Science topic

Cell Viability - Science topic

Explore the latest questions and answers in Cell Viability, and find Cell Viability experts.
Questions related to Cell Viability
  • asked a question related to Cell Viability
Question
3 answers
I am delivering propidium iodide (PI) to live cells through liposomes. I would like to incubate cells and liposomes over a period x of time. Say x is 12h. Delivery will be random and might be, in some cases, immediate. PI will thus be, in these cases, in these cells for 12h.
I would like to be able to image the PI inside the cell. However, PI is toxic to cells - how long can a cell last with (a certain concentration of?) PI, before it degrades and can no longer be imaged?
Thank you in advance for your help.
Relevant answer
Answer
Hey. I have a related question: Do you know WHY is PI toxic to cells? We have some weird behavior after minutes of exposure. Thanks in advance for your time!
  • asked a question related to Cell Viability
Question
1 answer
Hello. I am trying to test my designed peptide binding towards MDA-MB-231 and MCF-7 cell lines. However, I first need to dissolved my peptides as they are in lyophilized form.
The suggestion from my peptide synthesis service provider is to use 1 part ACN: 3 part water, which definitely is toxic to the cell lines. Alternatively, I can use DMSO as my solvent.
From my reading, concentration of >1% DMSO in my media would be cytotoxic to MDA-MB-231. I am now trying to run MTT assay to test the concentration that would be minimally cytotoxic to my cells. I am testing 1%, 0.5%, 0.25 %, 0.1% (v/v) DMSO in media.
However, I am curious does the concentration of this DMSO affect my peptide solubility in the solution? If let say I want to prepare 10uM concentration of my peptide, then if I am to prepare it by dissolving it in 100% DMSO, when i dilute the DMSO to 1%, wouldn't that also dilute my peptide? Or do I need to prepare higher than usual?
My concern is that my peptide sample is limited (10mg) per peptide so I don't want to use up whole sample as I have another assay to run.
Any advise on this? Thanks for the help!
Relevant answer
Answer
In order to avoid toxic effects of DMSO on cells, DMSO concentration should be ≤ 0.1% when it is finally added to the culture.
Suppose your working concentration that is finally added to the culture is 10uM, then you will have to prepare a stock of 10mM of your peptide dissolved in 100% DMSO. When you dilute your stock (10mM) 1000 times with culture media, you will obtain 10uM of working solution. At the same time, 100% DMSO will also get diluted 1000 times giving a concentration of 0.1%. So, you will have 10uM of peptide working solution in 0.1% DMSO when you finally add it to the cells.
If you have limited peptide sample, then you will have to run a small experiment to determine the level at which DMSO toxicity begins. Some cell lines can tolerate up to 1% DMSO without severe cytotoxicity. If MDA-MB-231 and MCF-7 cell lines can tolerate up to 1% DMSO, you may be able to save peptide sample for another assay because you will now be able to make a stock peptide solution of less than 10mM.
Best.
  • asked a question related to Cell Viability
Question
1 answer
Hello everyone,
I am currently working on a project involving primary cultures of breast cancer tissue samples, and I have a few questions regarding preservation:
How long can I safely preserve tissue samples at 4°C without compromising cell viability and functionality?
Are there specific preservation media or additives you recommend for maintaining tissue integrity during this period?
What methods or assays can I use to assess the viability of the cells after preservation?
I appreciate any insights or experiences you can share!
Thank you!
Relevant answer
Answer
Dear Amalia Kotsifaki,
How long can I safely preserve tissue samples at 4°C without compromising cell viability and functionality?
For best results, you may use the tissue samples within 24 hours. If you are unable to work on the tissues immediately, you may store the tissues in sterile culture medium at 4 degree C until use. You may successfully isolate cells from tissue samples stored at 4 degree C up to a maximum of 24-48 hours. Please note the longer the time from surgery to culture, the more the tisue samples are likely to deteriorate. Tissue samples used to generate primary cultures cannot be stored long-term, unlike the tissues used to extract RNA/DNA, which can be stored at -80 degree C using special preservatives.
Are there specific preservation media or additives you recommend for maintaining tissue integrity during this period?
There are no specific preservative media to maintain the integrity of the tissue samples when stored at 4 degree C except the sterile culture medium. Although most surgical specimens are sterile when removed, problems may arise with subsequent handling. Therefore, additives in the form of antibiotic/antimycotic should be added to the culture medium to prevent contamination. Wash the tissues thoroughly well in the medium containing antibiotic/antimycotic at the start of the procedure.
What methods or assays can I use to assess the viability of the cells after preservation?
You may apply simple method like cell counting using viability dyes such as trypan blue or Calcein-AM to check for the percentage of viable cells.
Best.
  • asked a question related to Cell Viability
Question
2 answers
I am have purified an bioactive compound (drug) from a plant. I need to perform an MTT assay to check cell viability and to determine IC 50 value.
After lyophilizing of HPLC fraction the compound is not visible and also according to the HPLC report the concentration of the compound is very less.
What should be my approach?
Relevant answer
Answer
the same procedure you can opt for your MTT assay
  • asked a question related to Cell Viability
Question
1 answer
When IC50 cannot be obtained according to the cell viability chart, what should be the dose selection under cell culture conditions? Would it be beneficial to go with sham control?
For example, for this chart 1) control vs. 0.3 mg/ml, or 2) 0.0025 mg/ml (sham) vs. 0.05mg/ml. Which one should be preferred?
What reference can I give to base this on literature and how should I explain it academically?
Thank you in advance for your help.
Relevant answer
Answer
To my opinion, the product under investigation is not cytotoxic in the range of concentrations tested. There is no real answer to your question.
  • asked a question related to Cell Viability
Question
1 answer
If I wanted to do cell viability using mts assay, the treatment need to be diluted with complete media or basal media? What happen if I used complete media to dilute the treatmemt and do cell viability test?
Relevant answer
Dear Melonney,
I recommend diluting in complete media (appropriate culture medium + 10% FBS or other serum). Reducing the concentration of serum during the treatment is an additional factor that might influence cell viability. Remember also what the diluent of your treatment is. If it is DMSO, do not exceed the final concentration of DMSO of 0.1%. Good luck!
  • asked a question related to Cell Viability
Question
2 answers
I intend to analyze some aspects of cellular immune response after vaccination on those who didn't seroconvert, comparing with a control group of people who seroconverted, and at different periods (including pre-vaccination). Since I can't tell who will or will not seroconvert, I'll have to collect and keep blood samples from everybody pre-vaccination and later select and analyze those who didn't seroconvert (which are the minority). Given that isolation of PBMC with a density gradient is a time consuming procedure and I'll have numerous samples, I wonder if I could just separate and freeze the buffy coat from all samples. Later on when I get the serology results, I could isolate PBMC from the thawed buffy coat with Ficoll only from selected samples. I know cell viability will probably be lower compared with that from fresh blood. Does anyone have this experience? Can anyone help me by giving some advice? Thanks in advance! 😊
Relevant answer
Answer
Yes, you can. PBMCs from frozen buffy coats would become necessary when there are studies with elevated number of participants and for sample collections in multi-center studies.
You may follow the below protocol.
1. Centrifuge the collected blood at 2000 x g for 15 mins at 4 deg C in a swinging bucket rotor, switching-off the centrifuge brakes.
2. Collect the cloudy buffy coat (the layer between the upper plasma phase and the lower phase containing most of the erythrocytes), containing the enriched PBMC fraction into a cryovial tube.
3. Resuspend the buffy coat in freezing medium containing 90% FBS and 10% DMSO.
4. Freeze the vial in a controlled rate freezing container at -80-degree C. For longer storage, you may store cells in liquid nitrogen.
5. During thawing of buffy coat, add the same volume of PBS to the thawed buffy coat.
6. The diluted buffy coat is gently layered on Ficoll (centrifuged at 3000 rpm, 20 minutes, refrigerated centrifuge, swing rotor, break off).
7. Collect the PBMC layer.
8. PBMCs are then washed with 5 ml PBS two times.
Best.
  • asked a question related to Cell Viability
Question
5 answers
I was performing an MTT assay to check cell viability. Due to a technical issue in the plate reader I was using, I had to read the plate at 540 nm wavelength. Would the results still be credible?
Relevant answer
Answer
Hello Bahar Ksh
If you have to read the plate at 540 nm wavelength, use DMSO as the solubilizing solution.
If you are using acidic isopropanol as the solubilizing solvent, the absorption maximum is at 570 nm. But for DMSO as the solubilizing agent, the absorption maximum shifts to 540 nm.
Best.
  • asked a question related to Cell Viability
Question
2 answers
I need to perform an MTT assay on MDA-MB-231 cells to determine cell viability. I am using free docetaxel (DTX) as my positive control for my MTT assay and my supervisor has asked me to dissolve my DTX in DMSO. However, it has been specified that my vehicle for the positive control as well as the positive control itself cannot contain more than 0.1% DMSO otherwise it will be too cytotoxic and we won't know whether it is my treatments killing the cells, or the DMSO.
I need to work with concentrations 10, 20, 30, 40, 50, 60, 70, 80 and 100µg/mL of DTX to treat my cells with. Therefore, how do I create a stock as well as serial dilutions of my DTX in DMSO and DMEM media such that the final concentration of DMSO that I add in the wells is less than 0.1%?
