Science topic

Tumor Cell Culture - Science topic

Explore the latest questions and answers in Tumor Cell Culture, and find Tumor Cell Culture experts.
Questions related to Tumor Cell Culture
  • asked a question related to Tumor Cell Culture
Question
5 answers
Hi guys,
I'm working on the tumor mouse model. In general, I prepared tumor cells and injected cells into the flank of mice subcutaneously.
After around 3 weeks, I found, in the same group, there's a variation in the tumor weight. For instance, some tumor were around 1.2g, some 0.6g.
Could you give me some suggections or tips about how do tumor inejction?
Which type of needle do you need for injection? or How to prepare tumor cells suspension? or whatever something you think is important to do that successfully
Many thanks :)
Relevant answer
Answer
Thank you so much for your advice. Nurul Ashikin Mohamed Shahrehan
  • asked a question related to Tumor Cell Culture
Question
3 answers
Hello,
I am trying to transduce my CD4/CD8 T-cells with lentivector using retronectin from Takara (#T100B) at 50ug/ml, after they are activated by CD3/CD28 for 2 days.
The official protocol says that "it's important that the target cells be in logarithmic growth phase", but how this state may be monitored?
How to choose the "perfect moment" for transduction via retronectin?
Thanks in advance!
Relevant answer
Answer
Well said Aledjandrina
  • asked a question related to Tumor Cell Culture
Question
12 answers
Need this cell line urgently....
Relevant answer
Answer
Hi Gurmeet
We worked long back with 4T1 cell line, and not sure if my lab still have a cryovials of 4T1, But, you can try contacting Dr. Satish (sathish.vemuri@gmail.com) and give my reference.
Regards
Rajkiran
  • asked a question related to Tumor Cell Culture
Question
3 answers
Dear brain tumor and U-87MG gurus,
Greetings!
Recently I bought a vial of U-87MG from ATCC and started culturing them following ATCC's SOP. However, I ran into a problem that the cells tend to grow in big spheres (attached pictures 1, 2, 3), rather than the nicely spreadout monolayer shown in ATCC website (attached picture 4). This bugs me a lot as I'm not sure whether I have a problem here, especially after comparing the morphology of cells I got with the one posted on ATCC wesbite. I'm wondering if anyone had this issue and would much appreciate for any insight.
Details about the culture:
1. Media: I have used ATCC's recommended media, which is ATCC's EMEM+10%FBS (not heat-inactivated)+P/S. I have also used Sigma's high-glucose DMEM+10%FBS (not heat-inactivated)+P/S+GlutaMax+Na-Pyruvate+NEAA+HEPES. However, both conditions resulted in similar problem as mentioned above. 
2. Plates: I have used regular TC-treated plates from two different vendors (TPP and Sarstedt), and the issue happens in both.
3. Seeding density: 4x10^4 cells/cm2, i.e. seeding 6 million cells per 15cm plate
4. About attached pictures 1/2/3: seeded in 15cm plates as mentioned above in Sigma's high-glucose DMEM+10%FBS (not heat-inactivated)+P/S+GlutaMax+Na-Pyruvate+NEAA+HEPES for 5 days. Media change every other day didn't seem to address or improve the issue.
5. I sent the attached pictures 1/2/3 to ATCC tech support for consultation, they concluded I'm having contamination, as judged from the morphology of cells in the culture. I have not tested mycoplasma contamination for these cultures yet.
6. I've also tried longer trypin treatment time (20min at 37C), which seemed to help the cells to spread out nicely on the 2nd day, however, the problem came back on the 4th day.
7. The culture shown in the pictures is about the 10th passage of the cells from the received vial from ATCC.
8. I had some experience on adherent culture of glioma stem cells on laminin-coated plates. Those cells will appear like the pictures 1/2/3 here if I grow them in regular TC-treated plates. I guess I'm not surprised to see U-87MG is growing in a similar way with big spheres on TC-treated plates, but the ATCC culture picture destroyed my guess here with very nice monolayer culture....
Thank you very much in advance for your time, consideration and help!
Best,
Kai
Relevant answer
Answer
With the U87 cells, to prevent the clusters I do multiple passages with my pipette and I get good results
  • asked a question related to Tumor Cell Culture
Question
1 answer
Please take a look at the attached file.
I irradiated cells using a fractionation regime of 3 x 1 Gy after exposure to a substance in different concentrations.
I made an XY table with the determined SFs and plotted a graph using the LQ-model.
The equation I used was Y=exp(-3*(A*3*X + B*3*X^2)). Its an edition of the provided equation Y=exp(-1*(A*X + B*X^2)) in regard to the fractionation regime.
To determine the AUC I used the standard analyzing tool that Graphpad provided.
Could someone tell me, if this is right or if I mistaken somewhere?
Tank you very much in advance!
Relevant answer
Answer
There are two, very different, ways to model an LQ model. The first assumes that the fractionated curve continues along the single dose curve. The second assumes that there is full recovery from each fraction and therefore the initial curve is repeated from the previous dose SF. The area between these curves is called the "envelope of additivity". See G.G. Steel or Peckham and Steel for more on this addition of survival curves for multifractionated doses. Interestingly, ionizing radiations (with shouldered survival curves) tend to repeat the initial portion of the curve (so-called repair of sublethal damage, but actually split-dose recovery), while some alkalizing agents, such as Bleomycin, have their curve continue along the single-dose curve (no split-dose recovery).
