 mg/ml and in my last experiment I tried with U=0.1 for the amylase but my plot still behaves as a linear plot.
I dont know what am I doing wrong. Does any one can share a protocol where you managed to construct the MM plot for this enzyme, please?
Relevant answer
Answer
Make sure the substrate you are using is of high purity. The same goes for the enzyme. Low-quality reagents produce unreliable results.
  • asked a question related to Enzymes
Question
1 answer
I have insert having flanking regions with XbaI site and and a vector digested with XbaI. How can I prevent self ligation in insert as well as vector before ligation?
Relevant answer
Answer
Usually what you do in such case, is that you dephosphorylate your vector with a phosphatase to prevent self-ligation and then perform the ligation.
If the insert self-ligates, it will decrease yield of your ligation, but it won't give you colonies, because it doesn't have ori, selection etc.
The vector should not be able to self-ligate so you shouldn't get colonies with empty vector.
And the vector and insert should ligate at one strand, which is sufficient and bacteria will repair the single strand nick.
However, if possible, you should avoid ligating through the same RE, as it is difficult to ligate and it may insert in both directions (to check quickly, you can design one primer for vector and one for insert and perform colony PCR to quickly distinguish).
If your ligation wouldn't work, I would recommend designing primers for fusion PCR to get your insert into the plasmid quickly.
  • asked a question related to Enzymes
Question
3 answers
I need CEL1 enzyme to use it in my laboratory work to complete my PhD thesis. Unfortunately, I didn't find it anywhere. So I'm waiting for any request about companies name where is available or anyone who sell it for me.
thank you.
Relevant answer
Answer
Unfortunately I couldnt find any company to sell it online, which is a bit weird, but as you probably know, CEL I Endonuclease is naturally exists in Celery juice, which normally utilized for detection of heteroduplex and SNPs, specifically in confirmation of genome editing by CRISPR-Cas. In this link you will find a protocol which has been described an extraction method of CEL1 from Celery juice which could be done by your own.
Also, based on your target in working with such a Endonuclease, you may be able to substitute it by T7E1 which is more common than CEL1 and could be purchased from NEB.
Good luck
  • asked a question related to Enzymes
Question
3 answers
I performed crosslinking using a 10 mg pellet and 5 ml of diluted 2.5% glutaraldehyde for a period of 2 hours. Upon attempting to dissolve the crosslinked enzyme for activity assay, I encountered inaccurate dissolution. Moreover, when compared to the free enzyme, the crosslinked enzyme did not exhibit activity in the activity assay. However, increasing the concentration of the crosslinked enzyme resulted in activity, which is in contrast to the behavior of the free enzyme, which exhibited activity at a concentration of only 100 ng. What further steps can I take to address this issue?
Relevant answer
Answer
Cyanogen bromide conjugation possibly would be a good choice for immobilizion while preserving the enzyme activity...a type of mild biologic attachement procedure....
  • asked a question related to Enzymes
Question
6 answers
Hi everyone.
In our lab we genotype samples for our experiments. Apparently we see a band/bands in our control neg ( MQ water). we have changed everything, the buffers, water, primers, restriction enzyme. but still, we see this band. I am not sure if this is contamination or not.
Do you have any idea how to solve it?
Relevant answer
Answer
Hi,
Thanks so much again, I will try BLASTing and check that out.
  • asked a question related to Enzymes
Question
1 answer
Hello,
I am trying to measure the Kd of an enzyme. I used a fluorescence-based kit and I did a dose-response for the substrate.
I measured over 2 hours the fluorescence intensity.
How am I supposed to measure Kd? and which time point should I choose?
Thanks.
Abir
Relevant answer
Answer
One doesn't usually measure the Kd (equilibrium dissociation constant,
(k-1/k+1) of an enzyme-substrate pair, except in special cases or with special techniques. The usual measurement is the Michaelis constant Km [(k-1 + k+2)/k-1]. The main reason for this is that the reaction is occurring during the measurement, so the substrate gets used up, hence it's concentration is decreasing all the time.
The special case is if the value of k+2 (the rate constant for conversion of the enzyme-substrate complex to enzyme+product) is much lower than k-1 (the rate constant for dissociation of the enzyme-substrate complex to enzyme+substrate), in which case the Km is essentially the same as the Kd. You don't usually have sufficient information to make this assumption, unless it's a very slow enzyme reaction.
The special technique is pre-steady-state kinetics. You need special stop-flow or quench-flow apparatus for this because the measurement is made on a millisecond time scale. You also need some optical (absorbance or fluorescence) readout of substrate binding.
To measure Km kinetically in the usual manner, you must measure the initial rate of the reaction at several substrate concentrations. Plot the product measurements at multiple evenly-spaced time points at each substrate concentration. Draw a straight line starting from the origin and tangent to the earliest time point measurements, including all the data until the reaction starts to slow down. The slope of the tangent line is the initial rate.
Next plot the initial rate on the y-axis versus the substrate concentration on the x-axis. Use a suitable computer program to perform a nonlinear regression to the Michaelis-Menten equation to get the value of Km and Vmax. The substrate concentrations used for the analysis should cover a range below and above the Km to show the curvature of the plot.
  • asked a question related to Enzymes
Question
4 answers
If an active site mutant knocks out product formation at all excessive concentrations of substrate and at all excessive concentrations enzyme, but substrate binding affinity via anisotropy shows no difference in binding affinity for wildtype versus active site mutant enzyme, is kcat = 0? But if kcat = 0, then by the relationship of Michaelis constant (KM) to kcat then KM = KD.
How do you show to reviewers that the active site mutant is dead? If the wildtype mutant starts producing product in seconds, are you supposed to measure the reaction for the mutant for an hour at zero concentration of substrate and at 100fold excess substrate concentration of the K_M for the wildtype enzyme for the active site mutant, and show the time course?
Are there any publications that show a mutant enzyme is not just slow to produce product but is rather incapable of producing product but can still bind substrate? Examples would be greatly appreciated!
Relevant answer
Answer
Short answer - I would say yes. If an enzyme can't turn over products on any time scale, its turnover number would be 0.
I agree that if kcat = 0, then effectively your KM is just the ratio of kon and koff, that is, it is equal to the KD. If one was simply measuring binding of the substrate to the enzyme, I can definitely see that replacement of a residue critical to the mechanism, but not the binding and orientation of the substrate, would generate a variant that does not turn over product but still has a similar or identical KD for the substrate. This says nothing about the turnover number (or lack thereof).
How would I show a reviewer that a binding-competent enzyme is catalytically inactive? I think what you have described is going above and beyond. Extending a reaction for 60 min at 100 x KM to prove no activity exists feels a bit extreme. Even if the measured kcat is very tiny but not zero, it would be a moot point as this has absolutely no biological relevance.
If your wild-type enzyme has a kcat > 1 s-1 like you mention, I think it would be reasonable to measure the initial rates of i) the wild-type enzyme, ii) the catalytic mutant, and iii) a matched enzyme-free reaction using the same reaction conditions for all on whatever time scale is appropriate for your enzyme and its relative rate.
With enough replicates, you can use appropriate statistics to compare the grouped samples. Showing that there is no statistical difference in the rate of reaction between an experiment with no enzyme at all versus a catalytic mutant would be a strong argument that no activity exists, i.e. kcat must equal 0.
You will see in this paper we use an LC-MS assay to detect the presence/absence of product in active site replacements as justification for a dead enzyme, in addition to a colorimetric assay where we did the above to justify activity as "n.d." We did not measure affinity to the substrate but the binding site is > 15 angstroms away in this case.
Hope this helps!
ACA
  • asked a question related to Enzymes
Question
1 answer
I hope someone can help me with this! I am currently working with one enzyme expressed in E. coli. We want to scale up the process to an industrial production (no Academia involved).
The regulation of E. coli BL21 cells is not clear to me. Can be used for commercial/industrial use if used for enzyme production?
If not, what other bacteria can be used instead?
If we use a pET vector system, can this be used for industrial production as well?
Any discussion, advice, or document will be very much appreciated!