Relevant answer
Answer
Hello Lauren,
To prepare a series of dilution of docetaxel (DTX), you may prepare a stock of 100mg/ml DTX in 100% DMSO.
For the concentrations you have mentioned, you may go about as follows.
For 10ug/ml (0.01mg/ml): Dilute 100mg/ml stock 1:10000 times using DMEM which means DMSO will be diluted 10000 times to give a concentration of 0.01%.
Similarly, for 20ug/ml (0.02mg/ml): Dilute 100mg/ml stock 1:5000 times using DMEM which means DMSO will be diluted 5000 times to give a concentration of 0.02%.
For 30ug/ml (0.03mg/ml): Dilute 100mg/ml stock 1:3333 times using DMEM which means DMSO will be diluted 3333 times to give a concentration of 0.03%.
For 40ug/ml (0.04mg/ml): Dilute 100mg/ml stock 1:2500 times using DMEM which means DMSO will be diluted 2500 times to give a concentration of 0.04%.
For 50ug/ml (0.05mg/ml): Dilute 100mg/ml stock 1:2000 times using DMEM which means DMSO will be diluted 2000 times to give a concentration of 0.05%.
For 60ug/ml (0.06mg/ml): Dilute 100mg/ml stock 1:1666 times using DMEM which means DMSO will be dilute 1666 times to give a concentration of 0.06%.
For 70ug/ml (0.07mg/ml): Dilute 100mg/ml stock 1:1428 times using DMEM which means DMSO will be dilute 1428 times to give a concentration of 0.07%.
For 80ug/ml (0.08mg/ml): Dilute 100mg/ml stock 1:1250 times using DMEM which means DMSO will be diluted 1250 times to give a concentration of 0.08%.
For 100ug/ml (0.1mg/ml): Dilute 100mg/ml stock 1:1000 times using DMEM which means DMSO will be diluted 1000 times to give a concentration of 0.1%.
So, for all concentrations, you will have DMSO concentration less than 0.1% except 100ug/ml, which will have DMSO concentration equal to 0.1%.
You should have a vehicle control for each dilution. For instance, for 10ug/ml working concentration, you should have a vehicle control containing 0.01% DMSO only. Similarly, for other concentrations you may do the same.
In a similar manner as above, you may prepare the positive control in DMSO containing less than and equal to 0.1% DMSO.
Best.
  • asked a question related to Cell Viability
Question
1 answer
Hi Everyone
First year PhD student in London here, I'm new to Caco-2 cells.
I have two issues that are bugging me. Does anyone have any advice on how I can navigate these problems?
The first one, as shown in the first image, where cells seem to clump together in a wall like structure, and there are cells growing on one side but none on the other side of this 'wall'. This is super annoying because I seed the cells and let them grow for a week, only to find that the cells on one side of the plate have died or disappeared so I essentially waste a week and media.
The second one, are the black rod like structures bacteria? I'm treating my cells with media in the absence of antibiotics for a gentamicin protection assay for 24 hours but normally, I do use pen-strep 1%. Cell viability in the potentially infected regions appears to be severely limited. However, the media is not cloudy?!
Thanks so much!
Pranaya
Relevant answer
Answer
Hello,
Did you solved your problems ? Because I have the same issues when I grow my caco2 in 96 plates ! I find out when I change the media and I delivered the media too "strong" the cell mat take-off.. Beside the facts that I have to be more gentle I think there is an adherence problem...
  • asked a question related to Cell Viability
Question
2 answers
I am currently testing the effect of bacterial filtrates on cancer cells , after seeding the cells I tested the bacterial filtrates against them , and got images of all 96 wells using inverted microscope. How can I calculate the % of viability without staining the cells or doing MTT ?
i would appreciate any help
Relevant answer
Answer
Thank you for replying Mr.Jianfeng Lin ,
Adding live/dead staining is a good idea , but I already have the images after 24 h incubation that’s why I was looking for a method to do the counting without any staining . After incubating cancer cells with bacterial filtrates their morphology changes to get round-up and circular .
XTT is a good suggestion though would you mind recommending some publications related to this assay ?
  • asked a question related to Cell Viability
Question
2 answers
Hello, I am currently conducting research on ovarian tumour cells and the use of chemotherapies with dietary metabolites and their combinations to see if there is a difference in cell viability. I have conducted tumour cell assays and have formed bar graphs with the normalised data so the x axis is the therapy concentration e.g pemetrexed at 100nM, 1um, 10um and 100um and the y axis is cell viability (%). I am unsure on how to analyse this data, I have chosen ordinary one-way ANOVA as the independent groups could be the different treatments with different doses to see if there is a difference in cell viability. I have run the tests and they seemed to have work but I have no actual understanding of the results and I also used one sample t-test, which again has given me results but I am not too sure what is showing. I got told that it isn’t clear what comparisons I am making – a given observation must be significantly changed compared to something else – but what? Any advice on what statistical analysis I should be using?
Relevant answer
Answer
In a nutshell, 'one-way' or 'two-way' ANOVA means the number of factors in your experiment. One-way: one factor. Two-way: two factors. A factor means the variable, which also means the factor you are actively changing, in this case, your dose. Cell viability is the outcome, which you are not actively changing.
I haven't really done much of t-test, but from my understanding it can only compare 2 treatment doses... although it seems that results still show up for you. I agree with the recommendations Muhammad Ilyas suggested, that one-way ANOVA seems to be more appropriate for your experiment, because it can compare multiple (>2) doses.
If you want to know more, try reading the prism statistics guides. They provide pretty neat explanations on how to choose the right statistical test. Here is a link about one-way ANOVA, where you can read the session titled 'Is there only one factor?' They also talk about t-tests and two-way ANOVA elsewhere.
Hope this helps!
  • asked a question related to Cell Viability
Question
3 answers
Hello,
My E. coli cells express both green fluorescent protein as well as mCherry. So I need a fluorescent stain of color other than green and red fluorescence to enumerate their viability. Please suggest. Thanks in advance.
Relevant answer
Answer
May consider using Permai fluorescence dye.
  • asked a question related to Cell Viability
Question
3 answers
the best cell lines for myeloid leukaemia for transfection
Relevant answer
Answer
Thanks Bill Chi Shun Ho
What about Kg1a?
  • asked a question related to Cell Viability
Question
1 answer
Dear colleagues,
I need to transfect a 10 kb plasmid into Caski cells and am hoping to use Lipofectamine 3000 for the transfection. If anyone has experience using Lipofectamine 3000 specifically in Caski cells, I would greatly appreciate if you could share your transfection protocol. Specifically, tips on:
Ideal plasmid:Lipofectamine 3000 ratio
Optimal cell seeding density
Lipofectamine reagent dilution and complexation steps
I want to achieve high transfection efficiency while maintaining good cell viability. Please let me know the optimal conditions and procedures you’ve used for transfecting plasmids into Caski cells via Lipofectamine 3000. Thank you so much for taking the time to share your experience!
Relevant answer
Answer
Have you done it, if yes please let me know the protocol. Thanks
  • asked a question related to Cell Viability
Question
3 answers
MTT assay
Relevant answer
Answer
Hi Dr. Zunika Amit
I agree with Dr Anton Lennikov .
There are many compounds which show increase in the cell proliferation above the control cells. This increase in proliferation at the optimal drug concentration sometimes leads to increase in the cell number more than the control number. In this case you will observe the % cell viability of the treated cells above 100% which is quite common in many experiments. I have observed similar effects in compounds against highly proliferative cells such as RAW 264.7, N9 microglia, HMC-3 and many other cells.
Hope it helps,
Thanks,
  • asked a question related to Cell Viability
Question
2 answers
Why does a negative or a 0 for blank mean? Does this mean that cell viability has stopped?
Relevant answer
Answer
Hey there Maya Osman,
Dealing with Alamar Blue assays can sometimes throw us curveballs, right? Here's the deal: getting a negative or zero value for the blank in your Maya Osman Alamar Blue assay doesn't necessarily spell doom for your Maya Osman cell viability.
First off, let's understand what the blank represents. It's essentially your Maya Osman baseline, the control that gives you Maya Osman a reference point for comparison with your Maya Osman experimental samples. So, when you're Maya Osman hitting a negative or zero, it's indicating that the background absorbance or fluorescence is essentially negligible.
Now, does this mean your Maya Osman cells have completely given up the ghost? Not necessarily. What it could indicate is that either there was an issue with the reagents or the assay setup, or your Maya Osman samples just happen to be super clean with minimal background interference.
However, it's always wise to double-check your procedure, make sure your Maya Osman reagents are fresh and properly handled, and perhaps consider running a positive control to validate your Maya Osman assay setup. Sometimes, a negative or zero blank can indeed signal a problem, but it's not an automatic death knell for your Maya Osman cell viability assessment.
Stay vigilant, keep experimenting, and let's crack this puzzle together! Cheers!
  • asked a question related to Cell Viability
Question
2 answers
I am working with a formulation of calcium phosphate bone cement reinforced with graphene, and I will perform an MTT assay to evaluate the cell viability of this material. However, I need to know which bone cell type is most commonly used for this assay. Thank you in advance.
Relevant answer
Answer
Malcolm Nobre Thank you very much for the information!
  • asked a question related to Cell Viability
Question
2 answers
Hello,
I am Mahmuda, now I am working with DG44 cell culture. So far, my cell culture viability improved to 90%. However, before transfection, my cell viability decreased to 70%. I am very disappointed with the results of this culture.