  • asked a question related to Tumor Cell Culture
Question
5 answers
I have recently started cell seeding inside my PDMS microfluidic device with syringe pump. I am actually trying to form tumor spheroids inside microwells coated with pluronics. However, the MCF7 cells instead of forming spheroids are attaching to PDMS surface. Please let me know if you have faced similar problem while forming spheroids on PDMS microfluidic devices
Relevant answer
Answer
try BSA coating on PDMS
  • asked a question related to Tumor Cell Culture
Question
3 answers
I am processing high-grade serous ovarian cancer samples and trying to grow tumor cell culture for subsequent functional assays. Any recommendation on the dissociation process, enzyme mix and/or culture media?
Relevant answer
Answer
...in addition to the previous comments following papers may help
doi:10.1038/nprot.2006.328
DOI: 10.1038/ncomms8419
Good luck
  • asked a question related to Tumor Cell Culture
Question
3 answers
We did qpcr using our sample tumor cDNA to compare with four different normal human tussie (colon, prostate, thymus, and spleen). I've calculated the 2^-ΔΔCt to be 7.08, 3.10, 0.17, 21.2. I understand that this means the gene expression level of the tumor sample is 7.08, 3.10, 0.17, 21.2 times of control tissue. I'm not sure if how much 2^-ΔΔCt would suggest mutations and can I conclude this to be tissue specific changes as the highest result is in spleen?
Relevant answer
Answer
I would ask a question about fold change calculation,{ FC=2^-(ΔΔCt) or FC=2^(ΔΔCt)}, when did you use the minus and when you did the calculation without minus?
  • asked a question related to Tumor Cell Culture
Question
4 answers
Dear All,
I am taking over the Master's student's project and I am a new PhD student. We have experiments involving tumour implantation in mice. The MSc student is used to scraping cells down when passaging and only uses trypsin to detach the tumour cells for implantation.
In my previous lab, I was taught to trypsinise as it is less damaging to cells when passaging and that apoptotic cells could influence culture environment. Therefore, I have been using trypsin (5 minutes, 37 degree C). However, I have noted a difference in cell growth as the MSc student is used to passaging at very, very low concentrations of 1:1000 whilst I resorted to passaging 1:10 every 3-4 days. I have been keeping track of the growth rate of my cells which is about 3.1-3.7% (quite consistent) but they seem to be growing much slower than the MSc student.
I consulted the MSc student and I was told to swap to scraping. I am just curious as to how this may affect the cells and whether anyone has ever noted such a huge difference.
Note: We re-use the T75 flask (hence, why I decided on trypsinising to cause less damage to my cells).
We mostly run FACs and IHCs in our lab if that will help determine which method would be more suited to our cause.
Thanks.
Relevant answer
Answer
Hi Darelynn,
In my experience if you leave EO771 with trypsine for too long, they lose their adhesion proteins and they are not able to recover them. You will notice that cells are alive but they never atach to the plate. For this cells, 5 minutes is too long. I leave them in trypsine for 1 min at 37 degrees to do the passages and it works just fine.
  • asked a question related to Tumor Cell Culture
Question
4 answers
Hi, I am currently design a assay for testing the cytotoxicity of primary T cells against tumor cell lines. Here is the design:
Cryo-preserved PBMCs were retrieved and cultured with 9ug/mL PHA, 20%FBS RPMI1640 medium for 48 hours. Then the PBMCs were stained with CD3 antibody and were sorted for CD3+ T cells. The adherent tumor cell lines A549 were seeded the day before add T cells in 96 well plate (3000 cells/well). Tumor cells were stained with LIVE/DEAD® Viability/Cytotoxicity Kit (Themo Fisher: L3224). After T cell purification and tumor cell staining, we add T cells to tumor cells in 200 ul medium. Finally, the cytotoxicity were observed in In-Cell-Analyzer 2200 (GE) for 6 hours by real-time imaging and count for red signal of dead tumor cells. The RNA of T cells were collected after recording of images, and cytotoxicity genes expression, like IFNG, PRF1, GZMB, will be run on real-time qPCR. CD3 will be used as housekeeping gene.
I know the PHA will activate T cells and stimulate them proliferating via CD2 and calcium influx pathway. And I do observe obvious cytotoxicity in the preliminary experiments. Could anybody give me some advices to improve this method?  . 
Relevant answer
Answer
I think that 3000 cells are not enough to isolate RNA to run qPCR.
Plus, cytotoxicity of T cells is not very strong and 6 hour of co-culture maybe is not long engough.
  • asked a question related to Tumor Cell Culture
Question
3 answers
I am generating Luciferase-expressing stable Tumor Cell Lines.
First i co-transfected the Lentiviral plasmid containing the luciferase gene with 293t cell and produce the virus. Than i transduce the tumor cell line with the susp collected from from 293t transfected cell.
How should i analyzed that my tumor cell line express the luciferase successfully? I study some papers for protocol , but their plasmid have GFP reporter gene, so its quit easy to detect. But mine plasmid don't have GFP or any other reporter gene.
thanks.
Relevant answer
Answer
Since you are trying to generate a stable recombinant cell line, I think you can first screen the lentivirus-transduced cells using an antibiotic such as puromycin if there was any selectable marker in your lentiviral construct.
  • asked a question related to Tumor Cell Culture
Question
6 answers
I'm trying to study NAD+ metabolism in cancer cells. I'd like to study the levels of NAD+, nicotinamide and nicotinamide mononucleotide (NMN). Assay kits for ATP and NAD+ are available but I haven't found any for NMN or nicotinamide. Any suggestions?
  • asked a question related to Tumor Cell Culture
Question
5 answers
I'm interested in culturing solid primary tumor cells using trypsin. Does anyone have any good protocols or literature they could send my way please?
So far this is what I've found but I'd like to have a cheaper/more recent alternative
Thanks!