Thanks
Relevant answer
Answer
Hi Irene,
E. coli BL21 is one of the most used strains for large-scale (industrial) production of recombinant proteins.
You can check the following link for other E. coli strains for the expression of heterologous proteins:
Or follow the paper below for more information:
Miroux B. and Walker J.E. (1996) Over-production of Proteins in Escherichia coli: Mutant Hosts that Allow Synthesis of some Membrane Proteins and Globular Proteins at High Levels J. Mol. Biol. 260: 289-298.
I have personal experience using different BL21, Rosetta2(DE3) and pLysS strains of E. coli, and all of them express heterologous proteins pretty well for different applications. E. coli Rosetta2(DE3) is another best strain that can be used for the expression of proteins having rare codons.
However, these strains have a major problem of endotoxins which are the unwanted by-products of recombinant proteins purified from any Escherichia coli. As endotoxin could interfere with in vitro biological assays and is the major pathological factor, it must be removed or inactivated before in vivo administration.
Hope the above link and the reference will be helpful for you.
Good Luck,
Faheem
  • asked a question related to Enzymes
Question
1 answer
I have synthesized several compounds that only dissolved in DMSO. The problem arose when I'm doing an enzyme inhibition assay. The compound should dissolved in 5-10% DMSO, otherwise the assay cannot be done. Is it possible if i use 100% DMSO to make a stock solution of my compound and corrected the result using 100% DMSO as negative control ?
Relevant answer
Answer
Yes, you just need proper controls. Enzymes will have different tolerances for DMSO, and so the first step will be to measure your enzyme rate at different DMSO concentrations. Once you know the maximum DMSO tolerated without loss of activity/denaturing, you can design your inhibition assay. If your enzyme loses a lot of activity or denatures at these DMSO concentrations, you might have to try other solvents until you find something compatible.
For example, I worked with an enzyme that retained 100% activity up to 7% v/v DMSO and then started precipitating near 10% v/v. I then wanted to test an inhibitor from 1 mM to 1 µM so I prepared 20X stocks of each inhibitor concentration (20 mM to 20 µM) in 100% DMSO. From these stocks I added the inhibitor to the enzyme to a final concentration of 5% v/v, or 1:20 dilution and the inhibitor did not precipitate. The positive control receives just 5% v/v DMSO with no inhibitor. Using this design you can confidently measure the effect of the inhibitor while controlling for the effect of DMSO consistently across replicates.
Hope this helps!
ACA
  • asked a question related to Enzymes
Question
2 answers
I am currently working with one enzyme expressed in E. coli. We want to scale up the process to an industrial production (no Academia involved).
The regulation of E. coli BL21 cells is not clear to me. Can be used for commercial/industrial use if used for enzyme production?
If not, what other bacteria can be used instead?
If we use a pET vector system, can this be used for industrial production as well?
Relevant answer
Answer
Regulation varies greatly depending on the intended use of the enzyme.
Enzymes for medical purposes / food products each have specific and often rigorous criteria. These are usually defined by the government.
Other uses, e.g. water purification / chemical synthesis / sensing assays, may have more relaxed requirements for usage, handling and safety.
Moreover, regulation depends on whether the product is the enzyme or the enzyme-producing bacteria.
Regarding pET and the BL21 strain, I think that buying the strain/plasmid yourself (i.e. not getting it from another source) allows commercialization, but I could be wrong. Patenting may also be an issue in this case.
Consulting an existing company in your country could give great insight.
Bacillus subtilis has been used for protein expression before, and has commercial strains available (not sure if they are pET compatible). Another common organism for protein expression is the fungus P. pastoris.
  • asked a question related to Enzymes
Question
3 answers
We are working on monoclonal antibodies using a G1 synapt HR mass. we can take a good charge state envelope for proteins with molecular weight of less than 40 KD such as Gh hormone or CAD enzyme. However for monoclonal antibodies with molecular weight  of more than  100 KD such as Ritoximab, charge state envelope has low sensitivity and resolution so that the Maxent software can not calculate  molecular weight of MABs. Our instrument is 8K.
any comment is greatly appreciated.
Relevant answer
Answer
What is the mass range of the instrument?... It should be up to 8kDa to see both high and low charge states to fully characterize the mab. Nowadays vendors are adding biopharma options (software and ESI/Mass filter configuration) to increase the mass range of the mass analyzer to see the lower charge states therefore higher m/z responses...In this case, you may try supercharger agents to be added into the mobile phases to increase the z to get lower m/z ratios. It may be partially helpful in terms of observing a wider charge envelope (higher m/z signals can shift to lower m/z values due to the loaded protons and resulting higher z levels) and the lost signal might get back a bit.
  • asked a question related to Enzymes
Question
2 answers
I want to use the active site structure of carbonic anhydrase enzyme in my paper. Is it better to draw its structure myself with software such as Discovery Studio or Paymol or...? Or is it better to take this structure from other articles and use it in my article and give them a reference?
thank you
Relevant answer
Answer
So it is better to draw the structure of the active site of the enzyme myself.
thanks alot foryour guidance Dear Adam B Shapiro
  • asked a question related to Enzymes
Question
4 answers
Greetings everyone.
During my master's research, I focused on exploring the potential of a specific bacterial strain to produce antibacterial compounds. To achieve this, I used the technique of liquid-liquid fractionation (Extraction) using butanol for the bacterial culture broth. Then, I subjected the supernatant to freeze-drying and further dissolved a portion of the butanol crude extract in methanol for analysis using GC-MS. The results revealed the presence of two secondary metabolite compounds, notably beta-carboline and cyclo-l-proline-l-leucine.
I investigated the genes and enzymes of the bacteria, and it appears that the genes and enzymes that synthesize beta-carboline and related compounds were not present in the bacteria. I have the genomic sequence date of the bacteria. I have searched the genome database to identify any genes or enzymes associated with the production of beta-carboline. Unfortunately, no such gene or enzyme seems to be directly related to the synthesis of beta-carboline in this bacterium. Also, my investigations regarding the McbB enzyme (which is an enzyme that works for the production of beta-carboline) have unfortunately provided no evidence of its presence in my bacterial strain.
So my question Is there an alternative methodology or approach by which I could clarify the mechanisms used by this bacterial strain to produce these compounds? For instance, the synthesis of beta-carboline usually involves the enzymatic action of tryptophan decarboxylase, which catalyzes the conversion of tryptophan to tryptamine. However, my bacterial strain seemingly lacks this specific enzyme. I am hoping that if any practical strategies or methodologies exist, I will try them as a first step to finding some answers for the synthetic pathway.
I sincerely appreciate any insights or directions you can provide.
Relevant answer
I guess your doubt is related to Intra-Cellular metabolites (I guess you have chosen secondary metabolite compounds instead). You can have a look at my recent publication, which was published in RSC Molecular Omics recently. It may drive you to some extent in your research focus.
  • asked a question related to Enzymes
Question
3 answers
I had extracted dna from E. Colli which shown very low digestion even with Hf digestive enzyme It is suspected that becouse of 1-3 minute kept plasmid with Pd3 during plasmid Isolation caused supercoilling of Plasmid which hindering restriction digestion
Relevant answer
Answer
Heating does not remove supercoiling. It requires a nick or break in the DNA by an endonuclease or a topoisomerase or to remove it.
  • asked a question related to Enzymes
Question
2 answers
I cut a vector that already contain the promoter using BstBI enzyme. The electrophoresis gel result showed that no different between control and vector+plasmid cut BstBI enzyme. There are more than 1 band found in the gel. Does anyone have any idea why it is this way?
Relevant answer
Answer
Thank you very much Prof. Liger for your answer
  • asked a question related to Enzymes
Question
3 answers
Hello. This year we started using ITC in a bit unconventional way to study enzyme kinetics of beta-lactamase (there is already literature data that showed it works). The principle is fairly similiar to binding experiments, the difference is that after every injection of substrate to enzyme the baseline does not return to zero, but there is a slight displacement. This differences can than be converted to reaction rates if you know the ∆H of the reaction. The biggest problem we are facing is huge inconsistency in data, especially with controls, where we titrate just substrate to buffer (see the pictures). Also, at the beginning of each titration (both enzyme and controls) we get this endothermic dips, which is weird, because dilution heat should produce exotermic peaks, which are clearly showing up after couple of injections. Anyone has clue what might be going on or have some practical advice? Some useful experimental information: we use Affinity ITC, 190 ul sample cell, 30x2 ul injections with 100s spacing.