Is there any suggestion or input that can be given so that I can solve this problem?
Then, is there any particular trick to do for this DG44 culture?
Thank you for the help
Relevant answer
Answer
DG44 cells are a type of Chinese hamster ovary (CHO) cell line commonly used in biopharmaceutical research and production, particularly for the production of recombinant proteins and monoclonal antibodies. Several factors can affect the viability of DG44 cells, including:
  1. Culture Medium and Nutrients: The composition of the culture medium and the availability of essential nutrients are critical for cell viability. Ensure that the medium is properly formulated with appropriate concentrations of glucose, amino acids, vitamins, salts, and growth factors necessary for cell growth and metabolism.
  2. pH and Buffering Capacity: Maintaining the pH of the culture medium within the optimal range is crucial for cell viability. Fluctuations in pH can adversely affect cell growth and metabolism. Additionally, ensure that the culture medium has adequate buffering capacity to resist pH changes over time.
  3. Temperature: DG44 cells are typically cultured at 37°C in a CO2 incubator. Maintaining the proper temperature is essential for cell viability. Fluctuations in temperature can affect cellular processes and compromise cell health.
  4. Osmolality: The osmolality of the culture medium should be maintained within the physiological range to prevent osmotic stress on the cells. Hypo- or hyperosmotic conditions can lead to cell swelling or shrinkage, respectively, affecting cell viability.
  5. CO2 Levels: DG44 cells are typically cultured in a humidified atmosphere containing 5% CO2. CO2 is necessary for buffering the culture medium and maintaining the optimal pH for cell growth. Ensure that the CO2 levels are properly regulated to support cell viability.
  6. Cell Density and Confluence: Cell density and confluence in the culture vessel can affect cell viability. Overcrowding can lead to nutrient depletion, waste accumulation, and reduced viability. Conversely, low cell density may result in suboptimal growth and viability.
  7. Cell Passage and Subculture: Proper handling during cell passage and subculture is essential to maintain cell viability. Avoid over-trypsinization or excessive mechanical stress during cell dissociation, as it can damage the cells and decrease viability.
  8. Contamination: Contamination with bacteria, fungi, mycoplasma, or other microorganisms can compromise cell viability. Follow strict aseptic techniques and regularly monitor cultures for signs of contamination.
  9. Cell Line Health and Stability: The genetic stability and health of the DG44 cell line can impact cell viability over time. Monitor cell morphology, growth rate, and productivity regularly to ensure consistent performance.
By carefully optimizing these factors and maintaining appropriate culture conditions, you can enhance the viability and robustness of DG44 cell cultures for various applications in biopharmaceutical research and production.
  • asked a question related to Cell Viability
Question
1 answer
I have WST-8 powder, how to use it to prepare CCK8 solution for cell viability testing
Relevant answer
  • asked a question related to Cell Viability
Question
4 answers
I am new to using MTT assay. My experiment is to treat cells and see the cell viability after 24,48,72 hrs so should I grow cells in three different plates and put MTT reagent for each one separately or there is a way to read every 24 hrs until I finish the experiment?
Relevant answer
Answer
You should grow cells in three different 96-well plates. Once the cells have reached 80% confluency, you remove the growth media and add media containing various concentration of the candidate compound/drug and incubate all the three plates in the incubator at 37 degree C and 5%CO2.
After 24 hours, you may perform the MTT assay on the plate marked as 24 hours. Calculate the IC50 value for 24 hours. Then after 48hours, perform the MTT assay on the plate marked as 48 hours and calculate the IC50 value for 48 hours. Similarly, after 72 hours, perform MTT assay on the plate marked as 72 hours and calculate the IC50 value for 72 hours.
So, you grow cells in three different plates and put MTT reagent for each one separately at three different time points.
Best.
  • asked a question related to Cell Viability
Question
2 answers
Hello everyone!
MTT was performed after applying DEHP to HeLa cells. According to MTT results (24 hours of exposure), cell viability is significantly reduced at concentrations of 100 micromolar and above. In the 1-30 micromolar range, viability is almost the same. Next, I will check SOD, CAT, GPX levels with ELISA. What concentrations should I use in ELISA? Should I include IC50, IC30 or IC20?
I would be glad if you help me.
Thanks in advance!
Relevant answer
Answer
GPX requires a source of GSH to function, plus you don't want the external environment to become depleted in thiols and rich in disulphides. Therefore I would include G6P, G6PDH, GSH and glutathione reductase for the GPX to function for any extended period of time. SOD I personally question it's importance and the 'toxicity' of superoxide itself (see link)
Catalase functions well just added to the media, but works better with low concentrations of H2O2 when it can function as a peroxidase. Other oxidants of concern include Fe(IV) species.
In my experience many cells exhibit a threshold response, in terms of the observed toxicity, when ROS production (could use systems such as Xanthine/Xanthine oxidase, or glucose/glucose oxidase) exceeds ROS removal rates by the cells. This phenomena also observed by others.
Good luck.
  • asked a question related to Cell Viability
Question
6 answers
Our CO2 supply is outside in a cage and there is fairly long distance pipework going around the outside of the wall to the entry point in the wall of the lab. Since turning late Autumn/early winter, some of our cells are starting to look a bit odd. It seems to get worse the colder it gets. The incubators are reading 5% CO2 (so unlikely a leak), the temperature is correct too. They are newish incubators, only serviced recently (we are getting our own CO2 meter to check soon too). All reagents were replaced (several times). Two different people have had the same problem, so not user error either (both experienced users). Different batches of cells have been tried too. Its the first time Ive ever used a supply from outside, (its usually next to the incubator) so I was wondering if it had an effect on anything as Im running out of ideas. Many thanks
Relevant answer
Answer
Bill Chi Shun Ho Thank you.
  • asked a question related to Cell Viability
Question
4 answers
I am trying to thaw S2R+ cells from the frozen culture. The cells (1 ml of cell culture) were frozen at passage 9 and stored at -80. I followed the steps below to thaw the cells from the frozen stock:
1. Take the vial with the cells and thaw it at 30 degrees in a water bath.
2. As soon as the culture is thawed and liquid, remove the vial from the water bath, and clean it using 70% ethanol. Take the 1 ml cell culture from the vial and add it to a 10 ml falcon containing 4 ml of complete Schneiders media (Complete S2 Media: 10% FBS, 1% Pen-Strep, 89% Schnieders Medium). Mix them nicely with pipetting.
3. Place the falcon, now with 5 ml components (1 ml of culture from stock and 4 ml fresh media) in a centrifuge at 100g for 10 min.
4. Remove the supernatant, which would contain DMSO, and then add fresh 5 ml of media (2.5 Fresh S2 Complete Media + 2.5 Conditioned S2 Media) and add this culture to the T-25 flask and let it incubate for 3-5 days before starting passaging.
I have attached the images (phase contrast images), which were taken after P10 (one passage after the above procedure of thawing and reculturing the stock). I see a lot of dead cells (Counted using Luna Cell Counter--> Viability is approximately 40%).
Am I doing something wrong while I reculture the frozen stock?? Or is it alright and I should just clear out the dead cells using low centrifugation speed for 10 minutes or so?
Relevant answer
Answer
As other mentioned, cryopreserved cells should be stored in vapor phase of liquid nitrogen or below -125°C. It may be too late to response to OP, but the cells look fine. The S2 cells can be grown as adherent or suspension in S2 medium containing 10% heat-inactivated FBS. Do not use antibiotics for the first 24-48 hours as it would affect the recovery of the cells after thawing. They love high cell density in culture. Make sure you keep the conditioned medium for cryostorage and future use. For passaging, I usually just dilute the cells a few folds with fresh medium and keep ~20% conditioned medium.
  • asked a question related to Cell Viability
Question
1 answer
I am combining drug A (IC20 and IC50) with drug B (5 uM, 10 uM and 50 uM). Drug B reduces cell viability to 28% at 50 uM, while drug A IC20 combined with drug B 50 uM reduces cell viability to 32%. CompuSyn gives a CI=0.46. Is this correct? To have synergism shouldn't the viability be reduced to less than 28% when combining the 2 drugs at these concentrations?
Thanks for any help.
Relevant answer
Answer
You can use the combination index, which is a quantitative measure of drug interaction. It is calculated using the formula
CI = (D1/EDx1) + (D2/EDx2),
where D1 and D2 are the doses of drugs 1 and 2, and
EDx1 and EDx2 are the doses that produce x% effect individually.
A CI value less than 1 indicates synergism.
  • asked a question related to Cell Viability
Question
2 answers
Im evaluating cell viability in flow cytometry with propidium yordurate and I need to keep my cells alive in a solvent that can keep them up to 1 hr and can also be used to be injected in the flow cytometry.
Relevant answer
Answer
Hello Lucas,
Use HBSS (Hanks' Balanced Salt Solution) with calcium and magnesium, which is a balanced salt solution with trace amounts of glucose.
Cell viability is not compromised while cells are in HBSS since it is a balanced salt solution with glucose and it maintains osmolality and physiological pH. Cells will remain relatively happy and survive for at least 2 hours in HBSS.
So, you may use HBSS instead of PBS, and HBSS can also be used in flow cytometry.
Best.
  • asked a question related to Cell Viability
Question
2 answers
Hi,
I am curious to know what is the acceptable cytotoxicity levels for a drug/compound tested using MTT assay in vitro?