Relevant answer
  • asked a question related to Tumor Cell Culture
Question
6 answers
Hello,
I'm trying to get some mouse xenografts started for a gastric model that basically involves efficacy testing. What are some good human gastric cancer xenografts that give consistent tumors in nude mice with short latency?
Relevant answer
Answer
We have several models (http://altogenlabs.com/xenograft-models/gastric-xenograft-model/) all of which have been successful. If you desire some kind of specific trait in the cells (whether it be a pattern of expression or whatnot) then you'll need to do some background research on each cell line and see what suits you best. If you've never done a xenograft with gastric cancer, contact us directly and we'd be happy to assist.
  • asked a question related to Tumor Cell Culture
Question
4 answers
Does the NO product stays inside the cell or is it excreted in the extracellular medium? I am not sure if I shoud add the reagent in the intact cell (adhered in the bottom of the 96-wells plate), take out the treated medium and put fresh one instead, or if I should keep the old medium ?
Relevant answer
Answer
First, the Griess assay measures nitrite (and nitrate, if you use a reductase to convert it to nitrite) and not nitric oxide. Nitrite is an aqueous oxidation product of NO, and can be considered to be an indicator of NO bioavailability. If NO is being produced, then over time nitrite should accumulate in your media. So you want to sample media and use the Griess assay to measure the nitrite content in the media.
The longer you allow the cells to make the NO and thus the nitrite to accumulate in the media, the more likely you are to detect something. For example, if you don't detect any nitrite after 72 hours of culture, then you likley won't after 48 either. When you change the media, you reset the clock. Remember the volume of the media above the cells will determine the concentration, so try to keep the volume low, no more than 100 uL for a 96 well plate and be aware of possible evaporation related edge effects.
Finally, make sure you run your controls and calibration standard curve in the same media!
  • asked a question related to Tumor Cell Culture
Question
3 answers
Im working on the targeting of NPs to specific tumor cell and to relase the payloads there.
Relevant answer
Answer
Aptamers bind to a target molecule by providing a limited number of specific contact points imbedded in a larger, defined three-dimensional structure.
Aptamers might be difficult to generate for less expressed target proteins on cells.
  • asked a question related to Tumor Cell Culture
Question
5 answers
I need the media for conditioning of my DCs. Also I wish to check for the expression of phosphorylated STAT 3. Can You please help me out with the duration of conditioning.
Relevant answer
Answer
You can culture them in 1640+10%CS. Don't subculture too many generations, it may induce variation (the reason is none clearly).
  • asked a question related to Tumor Cell Culture
Question
10 answers
 I‘m studying a natural product's anti-tumor effect on melanoma. I was noticing it  triggered ER stress,  but at the same time, the autophagy related protein such as ATG5, ATG12 were down regulated. Also, the ratio of LC3-I and LC-II was elevated. As I know, the ER stress is usually considered as an inducer of autophagy. So I wondered if It possible to get such conclusion or do I need to do more experiments to prove that?
Relevant answer
Answer
You can see the following papers: 
Tyson, John J., et al. "Dynamic modeling of estrogen signaling and cell fate in breast cancer cells." Nature reviews. Cancer 11.7 (2011): 523.
Tavassoly, I., et al. "Dynamic modeling of the interaction between autophagy and apoptosis in mammalian cells." CPT: pharmacometrics & systems pharmacology 4.4 (2015): 263-272.
The interaction among ER stress , Autophagy and Apoptosis has a dynamics. If apoptosis is induced, it can reduce autophagy. 
  • asked a question related to Tumor Cell Culture
Question
3 answers
MG-63 cells, a line derived from an osteosarcoma, produce high yields of interferon after superinduction with polyinosinic acid polycytidylic acid, cycloheximide, and actinomycin D. Studies using MG-63 cells provide some important mechanistic clues concerning the details of the amplification process in tumors. Cells can also be transfected
Relevant answer
Answer
Because this cell line has the ability to retain a differentiated phenotype under culturing conditions. Additionally, the scientists prefer using MG-63 because it proliferates so fast compared to normal osteoblast. However using this cell line for implant related studies is controversial. From my experience, I would not recommend using it as a model for osteoblast because it behaves differently in many conditions. It is a cancer cell line
  • asked a question related to Tumor Cell Culture
Question
2 answers
Hi, I am planning to isolate single cells from human lung tumour tissue. I am wondering what is the best way to enzymatically degrade the tissue to isolate MOST of the cells (since the population of interest is very rare and I want to avoid false negatives when I do FACS analysis). I came across multiple collagenase cocktails which have been reported in literature, but not sure which is the best way forward. Which is the best collagenase to work with?
Any leads would be appreciated. I don't want to optimise too much since these are precious patient samples. Thank you! :)
Relevant answer
Answer
You can digest tissue in RPMI-1640 (2-3% serum) containing 0.8mg/ml Dispase and 0.2mg/ml Collagenase P (Roche). Tubes should be incubated at 37˚C in a water bath or incubator and gently invert at 5-10 min intervals to ensure the
contents are well-mixed. After 30 min,gently collect the cells in another tube and add the fresh media with collagenase and dispase. Total process will take around 60 min. Centrifuge the cells and plate them in fresh media.
  • asked a question related to Tumor Cell Culture
Question
4 answers
I want to culture glioma tumour spheres/neurospheres from Gl261 adherent glioma culture.I am going to use a media having DMEM/F12 supplemented with N2,bFGF and EGF.Could anyone please suggest how much N2 I should add to my media?
Thanks in advance for your valuable input.
Relevant answer
Answer
Thanks Chanchai!