  • asked a question related to Enzymes
Question
3 answers
I'm searching how to find enzyme DNA sequence.
I know what strain produce product, and I also know the biosynthesis pathway.
But I can't find how to solve this problem.
Now I'm studying some program, but it doesn't help.
What is the problem?, my ability? or my low knowledge?
Relevant answer
Answer
Isolate the enzyme, characterize it and send its strain for sequencing to know its sequence.
  • asked a question related to Enzymes
Question
3 answers
Hi all,
I'm attempting to clone a GC rich insert (500bp) into a vector that is approximately 5kb and also GC rich. After sequential digests (one enzyme works at 37 while the other works at 65 degrees Celsius), a 0.5% agarose gel reveals that the vector was efficiently cut as indicated by a 500bp shift down of the parent insert vector. Oddly, the 500 bp insert is barely visible. When blown out, the gel shows a smear near the 500 bp region. Is there a reason this is occurring? We are struggling to get any colonies to appear for diagnostic digests so any help would be appreciated.
Thank you!
Relevant answer
Answer
Hi
I agree with D. Liger. You should use more concentrated gel. If the problem still exists, you can increase the time of the restriction enzyme digest.
  • asked a question related to Enzymes
Question
1 answer
I used HEK293 cells as expression system, transiently transfected using PEI. Harvested on Day 4 or sometimes Day 5. The enzyme produced always show up as strong band in cell pellet in western blot , while there is no band in supernatant. While doing enzyme assay, this shows no activity.
I changed my expression system and used Expi293 cell line, which produces an active enzyme.
My questions:
1. Is this related to cell lines, but in past we have seen HEK293 cell line producing any other enzymes?
2. Does vector design plays any role in this?
Iram
Relevant answer
Answer
Expi293 cell lines
  • asked a question related to Enzymes
Question
2 answers
Hi everyone,
sorry if this is a simple question, or if I get anything wrong here, but I'm very outside of my comfort area when it comes to enzyme activity-related calculations.
So, I need to know the concentration of enzymes that a paper used for their stock solution of Catalase.
Here is a quote from the methods: "Enzyme activities of stock solutions were 3 mM/s for GOX and 998 s-1 for CAT. To obtain a defined, stable oxygen concentration of 2% on cell surface stock solutions were diluted by 1:10,000 for GOX and 1:1,000 for CAT."
For the GOX, I think I can manage to calculate the stock, but the catalase is the issue
So, the Kcat = 998 s-1
The Vmax = 1uM/ min = 0.0166uM/ s
For for the calculation: Kcat = Vmax / E[t] , is it as simple as rearranging it to E[t] = Vmax / Kcat?
That Vmax figure is from the data sheet of the catalase, with 1 unit being equal to 1uM H2O2 processed per minute (not 100% sure this is what is meant by Vmax), hence me dividing by 60 to get the uM per second.
I also know that 1mg of Catalase = 20,000U
The issue is that I don't know how to put this all together. I am currently trying to get more familiar with enzyme kinetics, but this is taking some time. I would very much appreciate if anyone could offer some advice to help speed things up so I can start with my experiments.
If I am missing some information here please let me know.
Best regards,
Ciarán
Relevant answer
Answer
The value kcat is the number of product molecules formed per molecule of enzyme per second, and is reported in units of s-1. So I don't think that the description of the stock solution in terms of s-1 makes sense , nor can it be converted to an enzyme concentration. Perhaps you mis-transcribed the units in your quotation from the published account, or there was an error in the paper.
For example, let's suppose the authors meant 998 µM/s as the activity of the stock solution, since the GOX stock solution was defined in similar terms (3 mM/s).
998 µmole/(L-s) = 59,880 µmole/(L-min) = 59,880 U
(The definition you gave for a unit of catalase activity is 1 µmole/(L-min).)
The specific activity you mentioned was 20,000 U/mg.
Thus the stock solution contained 59,880 U/(20,000 U/mg) = ~3 mg.
We don't have information about the volume of the stock solution, unfortunately, so we can't calculate its concentration.
  • asked a question related to Enzymes
Question
2 answers
Good morning, I am trying to degrade an RNA naturally resistant to degradation by RNases, I wanted to use MNase but it only degraded very little. My sample is in liquid medium, in 95uL of RPMI and I added 5uL of Reaction Buffer for a final concentration of 50mM Tris•HCl pH 8, 5mM CaCl2 and heated at 37 degrees for 30 min in the water bath, then I also tried it for an hour and no good results either. I have used 0.5uL (50 Units) and 1uL of enzyme.
I don't know if I'm doing it right, could you please help me, I don't know anyone who works with this enzyme, I've also tried RNAse A/T1 and the RNA doesn't degrade either. Thank you so much.
Relevant answer
Answer
Thanks Thilini for the information, I've been checking and the concentration of salts is critical for the enzyme, I must use a medium with less than 100 mM of salts, but the pH remains above 8, so it's best to use only PBS and/or or the enzyme buffer.
  • asked a question related to Enzymes
Question
9 answers
In order to perform Michaelis-Menten plot to calculate Km for oxa beta lactamase , I used nitrocefin as substrate , 100mM sodium phosphate di basic and 25mM sodium carbonate as buffer pH 7.3
Enzyme concentration 20nM
Substrate concentrations
1 uM
5 uM
10 uM
20 uM
30 uM
40 uM
50 uM
70 uM
80 uM
100 uM
Wavelength 490 nm
In order to calculate Vo ,
I plotted the absorbance values of each concentration vs time. The problem is , the slope for all the concentrations are same which means that Vo of all substrate concentrations are same.
Where is my mistake?
concentrations of the enzyme?
Concentration of the substrate?
Relevant answer
Answer
Ok, looking at your progress curves a few things spring to mind. Firstly, V0 is initial rate and should be taken from the first linear part of the curve, before it starts to level off. This is in practice somewhat arbitrary, but if you do this for your experiment here you should get different inital rates.
However, in your case this brings us to another issue, which is that, particularly in the higher substrate tests, your reaction is essentially over before you start measuring, so you will not get accurate initial rate estimates with this data. I would reduce the enzyme concentration or lower the temperature to get curves that look linear for longer (you need to slow down the reaction). Ideally, you highest substrate concebtration curve should look something like your lower concentrations do now.
  • asked a question related to Enzymes
Question
1 answer
Superoxide Dismutase Assay
Method
Activity of SOD has been determined in two ways:
1. Inhibition by the enzyme of an O2- dependent reaction.
2. Pulse radiolytic methods (Rigo et al. 1975) See: Beauchamp and Fridovich (1971); Misra and Fridovich (1972); Tyler (1975).
The method employed at Worthington is essentially that of Winterbourn et al. (1975) and is based on the ability of superoxide dismutase to inhibit the reduction of nitro-blue tetrazolium by superoxide. One unit is defined as that amount of enzyme causing half the maximum inhibition of NBT reduction. The reaction velocity will depend largely on somewhat variable assay conditions such as light intensity and reaction temperature. Calibration of the method in individual laboratories is recommended.
Reagents
  • 0.067 M Potassium phosphate buffer, pH 7.8
  • 0.1 M Ethylene diamine tetraacetic acid (EDTA) containing 0.3 mM sodium cyanide
  • 0.12 mM Riboflavin (store cold in a dark bottle)
  • 1.5 mM Nitroblue tetrazolium (NBT) (store cold)
Relevant answer
Answer
Cyanide is essential for inhibiting the respiratory process of mitochondria. Without the presence of cyanide, the data obtained would not specifically originate from Superoxide Dismutase (SOD), but rather from the enzymatic activities within the mitochondria themselves, which could potentially interact with Nitroblue Tetrazolium (NBT). The introduction of cyanide serves to redirect the reaction pathway from occurring within the mitochondria to being catalyzed by Superoxide Dismutase (SOD).