Relevant answer
Answer
In examining the effect of a compound or medicine, a concentration of a substance in which 50% of cells are alive and 50% are dead is called IC50.
  • asked a question related to Cell Viability
Question
4 answers
Hi guys!
Lentiviral CRISPR-Cas9 targeting guide RNA (gRNA) expressing vector is the most common method to generate a specific gene knockout cell line. However, during my recent research work, I found the RAW264.7 cell line quite difficult to be transduced by Lentivirus. Meanwhile, after transduction, RAW264.7 gets activated and polarized with poorer cell viability and slower proliferative rate, in which case it becomes tough to conduct the further study on it. As in the same situation for immortalized bone marrow-derived macrophages (iBMDMs). In the methods of some classic reseaches about RAW264.7 or iBMDMs, most researchers utilize Lentiviral transdution to fulfill genome editing. How can they avoid the abovementioned problems(poorer cell viability and slower proliferative rate after lentivirus transduction)?
I'm pretty new to this, so any help, tips, advice, or direction would be very helpful. Thank you in advance for any suggestions.
Relevant answer
Answer
@Aisyah Jaafar,Hello, we have tried many methods and finally succeeded with CRISPR/Cas9 and lentiviral vector to infect RAW264.7 cells.
  • asked a question related to Cell Viability
Question
1 answer
If we treat NHDF cells with ascobic acid for 1 hour at 33ug/mL and then irradiate the cells with a low dose of UVA we see a good antioxidant response and cell viability does not change compared to non-irradiated NHDF cells. However, if we followed this procedure but with a incubation of ascorbic acid for 24 hours, after irradiation we were unable to detect an antioxidant effect and we also observed an increase in cell viability.
We know that ascorbic acid can reduce our cell viability reagent, but we do not know why its antioxidant effect and viability depend on its incubation time. Do you have any idea?
Relevant answer
Answer
How do You measure antioxidant effect? How it differs (InYourHumbleOpinion) from cell "viability". What form of ascorbic acid are You using (cat. nr)?
  • asked a question related to Cell Viability
Question
2 answers
Hi everyone,
We are recently doing cell viability and for that we use Alamar Blue. The results after ascorbic acid treatment and UVA-irradiation show increase cell viability for cells that were treated with acid ascorbic. We think that there is no change of cell viability, that this increasing is due to increse in metabolic activity of those cells. Could it be possible? Do you have experience with a treatment that increse metabolic activity? There is some way to only see cell viability with Alamar Blue?
Thanks in advance,
Relevant answer
Answer
Thank you very much, we will try it!
  • asked a question related to Cell Viability
Question
1 answer
I have a compound (C23N3OH27) to repeat some results with a molecular weight of 361.48. The problem is that the results are not being the same, I am evaluating cell viability (K562 and KG1) with resazurin (24 hours of plating 20.000 cells/100uL, 24 hours of treatment 100uL, 4 hours of resazurin 20uL) and the results lead us to believe that it does not induce death in any of the cases. concentrations tested (30 uM, 20uM, 10uM, 5uM, 1uM), I have already evaluated cellular metabolism, resazurin, interaction of the compound with resazurin and none explains the reason for not repeating the results. I am suspicious that it could be my dilution, I used a table from a colleague that performs the calculation automatically. Could someone help me to do the dilution directly just so I can assess if it's correct? I have 5g powder of the compound which was diluted in 2305.34uL of 100% DMSO, which according to the table gave me a solution of 6,000uM, I don't know if that's correct.
obs: my controls (+/-) are responding well so I don't believe it's the resazurin or the plating
Thanks for all contributions!
I have attached the dilution table below.
Relevant answer
Answer
Sorry! I did not understand the calculations from the excel sheet as it is very complicated.
Could someone help me to do the dilution directly?”
Yes, let me make it simple.
The Molecular weight of the compound (C23N3OH27) is 361.48.
Then follow the sequence below.
361.48g -------- 1L -------- 1M
361.48g --------- 1L ------- 1000mM
0.36148g ---------- 1L ------ 1mM
361.48mg -------- 1000ml ------ 1mM
3.614 mg ----------- 10ml -------- 1mM
So, weigh 3.614mg of the compound in 10ml 100% DMSO to give 1mM stock.
You may prepare working solutions (30uM, 20uM, 10uM, 5uM, 1uM) as follows.
You may use the formula: C1V1=C2V2
C1= Concentration of stock solution (1mM)
V1= Volume of stock solution (X)
C2= Concentration of working solution (30uM)
V2= Volume of working solution (say 1ml)
Then,
1mM x X = 30uM x 1ml
1000uM x X = 30uM x 1ml
30/1000 = 0.03ml of stock i.e., add 30ul of stock solution to 970ul of media to give 1ml of 30uM working solution.
Similarly,
For 20uM
20/1000 = 0.02 ml of stock i.e., add 20ul of stock solution to 980ul of media to give 1ml of 20uM working solution.
For 10uM
10/1000= 0.01ml of stock i.e., add 10ul of stock solution to 990ul of media to give 1ml of 10uM working solution.
For 5uM
5/1000 = 0.005ml of stock i.e., add 5ul of stock solution to 995ul of media to give 1ml of 5uM working solution.
For 1uM
1/1000= 0.001ml of stock i.e., add 1ul of stock solution to 999ul of media to give 1ml of 1uM working solution.
Since 1ul is a very minute quantity to pipette, it may lead to error. So, you may dilute the stock by 1:10 to make a diluted stock (0.1mM). Then take 10ul of diluted stock (0.1mM) and add to 990ul of media to obtain 1uM working solution. Use this calculation for 1uM working solution instead of the above.
Best.
  • asked a question related to Cell Viability
Question
1 answer
I am writing to report a concerning issue I've encountered during my recent migration assay using RAW 264.7 cells. I have followed the standard protocol, but I am facing a serious challenge with cell viability and scratch healing in response to CXCL9 treatment.
Here is a detailed description of the experiment:
Cell Line: RAW 264.7 cells
Media: DMEM Free Serum (used to standardize the results, Raw cells 264.7 grow in DMEM with 10%)
Scratch Assay Setup:
Cells were plated in 12-well plates until they reached confluence.
Media was removed, and cells were washed with PBS.
A scratch was made using a 200μl tip.
Cells were washed again with PBS.
Fresh DMEM Free Serum media containing varying doses of CXCL9 cytokines was added.
The issue I'm facing is that after the addition of CXCL9, the scratch disappears quickly and cells seem to detach and float, indicating a loss of cell viability.
I would like to inquire whether I am following the correct protocol, if a reduced FBS media is preferable over serum-free media, or if you have any alternative suggestions for conducting the migration assay with RAW cells.
Relevant answer
Answer
I have a feeling you are not following the right protocol. Maybe the protocol provided below will help.
1. After the cells are 90-95% confluent, remove the spent media and add 1ml of 5 µg/mL of mitomycin C (prepared in culture medium) to each well. Mitomycin C will stop proliferation of cells for 2-5 h before scratching. So, in a way, it will help to ensure true detection of migration. Incubate the plate for 2 h at 37 °C and at 5% CO2.
2. Remove the medium with mitomycin C and wash the wells once with 1ml of 1X PBS.
3. Add 1ml of 1X PBS and manually scratch the wells vertically with a 200µl yellow pipette tip. Use a new pipette tip for each well.
4. Remove the PBS and then add the media containing varying doses of CXCL9. Then proceed as usual with the assay.
It is important that you stop the proliferation of cells. Without mitomycin C treatment, I feel the proliferating cells may be occupying the area created by the scratch and due to over confluency, the cells may be detaching from the substratum and floating.
Best.
  • asked a question related to Cell Viability
Question
2 answers
Usually neuroblastoma have low transfection efficacy (5-10%) when transfected with Lipofectamine 3000. So, this time I have tried electroporation with this condition,
poring pulse: 175V, 2.5ms, 50ms interval, 2 pulses, 10% decay rate, + polarity
transfer pulse: 20V, 50ms, 50ms interval, 5 pulses, 40% decay rate, +/- polarity
I have used this transfection condition with three seeding density (80K, 100K and 150K)in 24 well plate.
However, the cell viability is very low for RNA collection after 24 hour of electroporation.
Could anyone please suggest me how to improve the cell viability?
TIA.
Regards
Ayman
Relevant answer
Answer
Dear A.B Bayazid
Thank you for your reply.
The mentioned poring and transfer pulse conditions are used in my lab after optimization in case of SHSY5Y.
However, I was not getting enough cells may be because of technical errors as I am doing this first time.
So, following some literature protocols, after electroporation, I have seeded 1M cells in 6 well plate and 24 hour later transfer them to the 24 well plate (100K cells/ well) and collect cell lysate the next day.
In this way, I found around 80% confluency which is enough for RNA extraction.
Take care
Best wishes
Ayman
  • asked a question related to Cell Viability
Question
6 answers
I have used the MTT Assay to measure cancer cells' viability under an antioxidant compound's influence. But contrary to expectations, with the increase of antioxidant concentration from 5 μM to 150 μM, the viability not only did not decrease but also increased. In other words, with the increase in the concentration, the amount of light absorption increased. In your opinion, what is the reason for this technical error? Or what kind of problems could have occurred during the MTT Assay?