  • asked a question related to Tumor Cell Culture
Question
4 answers
Hello there,
I am working to isolate breast cancer stem cells from MDA-MB-231 using CD44+/CD24low positive selection on magnetic separation (Miltenyi Biotech column). Has anybody used this kit for solid tumor cells and not hematopoietic cells that the protocol is based on? I reach a good enrichment but I end with very low number of cells to work with. Any suggestion is helpful.
Relevant answer
Answer
As far as i know, almost all MDA-MB-231 cells are CD44+/CD24low or negative. I would suggest to perform flow cytometry before and after purification. A caution: If you are detaching cells with extensive trypsinization, then the cells may loose certain surface markers (potentially CD44 as well).
  • asked a question related to Tumor Cell Culture
Question
7 answers
I want to develop 4T1 tumor model. How many cells are needed to develop a subcutaneous tumor in Balb C mice?
Should I can use male Balb C mice in place of female?
If anyone have any clue about this, please guide me ASAP.
Relevant answer
Answer
Do the experiment to determine your tumor dose 100 (TD100), the minimal dose where all mice grown tumor.  Our TD100 for 4T1 is around 1 x 10^3 cells in the mammary fat pad. So we use 5 x 10^3 cells for our experiments. 
Cell lines can vary slightly, so it's best to find out what your own TD100 really is. Plus with a more minimal dose there is a better chance your therapy will work (if that's what you're testing). 
  • asked a question related to Tumor Cell Culture
Question
5 answers
I've begun to establish some PDX lines that consistently passage through mice. At this point, I'd like to make a cell line from these PDX tissues. When I dissociate the tumor tissue and plate it I end up with a lot of mouse stroma that eventually takes over the culture. I'm hoping someone has a good protocol for deriving a purified human tumor line from PDX tissue. We have a Miltenyi MACS system but have not have success using their mouse cell depletion kits after tumor dissociation. At this point any alternative approach could help get over this hurdle. 
Relevant answer
Answer
Hi Steven,
I was also going to suggest the mouse cell depletion kit. Actually, there are also kits for untouched isolation of stromal cells from syngeneic mouse tumors or human patient samples.
Best regards & good luck,
David
  • asked a question related to Tumor Cell Culture
Question
1 answer
Thank you
Relevant answer
Answer
Hey Siegel,
You can use this:
Starting material: Tissues stored as chucks under liquid nitrogen.
During handling the material is kept on ice. Before generating a lysate, the tissue is first cut into ~1mm cubes by using a razor blade on a glass plate held on ice.
The small cubes are then transferred into a hand held potter homogenizer with three ml ice-cold RIPA buffer per gram of tissue.
 Tough tissues like Prostate, Skin and Thyroid need incubated in the RIPA for 20
min on ice before starting homogenizing
 Medium tough tissues like Colon, Duodenum, Heart, Kidney, Skeletal Muscle or Tonsil, are kept in RIPA on ice for 10min before homogenizing
 Homogenize by pushing the piston slowly into the mix by continuously twisting the wrist thus turning the piston around its axe.
 Make sure all tissue chunks slide between the piston and the glass wall while homogenizing.
 Once the piston reaches the bottom, reverse the handling
 Keep the tissue submerged in the ice during the process
 Repeat the cycle until the tissue is liquefied
 Divide the liquefied tissues over 1.5ml tubes and centrifuge at 13,000rpm for 3 min at 4C
 Transfer the clear supernatant in new clearly labelled tubes. Take 20ul out for protein determination
 Protein determination is carried out using the BioRad Protein Determination Kit.
 Adjust the lysate to 5mg/ml by adding ice cold RIPA buffer
 Store in liquid nitrogen.
RIPA buffer: 20mM Tris-HCL pH7.4, 150mM Na\Cl, 1mM EDTA, 1% Triton-X100, 1%
sodium deoxycholate, 0.1% SDS with freshly added PMSF to 1mM and with freshly
added aprotinin and leupeptin to 5ug/ml just before use.
All the best.
  • asked a question related to Tumor Cell Culture
Question
5 answers
Hey Guys,
I am about to set up a drug release experiment and it requires the pH of buffer to be tumorigenic to mimic tumors. I went through literature and it tells that people have used pH ranging 5-6.8. This got me confused a bit as in which buffer I need to make. So, could you please tell me what exactly is the pH of tumor cells and what should be used for drug release experiment?
Thanks.
Relevant answer
Answer
It is depending on a lot of factors and it also amazingly varies within various areas of a same tumor.
See the attached articles,
Robert
  • asked a question related to Tumor Cell Culture
Question
5 answers
I'm interested in identifying T-cell populations in mouse tumor sections. I can do IHC or IF and I have paraffin or frozen sections.
Relevant answer
Answer
Hi,
I can confirm the abovementioned SySy anti-CD8a antibody works (is rabbit, not rat) in FFPE samples for mouse. 
I have not tried their aCD4, we use a rabbit Ab from Sino Biological (ref 50134-R001), also FFPE/mouse.
For CD3 we use DAKO ready-to-use solution (ref. IS50330; is for human but works).
Good luck,
 Daniele
  • asked a question related to Tumor Cell Culture
Question
2 answers
I induce tumor in mouse hindlimb using a very well known carcinogen and then at the onset of tumor initiation (visible bulge) I isolate the tumor, treat it with collagenase IV (0.2%, 2h) and plate the sieved cells in PLL-coated dishes. The cells are usually small and round and take 1-2 days to take their characteristic morphology, mostly fibroblast-like cell shapes. I have succeeded in isolating a mixed culture of cells using this protocol twice before but recently I repeated the same protocol thrice and failed in getting cells that take their characteristic morphology. I kept the cells for 5-7 days but they remained in their initial round phenotype. Several cells were adhered (I suspect its simply because of the PLL coated substratum), most cells were floating. I am completely amateur in primary cell culture and am unable to reason why I couldn't reproducethe same results with unchanged protocol, using which I could isolate cells 2-3 times before. I request for some suggestions.