  • asked a question related to Enzymes
Question
3 answers
Hi All and thanks in advance.
For long shelf life cleaning solutions, what concentration of protease and what stabilising agent should be added to hypochlorite solution content to preserve the activity of enzyme
Relevant answer
Answer
You will need a protease that can resist both the alkaline (pH 12-13 for the commercial, ~ 10% solution) and the strongly oxydising environment. It probably should be an endoprotease (cleaving in the middle) rather than an amino- or carboxypeptidase (cleaving at either end). You could try to search BRENDA, but it is a tall order.
You will find an exponential decay of the activity of the target protein as function of enzyme concentration and the contact time. Zero remaining activity is reached at infinite incubation time. You have to decide on how much remaining activity can be tolerated in your application.
  • asked a question related to Enzymes
Question
2 answers
I have received the sequence of plasmid, I aligned it with the gene sequence on APE plasmid editor and it is matched. But when I performed vector screening on ncbi, the sequence is strongly matched to vector. Can I make probe with this plasmid by cutting with relevant enzyme??
Relevant answer
Answer
It would appear that you have an insert of around 450nts cloned into your plasmid, based on the simplest explanation of the figure you attach. Does this sound like what you should have?
If the insert is correct then of course you can make a probe from it by releasing with appropriate restriction enzymes or by PCR amplifying the insert region.
  • asked a question related to Enzymes
Question
2 answers
We use Hind 111 is used almost every time but why? How many enzymes do we have to choose per experiment?
Relevant answer
Answer
thank you sir Lassaad Hedhili
  • asked a question related to Enzymes
Question
3 answers
Hello! I'm currently using the IntEnzyDB (https://intenzydb.accre.vanderbilt.edu/kinetics/list) for a machine learning project. During this process, I noticed that for some observations in the Kinetics Data table, the substrate_kinetics column contains multiple substrates in the form of a merged string separated by ";", which is presumably associated with multi-substrate enzymatic reactions. However, the Km entry of the wildtype enzyme sequence (column Km Wildtype) only shows one value, and the same goes for kcat. I've checked some of these reactions in UniprotKB, and there are separate Km values for different substrates. I've looked into the published paper, but I can't seem to find any relevant information. As I understand that the kinetics of multi-substrate systems could be complicated, I'd like to ask whether someone could kindly provide me with some guidance on how to interpret those entries in the database. I really appreciate your time and support!
Relevant answer
Answer
The database was probably created by automated text mining of the literature, with imperfect results. I've no idea why the two substrates are not on separate lines.
Below are the references at the UniProt site for P12268 (IMPDH2) associated with the kinetics data.
From the abstract of Hager et al (1995) comes the following:
"Type I and II IMPDH had kcat values of 1.8 and 1.4 sec-1, respectively, with Km values for IMP of 14 and 9 microM and Km values for NAD of 42 and 32 microM."
From the abstract of Carr et al (1993) comes the following:
"Substrate affinities were similar for types I and II with Km values of 18 and 9.3 microM, respectively, for IMP, and 46 and 32 microM, respectively, for NAD.kcat values were 1.5 and 1.3 s-1 at 37 degrees C for types I and II, respectively."
Source NAD Km IMP Km kcat
Hager 32 µM 9 µM 1.4 s-1
Carr 32 µM 9.3 µM 1.3 s-1
More references with kinetic data for this enzyme can be found in the BRENDA database here:
The values of kinetic measurements depend on the conditions of the measurement, among other things, so expect considerable variation between reports.
  • asked a question related to Enzymes
Question
2 answers
I want to do rolling circle amplification with phi 29 polymerase enzyme . Previous article show they use 10x reaction buffer. Can I use 10X reaction buffer from thermofisher for the PCR.
Relevant answer
Answer
Hi there,
Use the 10x stock of the buffer specific to the polymerase you intend to use.
  • asked a question related to Enzymes
Question
3 answers
I would like to performe the GRIESS reagent kit from biotium. The nitrate reductase enzyme (Sigma catalog no. N7265) has an activity ≥300 U/g. By protocol I am to use it at a final concentration of 300 U/L. The enzyme is in lyophilized powder form. How many microliters of grade water should I add? And how many microliters should I take of my solution to have final concentration of 300 U/L?
Please provide your suggestions.
Relevant answer
Answer
The product appears to be sold in units, in which case the u/g value has no bearing on what you need to do. The largest pack is 10 units, and you want 300U/L final conc: or 0.3U/ml. Your reconstituted stock therefore must be >0.3 U/ml if you are going to add it other reagents and still achieve a final concentration of 0.3U/ml. For example, you could reconstitute the 10U of enzyme in 333ul, which is 30U/ml i.e. 100x stock. Then you would add 1ul to 99ul to achieve the required conc.
The last sentence of your question however is unclear, as the concentration of enzyme once reconstituted won’t change regardless of how many uL you withdraw.
  • asked a question related to Enzymes
Question
6 answers
I'm designing a new activity for an undergraduate biology course about enzyme activity. I'm running into a slight complication. I've ordered a lyophilized powdered version of human salivary alpha amylase.
The issue is "how long can it be stored once dissolved?".
Most protocols say to use "freshly made" enzyme. But the amount needed for a lab section is too small to weigh out. Seriously, the bottle has 1000 Units and is only 10 milligrams. Diluting to 1 Unit/mL for a working concentration makes 1 Liter of enzyme solution. That is more than enough for an entire week of lab sections!
But, will it denature/lose too much activity in the refrigerator?
Looking for some practical advice!
Relevant answer
Answer
Were did you purchase this Amylase? Was there a specific reason why you are using a human source material? I work for a company that has Amylase from other sources and it is available as stabilized liquid that is treated with a protease inhibitor.
  • asked a question related to Enzymes
Question
3 answers
Hello all,
I am over-expressing acid sphingomyelinase enzyme in HeLa cells. Theenzyme shows expression on western blot but I do not have any activity in in vitro assays. I have checked the activity through Mass Spec as well as radioactivity. Please suggest where could I be going wrong? Thanks!
Relevant answer
Answer
Hi there,
If your enzyme of interest is produced then there might be an issue with its folding/stability (if not properly folded/matured the protein will exhibit no activity), cell extraction (possibly provoking the loss of enzyme activity) and/or with the assay itself (compatibility with the source of enzyme you use).
  • asked a question related to Enzymes
Question
1 answer
I want to see polymorphism of Tumour necrosis factor alpha (308 G>A, rs1800629) polymorphism. The sequence of that region is given below:
AGTTCTATCTTTTTCCTGCATCCTGTCTGGAAGTTAGAAGGAAACAGACC ACAGACCTGGTCCCCAAAAGAAATGGAGGCAATAGGTTTTGAGGGGCATG [G/A] GGACGGGGTTCAGCCTCCAGGGTCCTACACACAAATCAGTCAGTGGCCCA GAAGACCCCCCTCGGAATCGGAGCAGGGAGGATGGGGAGTGTGAGGGGTA
So, I need a restriction enzyme which will cut on the bracket region in presence of either adenine or guanine. Most of the articles, I have Found that NcoI enzymes have been used for this purpose.
NcoI enzyme cleaves when this sequence present:
5'  C ↓C  A  T  G  G   3' 3'  G  G  T  A  C ↑C   5'But this sequence is not present in the above sequence . Rather the sequence is "GCATGG"  As a result when I am using tools to find out restriction enzymes, NcoI enzyme shows" 0" cut. I did not not find any other restriction enzymes also which will cut on that site. So, my questions are: What may be the possible solution? Where I am doing mistakes on searching?
Relevant answer
Answer
Hi there,
obviously the sequence you provide is wrong! The actual sequence is not GGGGCATG [G/A] but GGGCCATG [G/A] (according to the following paper ) which results in the presence of one NcoI site if G is present and in the loss of the site if A is present...