Relevant answer
Answer
if you are looking at viability, did you use any other methods? mtt depends on mitochondria. and you are using antioxidants.
  • asked a question related to Cell Viability
Question
2 answers
Hello to all,
I am working on SH-SY5Y cells and I treated my cells with Rsl3 and ferostatin-1 (24 hours) but I could not see good cell viability. I would like to know how I can treat my cell so that fer-1 can prevent cell death caused by Rsl3.
With thanks for your attention
Parisa
Relevant answer
Firstly, I'd suggest ensuring that you're using appropriate concentrations of both RSL3 and Ferrostatin-1. The concentrations required can vary depending on the specifics of your experimental setup, including cell density and the duration of exposure. In many cases, concentrations are determined through a process of titration, starting at lower concentrations and slowly increasing until the desired effect is achieved.
  1. RSL3: This is an inducer of ferroptosis. Concentrations of RSL3 typically used in research range from 0.1 to 10 µM, depending on the specific cell line and the duration of the treatment.
  2. Ferrostatin-1 (Fer-1): This is a potent inhibitor of ferroptosis. The concentrations used can vary widely, from 0.1 µM to 20 µM, again depending on the specific cell line and duration of treatment.
Here's a general process to follow:
  1. Pretreat with Fer-1: To maximize the protective effects of Ferrostatin-1, you could pretreat your cells with Fer-1 for a certain time period (say 2 hours) before adding RSL3.
  2. Co-treatment with RSL3 and Fer-1: After the pretreatment period, add RSL3 while maintaining the presence of Fer-1 in the culture medium.
  3. Post-treatment with Fer-1: Continue to treat the cells with Fer-1 after removing RSL3 to ensure that any late-developing ferroptosis is also inhibited.
Remember to keep in mind the half-life of Fer-1, which is approximately 2 hours in cell culture medium, so repeated administration may be necessary for longer experiments.
Monitor cell viability using an appropriate assay such as trypan blue exclusion or MTT, and adjust concentrations as necessary. Note that while Fer-1 can prevent RSL3-induced cell death, it may not restore cell viability to 100%, as some cell death may still occur.
  • asked a question related to Cell Viability
Question
3 answers
I have done a number of MTT assay's for a study I am a part of, twice the results showed a very low viability and this crystal formation. I did a later assay following the same procedure, using the same cell line, and same MTT with viable results. What would cause this? Could the MTT mixed for that trial being exposed to light have anything to do with it?
Relevant answer
Answer
I would think the MTT is reduced (by chemically or enzymatic processes in cells) to yield insoluble formazan. The formazan likely will not precipitate immediately, and thus much of the formazan likely will form small purple crystals in the external media. I think these particles form the nucleus around which buffer components are crystallizing. Depending on the pH, buffer components such as EDTA, and phosphate salts readily crystallize.
  • asked a question related to Cell Viability
Question
4 answers
I isolated and differentiated bone marrow-derived macrophages from mice. After differentiation I frozen them and kept them in liquid nitrogen. Then, I thaw them and let them recover for 3-5 days before polarization.
They proliferate perfectly with the differentiation medium (DMEM + 20% FBS + 30% L929 SN), and I can keep them in culture for, at least, 2 weeks after thawing. To induce polarization I split the cells, count and seed them at a density of 100-150,000 cells/cm2 in classical medium (DMEM + 10% FBS). I culture them for 24h with this medium, after I change the medium to the one with the specific cytokines to induce anti- and pro-inflammatory polarization (IL-10 and protein homogenate from injured muscle, respectively) for 48h. After 72h from seeding, I get a viability of 10-15%, which is too low.
I have several questions for which I haven't found a clear answer yet.
  • Can the incubation of 24h with classical medium before polarization can affect BMDM viability? Should I reduce this time?
  • Is it possible to induce anti- and pro-inflammatory polarization with the presence of M-CSF (L929 SN)? I have read that the M-CSF promotes and anti-inflammatory phenotype. Would it be possible to induce a pro-inflammatory state even with the presence of M-CSF)
I attach the photos from the BMDM after 24h of seeding with differentiation or classical medium.
Hope you can help me!
Thanks :)
Relevant answer
Answer
1-It does not affect the polarization property of macrophages. The incubation of 24 hours with the classical medium before polarization does not negatively impact BMDM viability or alter their ability to undergo polarization. This incubation time is commonly used to allow macrophages to adhere and recover from any stresses induced during isolation. As a result, it does not interfere with the subsequent polarization process and allows the cells to maintain their normal function during polarization.
2-M-CSF (Macrophage Colony-Stimulating Factor) plays a critical role in promoting the differentiation of monocytes into macrophages and is involved in developing macrophages with an anti-inflammatory or M2 phenotype. However, it is essential to note that M-CSF alone typically results in a basal level of anti-inflammatory properties in macrophages, and it does not induce a robust pro-inflammatory response.
To induce a pro-inflammatory state in macrophages, it is common to use other stimuli, such as pro-inflammatory cytokines like interferon-gamma (IFN-gamma) and lipopolysaccharide (LPS). These stimuli promote the polarization of macrophages into the M1 or pro-inflammatory phenotype, characterized by the secretion of pro-inflammatory cytokines like TNF-alpha, IL-1beta, and IL-6. Add other factors like IL-4 and IL-13 along with M-CSF to induce a pro-resolving or anti-inflammatory phenotype.
  • asked a question related to Cell Viability
Question
1 answer
I'm doing primary tumor spheroids to a drug screening. We're having success to make the spheroids but I'm not having succes to dissociate the spheroids to evaluate the cell viability (using trypan blue to count).
I tryed to use trypsin 5 and 10 min (37ºC) and Accutase 10 min (R.T.) and the spheroids not dissociated completely. Could you help me?
p.s.: Spheroids were plated with 5000 cells, so they are ver small to dissociate mecanically.
Thank you!
Relevant answer
Answer
Collagenase at +4C
  • asked a question related to Cell Viability
Question
1 answer
I checked the literature in detail to compare different cell viability, cytotoxicity studies to check the biocompatibility of biomaterials. In some articles, they use the 'leachable-conditioned medium' method to check the biocompatibility. For this purpose first, they incubate biomaterials in cell culture medium without FBS after that they use this 'conditioned medium' with FBS in cell seeding. I am wondering in this first step why they don't add FBS directly to cell culture media and is it important for this procedure. I also saw different methods including FBS.
I am waiting for your response. Thanks:))
Relevant answer
Answer
FBS contains cell adhesion proteins as well as growth factors. Conditioned medium contains secreted cell adhesion and growth factor products produced by cells in culture. Added FBS to conditioned medium would cover up or inhibit effects of the conditioned medium.
  • asked a question related to Cell Viability
Question
4 answers
I want to check the cytotoxicity of my material. I am using MTT assay kit. The protocol is provided in the booklet with kit how to perform the assay. Is it better to follow the protocol provided in the kit or use protocol given in literature review. As in literature review different protocols are given performed with different kits.
Relevant answer
Answer
Since you are using the MTT assay kit, please follow the protocol provided in the kit.
Usually, in the MTT assay kit the MTT reagent powder is provided. You will have to prepare the MTT reagent as per the kit’s instruction namely, adding the required volume of cell-based assay buffer in the MTT vial and completely dissolving the powder. The MTT powder may dissolve slowly in the buffer. Vigorous vortexing will be needed to dissolve the powder completely. MTT solution appears bright yellow in color. You may add 10ul (or the required volume as mentioned in the kit’s protocol) of the reconstituted MTT reagent per well.
If you do not consume the MTT reagent in a single experiment, you need to store the reconstituted vial at -20 degree C in amber colored bottle until further use.
Different MTT kits have different protocol, but the objective remains the same (i.e., measuring cytotoxicity). You may have different protocols in the literature for MTT assay which may be designed as per one’s needs. For example, in the MTT assay you may use any one of the solubilization solution (like DMSO or acidified isopropanol solution, or a solution of the detergent sodium dodecyl sulfate in diluted hydrochloric acid) to dissolve the insoluble purple formazan product into a colored solution. The solubilization solution is also provided in the kit (the composition of which is not disclosed due to proprietary information) which you will have to use.
Since you have the MTT assay kit available with you, it is best in such a situation to follow the protocol provided in the kit.
Best.
  • asked a question related to Cell Viability
Question
1 answer
what can be the working conc of the dye in a 96-well plate experiment?
Relevant answer
Answer
You may prepare Resazurin (solid) at 0.015% in PBS pH 7.2.
You may weigh 1.5mg in 10ml PBS, vortex and filter sterilize (using 0.22 um filter). You may add 20ul of Resazurin solution (0.15mg/ml) per 100ul suspension per well in 96-well plate to check cell viability.
Best.
  • asked a question related to Cell Viability
Question
10 answers
As you are aware, Acridine orange is an intercalating dye that can permeate both live and dead cells. AO will stain all nucleated cells to generate green fluorescence. Propidium iodide can only enter dead cells with poor membrane integrity, so it will stain all dead nucleated cells to generate red fluorescence.
So, it is basically straightforward to calculate the cell non-viability using the PI. However, the AO enters all cells regardless of alive or dead. So how can I analyze the raw data to get meaningful cell viability/non-viability percentages?
I have positive and negative controls, and blank wells as well.
Thank you in advance.