Relevant answer
Answer
Its better if you re check the total  incubation time for collagenase. if it is too long,cell will not adhere to the surface. additionally, as lorenzo suggested,increase the serum concentration.    
  • asked a question related to Tumor Cell Culture
Question
2 answers
We have a breast cancer mouse model from which we want to extract the tumour, dissociate in single cells and then remove all non-tumour cells to have a pure population of tumour cells to inject in NOD0SCID mice.
Does anyone knows how to remove non-tumour cells?
Specific markers for fibroblast and others non-tumour cells...
Relevant answer
Answer
Can you instead of selecting the non-tumour cells, select for the tumour cells using for example ER status or any CD markers and FACS sorting?
  • asked a question related to Tumor Cell Culture
Question
4 answers
Does there is any drugs that can specific kill T cells without harm NK cells?
Relevant answer
Answer
Hi there,
If you work with human buffy coat, try to deplete T cells during ficoll gradient centrifugation with RosetteSep CD3 depletion kit (StemCell) and then negative selection. This way I got >98% purity at the end.
  • asked a question related to Tumor Cell Culture
Question
3 answers
I suspect that my knockout tumor cells are more susceptible to granzyme-mediated killing.  Is there an in vitro assay to incubate tumor cells with granzyme (and perforin?) to induce cell death?  I thought this might be easier before attempting to incubate my tumor cells with isolated NKs or CTLs.
Any references or protocols would be greatly appreciated!
  • asked a question related to Tumor Cell Culture
Question
5 answers
I want to draw the tumor clone evolution as follows, does anyone can help me which software i should use?
Relevant answer
Answer
Illustrator would be the easiest program to use for an image like this. It can do all of the things I'm seeing in the image you posted, and whatever image you make is easily scaleable without becoming pixelated-- good for publication. You can set the outlines and fills to be whatever color you want, add gradients, etc. Use layers to separate the different parts of the image so you can tweak them as needed.
 Lynda.com has a lot of tutorials that are free, and there are also some good ones on YouTube.
  • asked a question related to Tumor Cell Culture
Question
3 answers
I am working on the effect of fenofibrate on the tumor cell metastasis by using Balb/c mice as the animal model. Now I have a problem which it's the fenofibrate is not soluble in water, so I used stock DMSO as a solvent. The required dose of FF was100mg/1000g, so I will need to prepare this dose by using a high amount of DMSO because the solubility of FF in DMSO IS 15mg/ml. AlL mice were severely tired after being  injected with this dose! Btw the control group also were died after being injected with DMSO alone !!!! any suggestions please!
Relevant answer
Answer
Did you try methanol as a solvant: you can go MAX at 10% vol / vol injection with this solvant.
You can also reduce DMSO at an acceptable standard concentration for in vivo experimen, using ultrasonication, and injecting your "solution" (which is not) intra peritoneally. The i.p. route, which cannot be used in clinics (excepted in some cases, e.g. ascitic ovarian cancer), usually greatly help phamacologists...
Robert
  • asked a question related to Tumor Cell Culture
Question
1 answer
Hi,
I am ex vivo activating CD8 T cells for an adoptive cell therapy model. I would like to freeze down batches of activated cells to use at later timepoints and in other assays. How well do the activated cells respond after freeze/thawing and how long should they be cultured after thawing before I look for cytokine production?
Thanks,
Relevant answer
Answer
Human CD8 T cells are better frozen when they are not so activated (let's say 10-15 days after stimulation). Usually when you thaw them (use DNAse, that helps!) you lose around 50 % of the cells, so be aware (we always freeze vials of at least 10-50 million cells if possible). Usually, we look for cytokine production the day after thawing (so day 0 you thaw the cells, day 1 you stimulate them and day 2 you collect the supernatants to measure cytokine production).
I hope this will help you!
  • asked a question related to Tumor Cell Culture
Question
1 answer
Hello everyone, I am starting to work on extracellular vesicles (EVs) so recently I did an exosome isolation from cell culture media of neuroglioma cells by using ultracentrifugation protocol. I checked the EVs size with Nanosight and obtained several pics: two major at 165 and 215 nm and two other but smaller at 355 and 535 nm (might be aggregates). Does anyone know if this profile looks like something I could expect or is that too much high for a classical EVs profile? Usually the major pic is around 100nm in accordance with literature data but it seems to be the expected profile from exosomes (a small portion of EVs from cells). Is there any cut-off I should consider more related to all type of EVs?
  • asked a question related to Tumor Cell Culture
Question
2 answers
Dear all,
I constructed a mutant 4T1 cell line using CRISPR and implanted it into the 4th mammary gland of BALB/c recently. The tumor grows well at the same rate as WT in the first 12 days but 2 weeks later some of the CRISPR-modified tumor regressed while some remains small about 4~5mm long. I have some guesses but I really need you guy's assitance to help me out.
1. Maybe it's just a good thing, a phenotype. The presence of my gene ruins the immunosurvilence so the mutant cells are attacked and ruled out by the immune cells.
2. The contamination of Mycoplasm. But I think the contaminated cells cannot grow into a tumor at all. I don't have so much experience, so any information about this is appreciated.