  • asked a question related to Enzymes
Question
1 answer
Hello, so i am scaling up a 10 L biorector to 1000 L bioreactor for enzyme production using corncob-based media. The product increased, but there's a lot of excess media left in the product. So, i want to minimize the media impurities before purifying the product. What should i assess regarding this matter? Thank you
Relevant answer
Answer
The key for fermentation scale up is to keep some key parameters the same or proportional. These important parameters may include mixing times, power to liquid volume ratio, oxygen transfer coefficient etc. The following are some useful article to guide fermentation scale up:
Article Scale-up of Industrial Microbial Processes
If there was no excessive medium at small scale, then the reason for excessive media at scale up is likely due to the unproportional scale up that changed some key parameters.
  • asked a question related to Enzymes
Question
1 answer
One Thing, Your Physics Book Many Equations Depend On Uniform Velocity And Acceleration Equation But Nothing Is Uniform All Time And Change Should Be And Must Be Come Every Where In Universe About Velocity And AccelerationAt Physics Books Those All Uniform Velocity And Acceleration Can Be More Or Less Correct For All Uniform Velocity And Acceleration World But Not For Practical Life Equation Cause Nothing Is All Time Uniform In This Life And In Heaven Also.When Moment Change And Time Change And When There Are Day,Night,Morning,Noon,Afternoon,Evening,Night That Moment Change Sky,Cloud,River,Pond,Mountain,Rain,Storm,Breeze,Drizling,Cats And Dogs And Others Then Also Velocity And Acceleration Of Life And Planet Change And When Mass Increase.Without There Would Not And Will Not Life Beauty.Life Would Be And Will All Time Alike And All Time Uniform And No Taste And Color Of Life.Kids Would Be Forever Kids,Parents Would Be Forever Parents. Universe.Its Research And Scientists Say Planets Movement And Rolling Never Follow Isac Newton Equation And Isac Newton Rule
Newton”s First Law:No Outside Force Motionless Elements Forever Motionless But Motion Elements All Time Motion.
Ans:Wrong Because No Outside Force Elements Has Inside Force.And Others Wise Actually Elements Are Not Only Electron,Proton,Neutron And Every Elements Has Different Characteristic And That’s Why Chemical Characteristics Different And That’s Why Their Strength And Characteristics Different And That’s Why Elements Not Only Electron,Proton,Neutron And There Are Many Atom Just Like Electron,Proton,Neutron That’s Why Characteristics Different And That’s Why Different Different Elements.There Are Many Atom(More Than 150 Atom Inside Every Chemistry Elements And Its Atom Number Is Vary Elements To Elements-If We Are See Chemistry Nucleaus And Chemistry(Organic Or Inorganic) Elements Or Reaction Under Plasma Microscope Or Satellite Frequency Microscope Or Very Strong Microscope Than We Will Be See Totally Different World Of Chemistry Which Is More Or Less Totally Different From Our Chemistry Book And Also Never Ever Only Chain Reaction Happens In Organic Chemistry.And Chemistry Nucleas Is That Which Secret Frequency And Secret Vein Just Like Leaf To Compact Others All Atom And Under Plasma Microscope Or Satellite Frequency Microscope We Will Be Able To See Accurately Nucleus Which Secret Frequency And Also Vein Just Like Leaf To Compact All Atoms.Nuclues Is Never Proton And Neutron…..If Nucleaus Is Only Proton And Neutron Than When Environment Partial Reaction Will Be Happen And Temperature,Air,Light,Color And Environment Different And Fraction Will Be Come Than When Proton Will Be Go Away And All Over Atom Distribution Will Be Broken And All Will Be Broken.So,There Are Many Atom Inside Elements. . Chemistry Basic Atom Not Only Electron,Protron Or Neutron And There Are Others Basic Atom Within Elements And That’s Why Characteristic Different And Vary Places To Places.Changeno Atom,Airono,Waterono,Cloudono,Massono,Environmentallono,Specifino,Tastetono,Reactono And Others Many Atom(Just Like+_*%#!) Within Elements And It Is Vary Environment To Environment And Places To Places Or Planet To Planet.Atom Distribution Equation-----q*v*theta(sin,cos,tan or others)/Time Or Changing Time----Here---q=Atom Charge---Velocity----Theta---Atom Position With TimeIf We Are See Chemistry Nucleaus And Chemistry(Organic Or Inorganic) Elements Or Reaction Under Plasma Microscope Or Satellite Frequency Microscope Than We Will Be See Totally Different World Of Chemistry Which Is More Or Less Totally Different From Our Chemistry Book.And Chemistry Nucleus Is That Which Secret Frequency And Secret Vein Just Like Leaf To Compact Others All Atom And Under Plasma Microscope Or Satellite Frequency Microscope We Will Be Able To See Accurately Nucleus Which Secret Frequency And Also Vein Just Like Leaf To Compact All Atoms
Now Without Outside Force Motionless Elements Never Forever Motionless Or Motion Elements Never Forever Motion Because They Have Specific Characteristics And Inside Temperature, Pressure And Atom Elements Just Like Electron,Proton,Neutron And They Will Do Reaction And Others Velocity And They Have Half Yearly Age Because Every Elements Should Be Do Reaction And Die Or Convert Or Change Because Planet Mass Increase Everytime And When Only If Single Something Change Than Change Everything.So,Aisac Newton Equation Should Be Wrong.Aisac Newton Equation Can Be True That Time When There Would Be No Constellation Or Universe Because Change Should Be Come Today Or Tomorrow And Every Time Because Nothing Is Uniform Velocity And Acceleration Here And In Heaven Life Forever.
Solve Equation:No Outside Force Motion Elements Can Be Motion But Once Inside Electron,Proton,Neutron And For Others Basic Atom Like Airono,Pressurono,Timeno,Frequencyiano,Reactionon,Relationono,Rationono,Waterono,Environmentolono And For Others Basic Atom Motion Elements Can Be Do Reaction And Once Motionless And It Can Be Slight Motion Increases But Once Motionless Cause There Are Many Many Basic Atom In Environmental Elements And Their Ratio And Velocity Different And That’s Why Characteristics Different And Sometimes Vary Place To Place Or Place And Distance Change And They Have Half Yearly Age And Environment Has Recycling Process And Partial Reaction And Its Gonna Convert Others Like Dhancha Plant When Die Then Convert As Green Fertilizer.And Same Words For Motionless Elements.And Motion Elements Never Wanna Forever Motion And Motionless Elements Never Wanna Forever Motionless Cause It Has Utter Energy And Basic Atom Reaction And Velocity And Ratio And Environmental Partial Reaction And Recycling Process For Life.
Now,Suppose Anything Is Motion And No Outside Force:Motion Elements=Total Unit( Time And Others)*Summation Of Velocity*Inside Utter Energy Change*(Like Basic Atom Ratio And Velocity Change And Environmental Partial Reaction And Recycling Process Change)=Last Step Can Be Motionless Or Sometimes Slight Motion Increases And Once Inert Elements And Scattered………… And Here Elements Came From Environment.And Motion Less Elements Will Be Slight Motion And Once Scattered And Equation Motionless Elements=Total Unit( Time And Others)*Summation Of Slight Increases Velocity*Inside Utter Energy Change*(Like Basic Atom Ratio And Velocity Change And Environmental Recycling Process Change))=Last Step Can Be Motionless Or Sometimes Slight Motion Increases And Once Inert Elements And Scattered.
Newton 2ndLaw:Force Proportional Momentum Change(Wrong)
Ans:When We Give Force On Elements It Can Be But Elements Has Also Own Force And Momentum.Example:Suppose Two Things Running:
M1=2, M2=50
V1=3,V2=80
K1=3,K2=80
So,It Should Be After Collusion Small Things Velocity Must Be Highly Increase And Big Things Velocity Also Increase And It Depends On Two Elements And Environment.
Solve:Force And Momentum Change Depends On Elements.Suppose One Ball When You Will Be Kick Then It Will Get Easily More Velocity But When You Will Convert Same Ball To Any Sheet And Give Same Force Then Its Momentum Change And Both Momentum Change Should Not Be Same.And Force And Momentum Change Depends On Elements And Size And Environment Also.Suppose You Are Wanna To Fall One Thing From Mountain Then Just Touch And It Will Be Get Huge Force And Velocity But Your Force Was Very Slight And If You Will Give Same Same Force On Normal Environment Then It Will Not Get Same Velocity And Force.So,Its Depend On Environment And Environmental Recycling Process.