Relevant answer
Answer
1. I've never used the well scanning feature, and I'm uncertain how you would use the results in a calculation. Perhaps, if the cells are attached to the plate in a patchy way, you could use it to find the best patch (brightest fluorescence, I suppose). However, it would be better to revise the cell plating method to get a continuous cell distribution throughout the well in order to get more consistent results. In that case, there would be no benefit to well scanning, since you would get about the same result from every position within the well.
2. I think the method you described, especially replacing the medium, will improve the result from the previous results you showed me by reducing the background. One thing to watch out for is if the background due to the fluorescent dyes sticking to the plastic of the wells is higher in empty wells than in wells containing cells, because the plastic is blocked by the cells. I can't think of any way to control for this. It may not matter if you use the ratio of the fluorescence of the two dyes as the readout.
You also should consider the length of time and the concentrations of the dyes as variables to be optimized in the experiment.
  • asked a question related to Cell Viability
Question
2 answers
In literature 30mM glucose concentration cause reduction in cell viability on HK2 cells but we tried different high glucose concentration even for 48 hours but no big difference on cell viability. what could be the possible reason for this outcome. How to address this problem, did anyone faced similar kind of issue while studying type 2 diabetics using HK2 cells
Relevant answer
Answer
High glucose induces apoptosis via up-regulation of Bim expression in HK2 cells. Upregulation of Bim impairs autophagy activity. When Bim is inhibited, HK2 cells is able to restore autophagy activity and protect themselves from injury induced by high glucose.
Bim is reported to be the ‘activator’ for Bax activation and mitochondrial apoptosis. Overexpression of Bim leads to the release of cytochrome C and apoptosis. In the attached paper, the result suggests that cytochrome C is released from mitochondria in HK2 cells undergoing apoptosis induced by high glucose. Importantly, when Bim expression was knocked down, high glucose could not induce cytochrome C release from mitochondria.
Please refer to the paper attached below for more information.
Best.
  • asked a question related to Cell Viability
Question
7 answers
Hi all, I have problem with my hepG2 cells.
So before covid lockdown, I froze my cells and put it in Mr Frosty in -80°C. I used Cryostor as the freezing media for 1st batch after I received the cells (since I was adviced that HepG2 cells viability is better with Cryostor). Though after that I replaced it with 10%DMSO in FBS due to the lack of Cryostor at that time. The numbers of the the cells I stocked varied between 3 to 3.8 X 10^6 cells per cryovial.
We transferred the tube to LN2 after about 3-4 weeks (we waited other cells to remove it to LN2 together).
And now we want to wake them up, but the HepG2 cells do not attach to the flask the next day after I thawed using standard thawing protocol (from ATCC). They become aggregates and are still floating in the media. Are they dead?
I tried with other tubes from different batches already, but the results are the same.
I wonder whether this is caused by the prolonged time in -80? The other cells are fine when we thaw them.
Or is this because of too many cells per vial? Though I saw other people froze 4 X 10^6 to 1 x 10^7 cells...
Relevant answer
Answer
"If trypsin is not neutralized after trypsinizing can cause cells to clump and not attach. After trypsinizing (Trypsin-EDTA Solution, 1X (ATCC 30-2101)) fully neutralize the trypsin- EDTA with complete growth medium and centrifuge gently then resuspend cells in fresh medium."
  • asked a question related to Cell Viability
Question
1 answer
When silver nanoparticle used as test compound for cytotoxicity studies (MTT), the results showed like the compound is toxic upto a concentration, then onwards increased cell viability with increasing concentration of the drug. Is it possible?
Relevant answer
Answer
Metallic nanopartucles interfer with MTT assay, so you have to carry out a complementary assay to assure the results of MTT.
  • asked a question related to Cell Viability
Question
6 answers
Hello, I am working with cell culture and I need some advice. I usually use trypsin to detach the cells from the flask, but I wonder if I can stop the trypsin reaction with serum-free medium instead of serum-containing medium. Is this possible or will it affect the cell viability and growth? How do you subculture your cells with trypsin? Thank you for your help.
Relevant answer
Answer
Considering the necessity of growth factors for cells even for a few minutes, I usually use medium with fbs 2%, but serum-free medium is not recommended.
  • asked a question related to Cell Viability
Question
8 answers
I have been having trouble culturing J774a.1 and at my wits end (I have posted a similar question before).
So my lab got a new vial of J774a.1 from ATCC. I thawed them in DMEM (from Hyclone). For splitting, I detach the cells using cell scrapper. The viability of the cells are at 50% for the first passage which is not surprising. However, the viability dropped to 30% subsequently, and then 20%. I changed the media to RPMI with HEPES and L-glutamine (from Hyclone) and the viability increased to 50% -60% but never more than that. Moreover, the cells only grow to 70% confluence at most. It could be my way of scrapping causing cell deaths though (I scrap as gently as I can).
So in my last ditched attempt, I seeded the cells into a non-tissue culture petri dish. They grew really well, with 90% viability. However, most of the cells are not attached (as expected). But when I seed them back into a TC flask/plates, they attach well, but again, do not grow beyond 70% confluence. Is this normal? I have yet to check for mycoplasma contamination yet though.
Any ideas how I should approach troubleshooting this issue? I am out of ideas. Please help!
Thank you so much!
Relevant answer
Answer
Hello everyone,
I am also working on J774. Once I thawed cells are round and fine. As start passaging them P2 or P3 level, they become get activated with long tentacles. Is this normal morphology of J774.? should I change the media composition?
Cells look stressed.
People often add HEPES or BME to control ROS generated by macrophages. I am speculating this could be the reason for activating them.
  • asked a question related to Cell Viability
Question
4 answers
I need to perform cytotoxicity studies with an isolated substance for a future formulation with oral administration. However, I don't know which cell lines to choose to test cell viability... Thanks in advance!
Relevant answer
Answer
When choosing cell lines for cytotoxicity studies, it's important to consider the target organ or tissue where the formulation will have an effect. Since you mentioned oral administration, you may want to select cell lines that mimic the gastrointestinal tract.
Here are a few commonly used cell lines for cytotoxicity studies related to oral administration:
  1. Caco-2: This human colon adenocarcinoma cell line is widely used as a model for intestinal absorption and transport studies. Caco-2 cells form tight junctions and exhibit properties similar to the small intestine, making them suitable for evaluating the potential toxicity and absorption of orally administered substances.
  2. HT29-MTX: This human colon adenocarcinoma cell line is derived from the same tissue as Caco-2 cells but produces higher levels of mucus, making it a suitable choice for studying mucosal interactions and the effects of substances on the mucus layer.
  3. HepG2: This human hepatocellular carcinoma cell line represents the liver, which plays a vital role in metabolism and detoxification. It can be used to assess the potential hepatotoxicity of orally administered substances, as the liver is responsible for processing many compounds.
  4. HEK293: Human embryonic kidney (HEK293) cells are commonly used for various applications, including cytotoxicity studies. While not specific to the gastrointestinal tract, these cells can provide a broad indication of cytotoxic effects and are relatively easy to culture.
  • asked a question related to Cell Viability
Question
1 answer
Let's assume that when the cell viability experiment was conducted using animal cells other than HeLa cells, the absorbance of the negative control (cells not treated with Doxorubicin) showed almost no difference from the value of the blank, and cell viability was not well measured. . In this case, please describe a strategy to solve the problem.
Relevant answer
Answer
Hi Jules Lee,
There are several factors that may impact the cell viability in this case.
1st case, Assuming you are using the same cell line, then it may due to that the dose assigned is not strong enough to inhibit the cell. In this case, a drug dosing curve will assist in finding the IC50 of the cell line.
2nd case, Assuming you are using different cell line, and only some are resistance to the same dosage of the drugs, then it indicate these cell lines tested is resistance to the drug inhibition. If not refer to 3rd case.
3rd case, the doxorubicin may be degraded. It may sound weird, but drug may be degraded, regarding how the storage are. In this case, an pre-test with a positive internal control (with cell that expected sensitive to doxorubicin) and negative internal control (with cell that expected resistance to doxorubicin) will give the answer. Also similar case can also occured in the cell viability kit used in this study.
4th case, the incompatible of the cell viability assay used. It is very rare for this case, but sometime, the cell line may not respond to the cell viability assay used. In this case, you may need to swap the viability assay, for instance alammar blue assay to cell titre Glo etc.
  • asked a question related to Cell Viability
Question
3 answers
Hello everyone
I have a question concerning the use of the Alamar blue cell viability test for cells encapsulated in an alginate-based hydrogel. Does this test fit with the gel? Does the compound reduced by the cells get out of the gel into the culture medium?
Relevant answer
Answer
Dr. Kaushik Shandilya has explained the context of Alamar blue usage. I will just say that due to their non-toxic nature and non-interference with hydrogel material, Alamar blue remains the best dye to study hydrogel-encapsulated cell viability.
We have used Alamar blue in study cell viability of MSCs encapsulated in sodium alginate. After staining with dye on day 1, we washed the beads and again incubated for further. like this we maintained cells for 5 days and measured the reduced resazurin directly proportional to cell viability.
Regards
Saurabh
  • asked a question related to Cell Viability
Question
5 answers
Hello,
I am measuring multiple different parameters in cells using fluorescent channels in flow. I would also like to assess well viability. Given that I take the same volume from each well, and do not define a stopping event, can I compare live events between wells as a viability measure? I understand that not all cells that appear live in flow are viable, thus the use of viability stains. Do such stains have to be used to generate viability data from flow?