3. The loss of cell viability. You know, it takes almost 2 months to finish the entire work of CRISPR. So I am afraid the 4T1 invasiveness will decrease. But for the same reason mentioned in 2, I think if viability is an issue the tumor will not appear at all. I need you guy's help to give me any experiences about this.
Thanks in advance for all your time and kind help, guys!
Relevant answer
Answer
Hi Thomas,
Thanks for your response! It's really helpful. It's a knockout single cell line where CRISPR introduces a one dp deletion at the DSB. I confirmed it by cloning the PCR pruducts into construct and by sequencing 4 colonies. Yes, tumor regression is one of our hypothesis of mutating this gene. Right now, I have one negative single colony from the CRISPR screening, so I plan to inject that one as a control. However, we haven't tested the potential offtargets but I have other two colonies with different mutation so I will inject them to confirm our phenotype. With these at hand, I still want to know what will happen if we use an aged cell line for xenograft. Do they not grow at all? Because only some of our KO cell line xenografts regressed but some still remain just smaller. I want to know can I rule out the possibility of passaging too many times by observing this phenomenon.
Thanks again and sincerely for your kind help!
  • asked a question related to Tumor Cell Culture
Question
9 answers
Dear all, 
I have tried to use 40:80% Percoll to isolate tumor-infiltrating lymphocytes from tumor tissue of CT26 BALB/c mice model. But when I used this method after collagenase digestion and centrifugation (300rpm, 25 min), no separated parts of any cell types were found. The tumor cells were still mixed in 40% Percoll layer which it should truly be sedimented at the bottom of the tube. I have tried to used the other concentrations of Percoll (75%, 70% and 65%) instead of 80% but it showed the same results. Does anyone have suggestions or experiences about this? I will very appreciate it.
Thanks!!       
Relevant answer
Answer
Instead of collagenase digestion you can simply finely chop and mechanically grind the tumor tissue in culture media and filter through 40um nylon mesh... then subject them to discontinuous Percoll gradient (we usually prefer 25-50-75 %). Ficoll 1077 is another feasible method. Unlike myeloid cells, lymphocytes can easily be separated from tumor tissue without digestion. One would like to avoid enzymatic digestion since it can also lead to shedding of cell surface markers.
  • asked a question related to Tumor Cell Culture
Question
5 answers
Hi all,
I have seeded and have been expanding a set of Organoids over the last 3 weeks. The Organoids are derived from Melanoma tumor. The method I have been using is to dissolve the matrigel in Cell recovery solution for 1-2 hours , spin down, remove Matrigel with Cell recovery solution, wash with PBS, add enough Matrigel for 3 times as many wells that were harvested, mix mechanically, distribute to the new wells. The main problem I have run into is that I am getting several wells that have very little cells transferred to them along with a few that have no cells at all. Half of the wells will have the majority of cells. Short of cutting back on the number of wells I expand to or painstakingly mechanically removing single colonies during transfer into the wells, I am at a loss. I know that if I can break up the masses that have developed over the weeks without damaging the cells I have a higher likelihood of expanding the cells to more wells. 
Does anyone have experience with this? Can you share a method you used?
Thanks.
Relevant answer
Answer
Hi Christopher, in our system we're using Detachin to disrupt spheroids after digestion of our Hyaluronic acid hydrogel. It works for most of cell spheroid and keep all surface markers intact for flow cytometry or re-seeding (97% of viability).
Don't hesitate to contact me for reference.
All the best
Chris
  • asked a question related to Tumor Cell Culture
Question
9 answers
I would like to establish a immunotherapy model by co-culturing human tumor cell lines with HLA-matched T cells. In particular I am interested in non small cell lung cancer (NSCLC) cell line Calu-1.The HLA phenotype for Calu-1 cells is as follows: HLA A10, A11, B15, Bw35. I need at least partially matched HLA-phenotype, i.e. matching the A-subclass. 
Is there any company that provide PBMCs which are characterized for HLA phenotype? 
Thanks,
Alessia 
Relevant answer
Answer
You can find the updated allele frequencies for the various HLA Alleles here (from NMDP in the US):
From there you can use the binomial theorem to calculate the probability of zero "successes" (i.e. HLA matches in your sample) across a certain number of "trials" (i.e. samples to screen):  http://stattrek.com/online-calculator/binomial.aspx
-Benjamin
  • asked a question related to Tumor Cell Culture
Question
8 answers
Hi, guys, I know it's not easy to culture primary cells because they have limited cell division.
But for us, we are only able to passage cells once or even worse.
1) Cells are either growing slowly or enter senescence easily.
2) After cell passaging, the morphology also changes .
We are culturing ovarian cancer cells, and use DMEM+EGF+FGF+B27 with 0~2% FBS.
I also attached some pictures here from your reference.
Does anyone  know what happen? and how to modify the culture condition to make the cells in proliferation and also maintain the morphology?
Thanks a lot!!
Relevant answer
Answer
Hi Jingwen - this is a very difficult question to answer.
Some cancers, whilst being very aggressive in the human, just do not thrive in 2D culture conditions (triple-negative breast cancers is one example, there are only a handful of these cell lines available because of this). Therefore it might just not be possible to establish a cell line from your primary cancer (so may not be anything that you are doing wrong).
Senescence can be triggered by a number of factors. Mainly it is caused  by telomere erosion but it can also occur due t "culture shock" (i.e. growth on plastic). However, saying that, you might want to check for telomerase activity in the cancer cells (if you can) just to verify that it is active.
For some primary cells growth in high glucose media can  hasten senescent onset - so  check which DMEM you are using (and if high glucose try a low glucose one).
You may also want to add some Insulin to you media (some ovarian cell lines require this).
You could also try increasing the FBS (i.e. 5-10%) with a portion of your cells to see if this helps.