Equation:F1=M1*Dx1/t1 And F2=Dx2/t2 Now After Accident F1*F2=M1*Dx1/t1*(Momentum Change That Distance And Time That Velocity With Time Change Unit With Environmental Change)*M2*Dx2/t2
Now F1**(Momentum Change That Distance And Time That Velocity With Time Change Unit With Environmental Change)=/<_(Not Equal)>_(Not Equal)F2*(Momentum Change That Distance And Time That Velocity With Time Change Unit With Environmental Change)
Newton 3rdLaw:Every Force Has Equal And Opposite Force(Wrong And It Can Be True For Slight Time But Not All Time) Ans:When We Give Force On Elements It Can Be But Elements Has Also Own Force And Momentum.Example:Suppose You Are On Mountain And Just Touch A Big Stone By Finger Which Will Be Go To Touch Soil And Get Huge Force When Newton Under Apple Tree And Break Newton Head.So,What Will Be Opposite Force Of Newton Broken Head.Or You Are Pushing A Pin At Wall By Hammer…Pin Is Going To Wall But Hammer Does Not Get Same Opposite Force.Suppose Two Things Running:
M1=2, M2=50
V1=3,V2=80
K1=3,K2=80
So,It Should Be After Collusion Small Things Velocity Must Be Highly Increase And Big Things Velocity Also Increase And It Depends On Two Elements And Environment.So,Elements Force Should Not Be Equal And Opposite. You Are Giving A Kick To A Truck And Truck Has No Reaction Force But Your Leg Is Broken.Truck Not Give Same And Equal Force.Newton Gave Bullet And Gun Equilibrium Force Equation But Now Many Bullet Donot Give Opposite Force After Release.And Same Equation For Boat Also.And I Am Giving Something Fall From Moutain Or Foorball Or Truck Velocity And Acceleration Equation.
So,It Should Be After Collusion Small Things Velocity Must Be Highly Increase And Big Things Velocity Also Increase And It Depends On Two Elements And Environment.So,Elements Force Should Not Be Equal And Opposite.
Equation:F1=M1*Dx1/t1 And F2=Dx2/t2 Now After Accident F1*F2=M1*Dx1/t1*(Momentum Change That Distance And Time That Velocity With Time Change Unit With Environmental Change)*M2*Dx2/t2
Now F1**(Momentum Change That Distance And Time That Velocity With Time Change Unit With Environmental Change)=/<_(Not Equal)>_(Not Equal)F2*(Momentum Change That Distance And Time That Velocity With Time Change Unit With Environmental Change)
Newton Gave Bullet And Gun Equilibrium Force Equation But Now Many Bullet Donot Give Opposite Force After Release.And Same Equation For Boat Also.And I Am Giving Something Fall From Moutain Or Foorball Or Truck Velocity And Acceleration Equation.
And For That Motion Of Rocket Equation Was Wrong Because It Was Depend On Newton”s 3rdLaw: When You Will Be Go From Soil To High Sky Than Gravitational Attraction Increase That Velocity Decrease But Once Near Unventilated Places Gravitational Attraction Decrease And Rocket Velocity Increase And Plane And Rocket Different Things But Alike.And Free Velocity Should Not Be All Time 9.8 ms-2.And It Should Be Different Elements To Elements Either Up And Down.So,When You Are To Go Up Than Every Second Before Reached Unventilated Place Velocity Should Be Decrease For Gravitational Attraction.
Motion Of Rocket:
Rocket Mass:M
Gas Mass: Δm (Delta Mass) In Every Seconds
One Second Gass Mass = Δm
Δt (Delta Time) Gass Mass= Δm (Delta Mass)/ Δt (Delta Time)
Now Force F=Ma
HERE, M=Rocket Mass
a=v/t,
Δm (Delta Mass)/ Δt (Delta Time)*t
.
Force=M(Rocket Mass)* Δm (Delta Mass)*Increase Gravitational Attraction That Deacrease Rocket Velocity With Environmental Temperature And Pressure And Others And Increase Velocity By Force With Environmental Temperature And Pressure/ Δt (Delta Time)
So,HereGass Mass Should Be Higher Than Or Bigger Than Rocket Mass And Everything Seconds It Will Change For According To Environment.So,Newton 3rd Law Should Not Be Work Here Beccause It You Are Use Newton 3rd Law Than Rocket Shuttle Should Be Broken For Environmental Pressue And Temperature And Rocket Should Be Fall From Sky.
Now Neednot And Shouldnot Use Rocket.Now You Can Use My Triangle Vehicle Which Will Be Go Electric Recycling.IDonotKnow,You Will Be Albe Or Not My Triangle Vehicle Cause It Has Very Sharp Design.You Can Try But May Be You Cannot Make My Triangle Vehicle Which Will Fly In The Sky,Run On Road,Swim On Water And Dive Under Water.And.You Can Try But It Has Very Sharp Design
Now………1.Anything Is Running That Motion And Last Point Reached Equation:
Let,Suppose One Thing Is Running On V Velocity
After One Second(1s) It Will Be Go=V*1
Now T Second It Will Be Go=V*T But Now What Will Be Its Velocity Change And Equation:
In One Second=V*1 That Dx1/t1 Now 2 Second It Will Be Dx2/t2 And 3 Second It Will Be Dx3/3 And 4 Second It Will Be Dx4/4.Now Dx Can Be Change From 1 Second Or 2 Second Or 3 Second Or 4 Second.Now What Will Be Motion Elements Reached Point Equation After Time:
It Will Be Dx1/t1+Dx2/2+Dx3/3+Dx4/4 That 4Seconds*Summation Mean Of Velocity Change Or Velocity Summation (Dx1/t1+Dx2/2+Dx3/3+Dx4/4) That 4*Summation Mean Of Velocity Change And Others All Plus In 4 Seconds.
That((Dx1/t1+Dx2/2+Dx3/3+Dx4/4).
So,Specific Point Reached Equation After Time And Velocity Change Will Be
Specific Point Reached=4*Summation Of Mean Of Velocity And Time Change That Distance And Time Change And Others All Plus In 4 Seconds.
Specific Point Reached=Total Unit(Time And Others)*Summation Of Velocity
And Specific Point Reached Mean Velocity=Total Unit(Time And Others)*Summation Of Acceleration(Acceleration Mean Change Of Velocity That Distance And Time Change)
So,All Time Changing Velocity(Not Uniform) And Acceleration Equation Is:
V1+V2+V3+V4+V5 And a1+a2+a3+a4+a5
SPR=Total Unit Or Total Time*Mean Summation Of Velocity Or Acceleration.
Now Last Velocity When Velocity Change All Time More Or Less.So Last Velocity And First Velocity(Not All Time Uniform):
SPR=V1+3*Mean Summation Of Three Velocity+V5
Now,S-3*Mean Summation Of Three Velocity=V1+V5
S-F(F3MSTV)=V1+V5
Now v5=E-V1…….Here (S-F=E)
Motion Of Falling Elements:
1stVelocity=0 But When Fall From Upper Sky Velocity Low But When Fall From Near Soil Velocity High For Attraction Cause(Lower Portion Gravitational Attraction Is High).
Suppose Falling Fruit From Ifel Tower Head And Every Meter Per Second ms-1 Velocity Increases Not All Time Alike 9.8 Ms-2. Cause Its Also Depends On Environment And Elements.Its Also Not Possible For All Uniform Equation Cause Elements To Elements 9.8 Vary.