Thanks!
Relevant answer
Answer
Hi David,
The short answer is that the use of a viability dye is required. Even with taking the same volume from each well, without a viability dye when you compare data the number of "live cells" in the gate could be different for many reasons other than non-viable cells (changes to adhesion, cellular proliferation, etc.). I would recommend using a viability dye or cell death kit (ApoTracker / Annexin V) to attain the information you are looking for. Our lab uses the following dyes extensively for our flow analysis:
https://www.thermofisher.com/order/catalog/product/L10119 (1:1000 dilution after resuspension in 50ul DMSO)
thermofisher.com/order/catalog/product/L34966?SID=srch-srp-L34966 (1:100 dilution after resuspension in 50ul DMSO)
Alternatively, some labs use Propidium Iodide or DAPI to determine viability since these dyes cannot enter live non-fixed cells.
Hope this helps
  • asked a question related to Cell Viability
Question
2 answers
I am trying to distinguish the pillar cells from thin and thick epithelial cells of Chinese mitten crab so that I can study using pH dyes regulatory capabilities of the individual cell types. Cells can not be fixed as I am doing live cell imaging and I should avoid other live cell dyes which may hamper cell viability. Attached is a picture of an intact gill lamella.
Relevant answer
Answer
@all Distinguishing pillar cells from epithelial cells in live crab gill lamella can be a challenging task, but there are a few methods that might be helpful:
  1. Brightfield microscopy: Pillar cells are typically more elongated and spindle-shaped than the surrounding epithelial cells, and they often have a distinct refractive index due to their high content of microtubules. This can make them stand out from other cells under brightfield illumination.
  2. Differential interference contrast (DIC) microscopy: Similar to brightfield microscopy, DIC microscopy uses polarized light to highlight differences in refractive index between different structures within a sample. This can make it easier to distinguish pillar cells from other cells in the gill lamella.
  3. Confocal microscopy: By using specific fluorescent dyes or antibodies that selectively label pillar cells or epithelial cells, confocal microscopy can provide high-resolution images that allow for clear identification of individual cell types. This method may require some optimization to ensure that the dyes or antibodies do not affect cell viability.
  4. Scanning electron microscopy (SEM): SEM can provide high-resolution images of the surface structure of the gill lamella, allowing for identification of specific cell types based on their morphology. However, this technique requires the sample to be fixed and dried, so it may not be suitable for live cell imaging.
It may be helpful to try a combination of these methods to determine the best approach for distinguishing pillar cells from epithelial cells in your specific experimental setup.
  • asked a question related to Cell Viability
Question
9 answers
Drug toxicity test against normal cells
Relevant answer
Answer
Yes, you can easily apply MTT or other simpler assays like Cell titer Glow from Promega on normal cells. I use normal cells all the time.
  • asked a question related to Cell Viability
Question
3 answers
Hi there,
I used L-NAME to treat cancer cells. Then I did LDH and MTT, according to these results, MTT showed the cell viability was above 100% compared with control, and LDH showed cytotoxicity was nearly unchaged.
However, I saw some studies suggested that L-NAME could inhibit cancer cells proliferation. I'm confused with it.
Does anyone have some experiences with it?
Many thanks.
Relevant answer
Answer
When doing MTT, be careful not to have too many cells. When it is excessive, cell viability can be seen more than control.
good luck
  • asked a question related to Cell Viability
Question
2 answers
Hi, I am performing MTT assay to measure cell viability of ovarian cancer cell line (A2780) upon treatment with Cisplatin. However, I am repeatedly getting the following results and not sure why I have values in negative.
Can someone suggest me a possible explanation?
Relevant answer
Answer
Because at that dose all the cells gets killed and the reading at 100uM and 1000uM is of may be the reading of liquid. So, you may conclude that at that point the no viable ovarian cancer cells are present i.e., 0% viability.
  • asked a question related to Cell Viability
Question
2 answers
I have done two tests to investigate the effect of a material to cell viability and how cytotoxic the material is.
Briefly, I seeded cells in a 96-well plate and introduce the cells with the media treated with the material, and then place them in the incubator for 24h before testing. The next day, I did the PrestoBlue test and followed by the LDH assay.
When I did the PrestoBlue, the control contains of huge number of cells which is making sense for me as it is a control. But when I did the LDH assay, the reading between control (media + cells only) is quite high compared to the blank (media only). From what I understand, this means that cells are dying in the media, but this result does not correspond with the PrestoBlue result.
Can anyone help me explain what is going on? Is it a user fault (my fault) or is there any scientific reason why the results are not corresponding to each other?
Relevant answer
Answer
Thank you for insight, it does really help me.
Just a follow-up question, does this means that during the PrestoBlue assay, everything was actually okay, but when I did the LDH assay (which I did about 30 minutes after the PrestoBlue assay), something had happened due to user’s fault causing higher reading for the control?
Because the way that I understand it was, when I did the PrestoBlue, the cell number is high which is a good data result, and as I did the LDH using the same media from the well for PrestoBlue and only a couple of minutes after the PrestoBlue, it should not have a high reading. But if it did, what actually user had done causing the reading to be high?
p/s: I really appreciate your time and insight! Thank you again
  • asked a question related to Cell Viability
Question
1 answer
I treated my cell (HL60 and Mec-1) with Graviola leaf extract and Vitamin C alone and in combination form but the viability of cell increased in all treatment and the assay which I used was MTT. Can anyone know the reason behind increase cell viability and how to overcome this problem?
Relevant answer
Answer
The MTT assay is finicky as it monitors mitochondrial "health" so agents that stimulate mitochondrial activity might result in an increase in baseline. One might suspect this to be true for cmpds with antioxidant capacity, such as citrate. Try another assay for viability if your goal is to measure cytotoxicity.
  • asked a question related to Cell Viability
Question
4 answers
Aslamo Alikom (Greetings) Everyone,
I've MTT results for the cell viability after an exposure to an inhibitor alone and combining it with another drug.
Now I wanna see how the other drug affects the IC50 of the main inhibitor.
Cell viability% for the drug alone and with combination ranges from 100 to nearly 40s and 30, respectively.
I was wondering whether I've to normalize the data before doing non-linear regression to calculate the IC50 on Graphpad Prism.
I would highly appreciate your advice and guidance in this matter. Thanks in advance
Relevant answer
Answer
When looking at the effect of inhibitor A on inhibitor B, the maximum signal at zero concentration of inhibitor B will decrease as the concentration of inhibitor A increases. This will reduce the range of values between the baseline (no cell growth) and zero inhibitor B, but it does not change the way you calculate the % inhibition at each concentration of B. Therefore, it is not necessary to normalize the data.
If the signal at no cell growth is called MIN, the signal at zero concentration of inhibitor B at any particular concentration of A is called MAXA, and the signal at any given concentration of inhibitor B at this concentration of A is called XA, then
% inhibition = 100[1-(XA-MIN)/(MAXA-MIN)]
and the equation to use for nonlinear regression is (for example)
% inhibition = 100[I]n/(IC50n+[I]n)
where [I] is inhibitor concentration and n is the Hill slope.
  • asked a question related to Cell Viability
Question
4 answers
actually, I want to calculate growth rate of my cho cells. I have its viable cell count, total cell count, and viability percentage. so is there any formula for growth rate calculation using these values?
Relevant answer
Answer
Hello Dear Misba,
To know the growth rate of your cells, you need to divide the final population of cells to the inicial population and take the natural logarithem of it and divide it to the time (per hours) and then multiply to 24.
I found this website to calculate the growth rate and doubling time
also the logic of calculation is provided in this website
  • asked a question related to Cell Viability
Question
1 answer
I did a MTT assay for a plant extract against L929 cells. But in readings, the cell viability percentage was increased more than the untreated cells, with increasing concentration.
Relevant answer
Answer
Dear Ameena,
the MTT assay is only indirectly a test for viability, as MTT is reduced by microsomal enzymes which oxidize NADH to provide the electrons for MTT reduction, depending on the metabolic state of the cells. Also, some compounds can directly interfere with MTT, thereby changing / increasing its absorption.
Ref.: Ulukaya, E.; Colakogullari, M.; Wood, E.J. Interference by anti-cancer chemotherapeutic agents in the MTT-tumor chemosensitivity assay. Chemotherapy 2004, 50, 43-50.
Here is also a useful reference on the biochemistry of MTT and other tetrazolium salts in cell assays:
Berridge, M.V.; Tan, A.S.; McCoy, K.D.; Wang, R. The biochemical and cellular basis of cell proliferation assays that use tetrazolium salts. Biochemica 1996, 4, 14-19.
In any case, you should look at the cells in the microscope and count them after treatment with plant extracts.
Hope this will help you to get correct answers.
Angela Otto
  • asked a question related to Cell Viability
Question
4 answers
I have been working with a ceramic based scaffold, and it is to be determined for cell viability, for which I have chosen MTT assay. Although I have tried seeding cells onto the scaffold or taking extracts of the scaffold and then treating them to the cells, I was unable to get proper results. In general, the problem faced was that the absorbance of media control is remaining higher than that of cell control, which in general shouldn't happen. It would be great if any researcher can help me sort out the issue or suggest any other method that can be followed to perform a cell viability assay.
Relevant answer
Answer
Thank you Ferry P W Melchels for the suggestions.