When making the initial splits try to keep the density of the splits fairly high i.e. 1 in 2 split rather than a 1:4. This increases the labour but some cells are density dependent and die when they get lonely. Initially high splits may help them survive until they can adapt to the culture conditions
A last suggestion: (and this is "grasping at straws" a bit) save the media from the cells before you split them and when you seed the new flasks feed the cells with  a 50:50 mix of old media (from the cells pre-split) and fresh media. (this is often called "conditioned media" in papers and protocols.).
One last thing - primary cancers are likely to be very heterogeneous (containing cells with numerous different DNA mutations) and 2D culture conditions will always cause a "selective pressure" (so faster growing cell or cells that can tolerate growth on plastic will come to dominate the culture over time). Because of this you will always get some "phenotypic drift" and hence morphological changes over time (so make a master bank of cells early).
Sorry I can't be more help!
Gary
  • asked a question related to Tumor Cell Culture
Question
3 answers
I am setting up an experiment where I use magnetic beads to positively select for CD4+ T cells from a C57BL/6 mouse (with a tumour, either untreated or with treatment) spleen, followed by expanding the population. I then want to do a direct coculture with a cancer cell line, where I will conduct an ELISA on the supernatant to assess cytokine production (IL-12 etc.). My question is, by expanding the CD4+ T cells ex vivo (using antiCD3/antiCD28 and IL-2), would I be selectively expanding a specific subset (Th1 over Tregs for example), thereby skewing my results? If so, how might I go about avoiding that?
Relevant answer
Answer
Expanding your T CD4+ cells derived from the spleen of a tumor bearing mouse (not a xenotransplant, since the host is immunocompromised) is not expected to yield a progeny that is specifically directed against the tumor. This happens only if you expand the CD4+ cells in the presence of a tumor-specific antigen or in contact with the tumor cells.
  • asked a question related to Tumor Cell Culture
Question
6 answers
Dear all,
I'm looking for a tumor cell line expressing OVA protein on the cell surface so that anti-OVA antibody in sera can bind to the cell through OVA protein.
Does anyone has that cell line or know where can I get it from?
Thank you very much!
Relevant answer
Answer
Hi, I would warmly suggest you to use the RIP-mOVA mice which express a membrane-bound form of ovalbumin under the dictates of the rat insulin gene promoter, consequently in the pancreatic islets and thymus (also, aberrantly, in the kidneys and testis). Please find attached the original paper. If you're specifically looking for a tumor cell line expressing OVA on the cell membrane, i would ask either Dr. Marc Jenkins (Univ. Minnesota, Minneapolis, MN) or Thomas F. Tedder's (Duke University, Durham, NC) groups for a stable B16/F10 melanoma cell line expressing membrane-bound OVA (B16/F10/mOVA). The paper is attached for further information. I hope this will help you!     
  • asked a question related to Tumor Cell Culture
Question
1 answer
So my question is, whether somebody has experience with a similar set-up as described below (co-culturing target & effector cells for several days and then look at the killing efficacy of the allospecific effector cells) and if so, how he/she sets up the Assay and what control he/she uses.
So what I want to look at is:
I co-culture 40.000 irradiated tumor cells (suspension cells) for 5 days with 200.000 effector cells and administer different treatments.
After those 5 days (when I have allospecific cells) I wash the cells twice and I add again 40.000 tumors cells (not irradiated).
After 16 h I aquire the data.
As a maximum release control I use 40.000 tumor cells and kill them with 2 % Triton, and to be honest for the spontaneous release I don't really know what to use... 
So far I didn't get a stable readout and often the killing comes near 100 % or even more in comparison to the Triton wells, which of course makes sense, since in the other wells there are also dead effector cells and maybe some of the tumor cells from the co-culturing weren't killed until then...
I'm really thankful for any help!
Relevant answer
Answer
Hi Stefan,
I have several questions about your methods.
1) are you adding the non-irradiated tumor cells to wells that already have irradiated tumor cells and effector cells? 
If this is the case, your mixed bag of cells is going to be difficult to analyze by LDH.  You won't know for sure what cell is releasing LDH without lots of controls.  By the sound of it, you have effector cells that are releasing LDH.  Do you know the viability of your effector cells after 5 days as well as after the additional 16 hour incubation?  Can you purify your effector cells away from the irradiated cells?  With 3 cells mix, you would need to ask- what is the baseline LDH release of irradiated cells co-cultured with non-irradiated target cells? what is the baseline LDH release of irradiate cells co-cultured with effector cells? what is the baseline LDH release of non-activated effector cells with non-irradiated target cells?
2) What are your effector cells?  16 hours sounds like a long time to kill a target cell. Sounds like you are using T cells.  I would recommend doing a time course.  You may find that 4 to 8 hours gives a better result.
3) Have you considered other methods? Perhaps staining your target cells with a dye and then analyzing apoptosis by flow cytometery?
  • asked a question related to Tumor Cell Culture
Question
4 answers
Hello, I need to generate tumors in nude mice with the cell lines SK MEL 28 (melanoma) and MDA MB 231 (breast cancer), both human tumoral cell lines. How many cells do i have to inject intradermically?
Thanks, 
Martin
Relevant answer
Answer
When We are seeding subcutaneous cells we will have a heterotopic tumor. I always injected 6x10e6 tumor cells in 40ul of phisiological solution or culture medium in nude animals of 6 weeks old. In most of models, the tumor appears between 3 to 7 days after inoculation . In the three days after inoculation, papula decreases and then the tumor grows fast.
Good Look
Denise
  • asked a question related to Tumor Cell Culture
Question
4 answers
Can a tumour secrete a product but not expressing the protein?