Here Suppose Vo=0 Now Vo+V1*1+V2*2+V3*3(Here Velocity Increases With Fall Attraction)
Now Last Velocity Will Be=Vo+Increases Attraction*Summation Of Mean Velocity
Suppose Increasing Attraction Velocity=g
So,Last Velocity Will Be=Increases Falling Attraction*Summation Mean Of Velocity*Total Time+Vo
So,Last Velocity Will Be=Mean Increases Falling Attraction Velocity*Summation Of Mean Velocity*Total Time+Vo (Some Error Can Be Come But Error Should Be Identify)
And So,After Soil Touch Velocity Equation Will Be=(V-Last Velocity Increases Attraction After Touch)=Increases Mean Falling Attraction Velocity*Summation Of Mean Velocity*Total Time+Vo
(If Need You Can V-LVIAAT Or V+LVIAAT)After Touch AsNecessity.Here Vo=0 Others Can Be
Now Last Velocity And Distance Equation Will Be That After Soil Touch Falling Elements Equation Will Be=Vo+Summation Of Mean Velocity*Mean Increases Falling Attraction Velocity*Total Time+Velocity-Last Velocity Increases Attraction*1unit
From Soil Low To High:VelocityDesreases With Gravitational Attraction But Once Near Two Point Of Planets That Near Unventilated Places Velocity Increases
That SPR=Vo+Summation Of Mean Velocity*Total Time Unit*Decreases Attraction Velocity+(V+Last Velocity Decreases)*I Unit.
Laws Of Static Friction:
1.Motionless Varnish React Inverse Of Elements Motion
Ans:It Can Be True But Not All Time.Suppose One Boat On River.And Boat Is Motionless But River Water Wave Velocity Increases For Air Environment.And Environment Has Changing And Recycling Process.So,Boat And Water Varnish Both Velocity Also Increases.So,Equation Is Going To Be Wrong.Now It Can Be Sometimes True Or Many Times Also Like Cycling On Road Or It Can Also True Motion Increases But Mass Huge And That’s Why Gonna To Be Slow Or Stop.
Equation:Motionles*Varnish Force*Last Velocity Will Be=(V-Last Velocity Increases Or Decreases Attraction)*Increases Or Decreasing Attraction*Summation Mean Of Velocity*Total Time+Vo
=(V-Last Velocity Increases Or Decreases Attraction)*Increases Or Decreases Mean Attraction Velocity*Summation Of Mean Velocity*Total Time+Vo …..Here Vo Can Be=0 Or Others
Equation2:Motionles*Varnish Force Proportional To Stop Or Obstruct Force
Ans:How It Can Be True Cause Water And Boat And Mountain Or Truck Or Cycle Or Football Example.Cause Sometimes Can Be Increases Or Decreases
Equation Just Like 1st And S1=U1VI And S2=U2V2 And So,Here Not Proportional.
Equation3:Angel Of Repose Proportional To Last Border Varnish
Answer:Suppose One Boat On Rivet Or One Oil Machine Machine By Cow.If You Will Be Give Slight Force On Boat Border Than Its Move More Cause Lower Touching Portion Varnish Is Soft And It It On Soil Or Muddy Then It Would Be Give More Force To Move.Suppose Football Player Corner Kick,ItsGonna Here And There And Many Times Not On Specific Cause Environmental Cause Air,Temperatire,Pressure And Own Utter Energy Cause.So,Its Depend On Environment And Utter Energy Of Both Elements.
Equation Like Before And You Can Use My New Mathmatics Equation But It Can Be Increases And Decreases As Necessity And Length And Angel Can Increases And Decreases And But It Will Be Very Helpful For Cow,Goat,Horse That Animal Organ Convert For People Like Bones,Eyes If You Are Modify And Change Them 2 Or 3 Places Or More As Organ Then You That People Can Use Those Normaly But Need Plant And Plant Enzyme And Others Enzyme And Color Convert Also.
Equation4:Varnish Mean Never Depends On Touching Places And Volume
Answer:Its Ultimately Wrong If You Read My Previous Upper And Others Equation.
Equation5:Varnish Mean Never Depends On Area
Answer:UltimatelyWrong.Cause One Big Thing And One Small Thing Varnish Should Not Be Same And Also Depends On Both Area And Environment Also Like Water,Desert,Normal,Pitch Road Or Normal Village Road And Environment Also.
I Am Giving You Roult Law Example:
When 2 kg Ice+2kg Water Convert To Water Then Its Is 4Kg But According To My Law When 2kg Ice+2Kg Water Convert To Water Then It Can 4.10 Or 3.90 Or 4.20 Kg Cause Environmental Elements Enter Within It.LikeTemperaure,Pressure,Light,Master Force And Others And Why ItsGonna To Be Convert Without Why It Will Be Convert.And When Enter Something Within And React And Then 2+2=4 Kg Should Not Be 4 Kg And It Will Be 4.10 Kg Or 3.90 Kg Or 4.20 Kg Depends On Environment.So,NewRoult Law Will Be According To Me: (n2+n1)/n1=D(P2- Or + P1)/P1
Or (n2+n1/n2)=D(P1- Or + P2)/P2
Or (n1+n2)=D(P1- Or + P2)
Or (n2+n1)=(P2- Or + P1)D………Here P1 And P2 Next Situation Of n1 And n2. D=K And According To My Previous Equation K=Pressure*Temperature/N(Atom)*Volume Or D=K That K=Temperature*Pressure/Volume
Graham Law: Motionless Pressure And Temperature Any Gas Diffusion Per Inverse Density Rot
Answer:Gas Diffusion r=KTPD/Delta Time Here K=Increase Or Decreases With Temperature And Pressure Like Attraction Or Repulsion Equation
MarkonicovLaw:R-CH=CH2+HX …..Here (=)=Two Double Bond Now Solve Equation Will Be R-CH-H-CH2-X It Will Be True Equation Reaction Cause Asymmetrical And Unconnected Or Not Related.
Now You Can Think All About Your Study Equation They Are True Or Not And There Are Fit For Life And Environment Not Cause I Guess They Are Not Fit For Environment And Life Cause All Uniform Velocity And Acceleration Equation And Modified Equation Which Is Not Fit For Life And Forever Cause Here And There And In Constellation Or Universe Nothing Is Uniform Velocity And Acceleration Equation.People,Vehicle And Planet And Constellation And Universe Never Walk Or Run Or Motion Or Motionless As Uniform Velocity And Acceleration Equation.ItsGonna Change When Time Change Or Mass More Or Less Change And Moment Change Like Strom Is Not Uniform Velocity,Tsunami,Rain,MoonLight,Sunlight,Vehicle And Others Nothing Is Uniform Velocity And Acceleration Motion Or Motionless But All Your Study Equation Uniformly Go.I Am Solving According To My Own.ButIts Your Matter Accept Or Not.Now You Can Think Your Own Way And Try To Solve If You Are Guess They Are Wrong.
And Albert Inestine And Salam Glass And Stephen Hocking Equation Wrongly Prove From My Plant Energy,Fragrance,Color And Others Planet Light Absorb Or Mine,Pit,Quarry Find Equation. .............Are Those True About About Uniform Velocity And Acceleration Equation And About Isaac Newton Equations Wrong & Error?.........Please See Attachment For Further Attachment And Discussion.
Relevant answer
Answer
I'm wondering if somebody has read this opus from the beginning to the end? I would award Mehadi the Nobel price in literature if he learns that Albert Einstein but not Albert Einestine created the theory of special relativity. Anyway, one more genius in RG
  • asked a question related to Enzymes
Question
5 answers
Dear colleagues,
For genomic library preparation we need to obtain Tn5 enzyme. Unfortunately, In-house-produced Tn5, which was purified on Ni-column, has a nuclease activity without loaded adapters. Сould this be due to the incomplete enzyme purificatoin from the gDNA using PEI? Is this related to accidental nuclease contamination?
Relevant answer
Answer
Thanks for the reply. So you forego the SENP2 cleavage between NI-NTA column and SEC?
  • asked a question related to Enzymes
Question
3 answers
Dear All,
Hope are you doing well.
I am working on beta lactamase inhibitors. I have beta-lactamase-producing bacteria and i will have to check the concentration and enzyme activity for beta-lactamase using nitrocefin. Kindly share the protocol for the same.