  • asked a question related to Cell Viability
Question
3 answers
I treated cells with a toxic compound and was trying to find the most suitable treatment concentration. After performing a cell viability test, I saw that the middle concentration reduced the cell viability, but higher concentrations did not cause a reduction in cell viability. Does someone know what the reason could be?
Relevant answer
Answer
Another possibility is that the compound was not soluble at the higher concentration and precipitated/crystallized.
  • asked a question related to Cell Viability
Question
3 answers
I am facing difficulty in estimating cell quantification in my scaffold. Usually live/dead assay and MTT is performed to check cell viability. But what if we can extract cells directly from scaffold and measure it using a cell counter.
Relevant answer
Answer
Why not determining total DNA amount?
  • asked a question related to Cell Viability
Question
5 answers
I am trying optimize 2L bioreactor process with SP2/0 cell line. This is the third batch which was initiated. The initial batch parameters are 0.5x10^6 cells/ml, 37 degree C, air and O2 cascade: 0.09 and 0.03 vvm. The viability of the cells used for inoculating the batch was between 90 to 92%. But i am observing one common trend during all batches. From first day onwards, though there is doubling of cells but the viability decreases as the days proceed. And from day 2 onwards the doubling ceases and viability reaches to 50%. By the end of third day i need the discard the batch.
I am unable to understand the situation. If any have ever observed this kind of situation while working with SP2/0 cell line. Please share their experiences.
Relevant answer
Answer
Seeding density optimized at 3E5 cells/mL . Don't put too high.
RPM is very important. What is your tip speed?
DO set at 40%
pH setting : Keep floating like in SF, then start to control from D3 at 7+/-0.2
  • asked a question related to Cell Viability
Question
2 answers
I am working on a probiotic that have anaerobic bacteria spp. This probiotic is available in capsules commercially from Seeking Health LLC USA.
I need to dissolve the capsule powder in anaerobic PBS. In resulting Bacteria suspension I want to check the viability of the suspended bacteria.
I did a literature survey and found that few authors are telling to handle it dry anaerobic conditions. Mix the powder in anaerobic PBS and homogenize it. After this I can check cell viability via flow cytometry.
Do the homogenization maintain the cell integrity and viability?
If yes, what are the homogenization conditions to maintain the cellular integrity and cell viability.
Can I use Trypan Blue dye exclusion method for the viability check?
Or only Fluorescent dyes (Flow Cytometry) are the option ?
Expert advises are needed, please help me in this.
Thanks
Regards
Anuj
Relevant answer
Answer
Thanks Alzeyadi!
I will implement your suggestions and let you know it is working or not
  • asked a question related to Cell Viability
Question
3 answers
We want to get single cell suspension for flowcytometry staning from mice colon lamina propria.
We can only make about 30-40% cell viability after digestion.
We have tried different enzyme concentration(1000U and 2000U for collegenase VIII, 5mg and 10mg for collegenase IV in 10mL) ; dierent digestion time (1h and 2h);different wash time(10min twice; 20min once; 20min twice); But none of them is capabel to make single cell suspension over 70%.
Why?
We used followed protocol:
PBS wash tissue
PBS + 30mM EDTA + 1mM DTT, 220rpm, 37°C 20min in Shaker-Incubator
PBS + 1mM DTT, 220rpm, 37°C 20min in Shaker-Incubator
Hand shaking for 1min
wash tissue with PBS and shear the tissue to 3mm slice
Digest with 100U/ml collegenase VIII and 150ug/ml DNase I for 1 hours
Pass cell through 70um filter
Stain cell with AO/PI to calculate cell viability.
Thanks so much for your suggestion.
Relevant answer
Answer
Adding to the above answers, solid tissues are particularly challenging to work with, usually requiring a combination of mechanical and enzymatic dissociation to produce a viable single-cell suspension.
In tissues, cell types are embedded to different degrees meaning that carefully selected enzyme combinations are required to release all cell types and to reflect the true heterogeneity of the sample. You should design your protocol in such a way that it considers not only the nature of the tissue but also the nature of the cells of interest.
Collagenase and DNase are among the most commonly used enzymes to prepare single-cell suspensions from tissue samples. Collagenase helps to break down components of the extracellular matrix and DNase helps to digest free DNA that can cause aggregates. But care must be taken in collagenase selection as some collagenases have proteolytic activity that can damage the protein target of interest. So, you should select the right collagenase and DNase.
Also, adding EDTA to buffers and using cell strainers will help to remove clumps and enhance the quality of single-cell suspension.
Cells should always be handled gently, without subjecting them to harsh vortexing or centrifugation forces.
Some tips which you could consider:
1. Use scalpel, blade, or scissors to increase total surface area.
2. Determine the optimal concentration of enzymes.
3. Determine the optimal temperature and time for digestion.
4. Use orbital shaker to assist in mechanical dissociation.
4. Centrifuge cells at 300–900 RCF.
5. Pipette gently and avoid bubbles.
6. Use polypropylene tubes and plates to avoid cell adhesion.
Good Luck!
  • asked a question related to Cell Viability
Question
3 answers
My work aims to observe cell viability through AO/PI staining. Although I found numerous AO/PI staining protocols, but most of them analyzed the viability through microplate reader or cell counter (countess).
We're trying an approach of observing these cells under microscope with fluorescence setting. I plan to seed my cells into 96 well plates, and later stain them with AO/PI staining. However, I couldn't find any detailed protocol for this.
It seemed like AO/PI stain was diluted with PBS, prior staining - but this was for microplate & countess protocols.
Should I dilute my AO/PI stain? if yes, how much volume is needed? I'd also like to know if there is any incubation period before observing my stained cells under microscope.
Thank you!
Relevant answer
Answer
AO/PI staining is a technique used to determine the viability of mammalian cells by staining them with acridine orange (AO) and propidium iodide (PI). The protocol for performing AO/PI staining on mammalian cells in 96 well plates is as follows:
  1. Harvest the cells: Harvest the cells from the culture dish by gently rinsing them with PBS and trypsinizing them according to the appropriate protocol for the cell type.
  2. Wash the cells: Wash the cells twice with PBS to remove any remaining trypsin or other contaminants.
  3. Resuspend the cells: Resuspend the cells in a small volume of PBS. The concentration of cells should be adjusted to a suitable concentration, typically around 5 x 10^5 cells/mL.
  4. Add AO/PI staining solution: Add a small volume of AO/PI staining solution to the cell suspension. The staining solution can be prepared by mixing equal parts of AO and PI in a suitable buffer such as PBS.
  5. Incubate the cells: Incubate the cells with the staining solution at room temperature for 10-15 minutes. This will allow the staining solution to penetrate the cells and bind to the DNA.
  6. Analyze the cells: Analyze the cells using a suitable method such as fluorescence microscopy or flow cytometry. The AO will fluoresce green, while the PI will fluoresce red. Cells with intact membranes will fluoresce green, while cells with damaged membranes will fluoresce red.
  7. Calculate the percentage of viable cells: Calculate the percentage of viable cells by dividing the number of green cells by the total number of cells and multiplying by 100.
AO/PI staining is a useful technique for determining the viability of mammalian cells in a 96 well plate format. It is important to follow the protocol carefully and use proper laboratory techniques to ensure accurate and reliable results.
  • asked a question related to Cell Viability
Question
2 answers
I'm using the standardised INFOGEST - Minekus (2014) consensus paper for my simulated in vitro digestion model but each time I use the digesta (intact, diluted 1:4 and 1:8) on the caco-2 cells to assess carotenoid uptake and transport, the cell viability is less than 10% based on the MTT assay. I strongly suspect the bile salt concentration I'm using could be toxic to the cells. Is there a step by step procedure I can use without necessarily using the test kit to verify the actual concentration of my bile salts?
Relevant answer
Did you ever find a solution? Looking into the same issues.
  • asked a question related to Cell Viability
Question
1 answer
Hello There
I´ve been trying to do a sulforhodamine cell viability test for NSC-34 and it is not working. I changed the protocol to minimize cell detachment (washing only once, using collagen) but when I finish, there is almost no cell in the well. I see that some papers use LDH assay to cell viability in NSC-34. Is it better to change or am I letting something pass?
Relevant answer
Answer
I suggest you use sulforhodamine B (SRB) assay for your cells. SRB assay is fast and sensitive. It is independent of cellular metabolism and the reproducibility of this assay is high.
Some of the steps in SRB assay you need to follow carefully.
1. When you add 50ul of 50% cold TCA to each well during fixation step, you add it directly to 200ul medium supernatant to give a final TCA concentration of 10% TCA, and while adding the micropipette tip should touch the culture medium surface at the edge of the well to avoid fluid shearing forces which could result in cell detachment and loss. Mixing is not required, as this could lead to some cells detaching from the bottom of the well.
2. If you decide to eliminate the media and add 100ul of 10% TCA directly to each well, be careful, because washing cultures with buffer prior to fixation to remove serum proteins would cause cell detachment and loss.
3. Wash the plate four times by submerging the plate in a tub with slow-running tap water and remove excess water by gently tapping the plate into a paper towel. After the last wash allow the plate to air-dry at room temperature. Cell monolayer detachment can occur if water is forced into the wells. Also, it is better to let the plate dry completely before continuing to next step.
The only disadvantage of SRB assay is that it is important to evaluate the linearity of the assay response and to use the correct number of cells.
Hope this helps!
Good Luck!