Thank you
  • asked a question related to Enzymes
Question
4 answers
detailed explanations and procedures if any
Relevant answer
Answer
For more info please folllow the arts links below:
Protocols for Extraction, Isolation, and Purification of Secondary Metabolites of Mushroom and Its Applications | SpringerLink
Isolation and Purification of Enzymes | SpringerLink
Methods for Isolation and Cultivation of Filamentous Fungi | SpringerLink
Isolation of Enzymes | SpringerLink
Isolation and Screening of Cellulolytic Filamentous Fungi - PubMed (nih.gov)
Methods for isolation and cultivation of filamentous fungi - PubMed (nih.gov)
Isolation, Purification, and Characterization of Fungal Laccase from Pleurotus sp. (hindawi.com)
  • asked a question related to Enzymes
Question
5 answers
Anyone have any experience using the EnzChek pyrophosphates kit in the presence of citrate. There appears to be something strongly absorbing at 360 nm prior to the addition of the last enzyme (PNP).
Relevant answer
Answer
Hi Ziyu Song ! I am afraid not. I changed the kit altogether.
  • asked a question related to Enzymes
Question
4 answers
Hello, I am planning an experiment to measure enzimatic activity, and i will take sample every 24hrs. The only problem is that substrate is non soluble... nevertheless, enzyme can still access to it. My idea is to keep the reaction on movement, but i would like any suggestion on how to lessen error when taking the sample, because last time i tried doing separate reactions in different tubes for each measure (e.g. i had 3 different tubes for T0, 3 dif. tubes for T2, and so on), but replicates wheren't as much as similiar as I expected them to be. My first thought was to make a reaction stock (on triplicate), and take a volume of it each time, but I believe that since the subtrate isnt soluble, each time I take a sample, subtrate might vary, and i will still get error...
Is there an alternative for this type of experiments?
If not, which way is better: a stock reaction or multiple reactions?
Thank you in advanced for reading.
Relevant answer
Answer
I think the most important point is to make sure the insoluble substrate is well-suspended at all times. Given that, I would expect to get the same results whether using a single volume sampled multiple times, or separate volumes for each time point.
Since enzymes can lose activity if mixed too vigorously, you have to find the optimal compromise between keeping the substrate suspended and causing damage to the enzyme from excessively vigorous mixing.
Oxidation of the enzyme (and substrate?) may also be a problem for such a long-term experiment. It might be helpful to flush the container with nitrogen to reduce oxidation.
  • asked a question related to Enzymes
Question
1 answer
I am working on Nitrocefin assay for screening of beta-lactamase inhibitors and for that I would need to generate the standard curve using Nitrocefin. For my assay, I would be using crude beta-lactamase enzyme and inhibitor which will be incubated and later Nitrocefin will be added and evaluated using microplate reader at 490nm. Can I plot standard curve using Nitrocefin (fixed concentration) with enzyme unit (varying concentrations) or Is there any other methodology ? Please suggest as I am not using any readymade kit and need to develop a simple working procedure in a lab.
Relevant answer
Answer
The enzyme activity is quantified by the initial rate of the absorbance increase (the slope of the A490 versus time curve at the start). To convert this into the rate of nitrocefin hydrolysis (µmoles/min), you need to measure the extinction coefficient difference at 490 nm between nitrocefin and its product.
Since you don't have the product, you can make it in situ by mixing various concentrations of nitrocefin with enough of the enzyme to completely convert it to product in a reasonable amount of time, subtracting any background from the same concentration of enzyme without nitrocefin (since it is an impure extract). Plot the absorbance at the endpoint versus the concentration. The slope of the plot is the extinction coefficient difference (Delta A490/µmolar).
Once you have that number you can convert the A490/min initial rate to µmolar/min by dividing by the extinction coefficient difference (A490/µmolar). Multiply by the reaction volume in liters to get µmole/min.
A nice advantage of this method is that it automatically accounts for the geometry of the measurement, i.e., whether it is in a cuvette in a spectrophotometer or a multiwell plate in a plate reader, as long as the standard curve is prepared under the same exact conditions as those in which the beta-lactamase assay is run.
  • asked a question related to Enzymes
Question
4 answers
We digested pUC57 with the XcmI enzyme, and a non-specific band at 1500bp appeared alongside our intended band (2750bp). To diagnose the issue, we attempted gel purification on both the digested and undigested products, but received the same result. Additionally, we tried heating the digested and undigested products to dissolve secondary plasmid formation, but the same result occurred.
is there anyone with same issue who can help us please?
Relevant answer
Answer
Dominique Liger We also tried that, with KpnI and BamHI there was no extra bond, but with XhoI and XcmI the extra band appears again(sometimes with XhoI and sometimes with XcmI)
  • asked a question related to Enzymes
Question
2 answers
Antibiotic resistance , enzyme production are types of screening method . Both have advantages and disadvantages so , in these two which method is more preferred to insert in vector and provides maximum results . Are there any considerations of these methods while performing gene cloning?
Relevant answer
Answer
Hi there,
Your question is a bit of unclear to me... When you mention enzyme production, do you mean the enzyme produced by the gene conferring resistance or do you mean the product of the gene of interest possibly cloned into the vector?
  • asked a question related to Enzymes
Question
1 answer
All other aspects of how the simulation runs is ready. I am now creating a class in python called "Enzyme", however the following problem is not yet solved:
In BRENDA I have found the Kcat and KM values for ATP and glucose for hexokinase. In this bisubstrate reaction, how can I compute the reaction rate if I know enzyme, ATP and glucose concentrations? The problem I have is that I need to get Vmax from somewhere but for bisubstrate reactions I cannot compute Vmax from Kcat (because there are two).
Is computing Vmax as the enzyme concentration multiplied by the lowest Kcat an option?
Am I making a mistake by using Michaelis-Menten kinetics?
All help is welcome and appreciated :)
Relevant answer
Answer
kcat is determined from Vmax originally, so you can use the simple formula
Vmax= kcat[E]. Even though there are 2 substrates, there is only one Vmax.
To calculate the initial rate of the reaction at any particular pair of substrate concentrations, you need the rate equation. The proper rate equation to use for a bisubstrate enzyme depends on the kinetic mechanism. (In the case of hexokinase, there are also allosteric effectors to consider.)
I saw an old paper on rat skeletal muscle hexokinase
which gave the kinetic mechanism as Ordered Bi Bi with ATP binding first. I don't know if this is the right mechanism for the enzyme you are concerned with. If so, the kinetic rate equation for the initial rate in the forward direction is
v = Vmax[A][B]/{KiaKmB+KmA[B]+KmB[A]+[A][B]}
where [A] is [ATP] and [B] is [glucose]. You also need to know the kinetic constants Kia, KmA and KmB to calculate the rate, unless [A]>>KmA and [B]>>KmB. You may have KmA and KmB already, but you still need Kia.
I saw another paper (https://www.jbc.org/article/S0021-9258(19)42887-1/pdf) that gave the kinetic mechanism as Random Bi Bi. The initial rate equations for the two mechanisms are mathematically indistinguishable, however.
If you are not restricted to initial rate conditions, then you need to use the complete rate equation incorporating the products ADP and G6P, and all the kinetic constants associated with them. The equations are shown in the two papers, but it's unlikely you will have all the necessary values to use them unless you or someone else has made a very thorough study of your enzyme.
  • asked a question related to Enzymes
Question
1 answer
I want to test a list of peroxidases against a specific metabolite.
Ideally, I want to find an enzyme that will catalyze the reaction. But even a feeble peroxidase catalytic activity would be a great start.
Any tips are welcome!
Relevant answer
Answer
There are several computational tools available that can assist in predicting potential enzyme-metabolite pairings and their catalytic activities. One commonly used approach is molecular docking, which involves predicting the binding affinity and orientation of a metabolite within the active site of an enzyme. Tools like AutoDock, SwissDock, and DOCK are popular for performing such calculations. Additionally, machine learning algorithms trained on enzyme-metabolite interaction data can be used for prediction. Resources like EnzymeMiner and BRENDA offer comprehensive databases that can aid in identifying peroxidases and their potential substrates. However, it is important to note that these computational predictions are not always accurate, and experimental validation is crucial to confirm the catalytic activity of enzymes against specific metabolites.