E.g: Negative CE expression but serum marker CEA is high?
Thanks
Relevant answer
Answer
oh great answers .... :) thank u
  • asked a question related to Tumor Cell Culture
Question
3 answers
Hi all,
Having a ton of trouble infecting my primary mouse T cells from spleen with lentivirus. I regularly am getting 5-10% infection rate and many people seem to say this is within the "norm" but I would love to just get a little bit higher. I've tried everything - retronectin, protamine sulfate, dextran, polybrene, IL media - but nothing seems to get the rate up. Does anyone have any tips or seem to think this is going to be it for me?
Thanks!
Relevant answer
Answer
Hi Lauren,
One solution is to change the envelope of lentiviral vectors. I presume that you use the VSVg envelope which is not the best pseudotype to transduce primary T cells. We have good results for T cells tranduction with the MV envelope.
Regards,
Nicolas
  • asked a question related to Tumor Cell Culture
Question
2 answers
Good Morning.
I should evaluate the expression of membrane protein in a luminal cancer cell lines (HER2-negative, ER-Positive) derived by spontaneous tumor growth in transgenic or not mouse.
Do you have an idea if such cell line exists?
Thank you very much
Kind Regard
Lorenzo
Relevant answer
Answer
Hi Lore! How are you?
On this paper you can find all the breast cancer cell line classified for molecular subtype. 
Cancer Cell. 2006 Dec;10(6):515-27.
A collection of breast cancer cell lines for the study of functionally distinct cancer subtypes.
Neve RM1, Chin K, Fridlyand J, Yeh J, Baehner FL, Fevr T, Clark L, Bayani N, Coppe JP, Tong F, Speed T, Spellman PT, DeVries S, Lapuk A, Wang NJ, Kuo WL, Stilwell JL, Pinkel D, Albertson DG, Waldman FM, McCormick F, Dickson RB, Johnson MD, Lippman M, Ethier S, Gazdar A, Gray JW.
See you soon!
  • asked a question related to Tumor Cell Culture
Question
4 answers
I'm looking for one Ab targeting CD44-HA interaction and one that doesn't. The aim is to use the Abs to disrupt CD44 adhesion/signaling in an in vitro tumor cell culture, eventually leading to further in vivo studies adminiestering Abs to mice. Alternatively, how feasible would be the use of hyaluronidases or HA oligosaccharides to inhibit CD44-HA interaction? In the case of HA oligosaccharides, which one(s) would be best as inhibitors?
Relevant answer
Answer
CD44 standard isoform withour variable exons (predicted M.W. is about 75kDa) is known to highly ind to hyaluronic acids as compared with CD44 variant. As mentioned above, clone IM7 is best recognized to block the interaction of CD44 and ECM.  
  • asked a question related to Tumor Cell Culture
Question
3 answers
I am having difficulty extracting RNA from fresh frozen tumor samples. I am having good extractions on about 10/18 samples. I am following the protocol exactly. I have recently noticed- when spinning the sample down after breaking up the tissue, I see a pellet, but I also see a filmy substance floating around the top of the supernatant. These samples are having 0 yield. Does anyone know what this is? Or advice on using this kit? 
Relevant answer
Answer
        You may try Trizol  to extract total RNA from your tumor tissue.From our experience,the RNA mini kit extract less RNA than the method of Trizol.You can have a comparison.
  • asked a question related to Tumor Cell Culture
Question
3 answers
how to monitor tumor propagation in lung after inoculation of cell lines in the lung. Is there any simple and reliable method by which it can be check the gradual increase in the size of tumor in lung. if any than please share the methods with references..... 
Relevant answer
Answer
Dear Birendra.
Do you have any in vivo Imaging System to visualize fluorescence or bioluminsicence?
Otherwise would be quite difficult to track (without killing) the metastatic process
  • asked a question related to Tumor Cell Culture
Question
2 answers
I was looking in the literature for prostate cancer C4-2 cells (CRPC) mouse xenografts that used enzalutamide, but I wasn't able to find any such papers. Could any of you help me with this? Thanks in advance. 
Relevant answer
Answer
Thank you very much. I greatly appreciate your help.
  • asked a question related to Tumor Cell Culture
Question
1 answer
We tried the model following the the current protocols in immunology: The TRAMP mouse as a model for prostate cancer. Unable to get tumors growing.
Relevant answer
Answer
Hi, take a look at these papers, if you've not already done before. In the last paper that I suggest you, 3x10^6 cells were implanted s.c. in the left flank of 20 weeks of age B6 mice. To me, they are a huge amount for a s.c. injection. Anyway, take into great consideration the age of your mice: it seems that as older they are as better is the TRAMP-C2 cell proliferation in vivo. Good luck!
  • asked a question related to Tumor Cell Culture
Question
8 answers
hepg2 cells looked floated and did not attach after 48 hours of revival process. 
Relevant answer
Answer
Dear Anam,
Maybe your cells are death for problems during freezing and/or thawing processes. 
Some aspects are important:
- Do not vortex, bang the flasks to dislodge the cells or centrifuge the cells at high speeds.
- DMSO percentage is critical, during cryopreservation with DMSO, it is natural that some of the preserved cells get stressed and ultimately dead and on thawing these dead cells comes in form of cell debris. If glycerol is used in the freezing medium, do not store it in light conditions (glycerol is gets converted to acrolein, which toxic to cells),
- During thawing process, make sure that you thaw the frozen cells quickly, but dilute them slowly using pre-warmed growth medium before plating.
Other different possibilities are:
- Fungi/yeast contamination: if you see filamentous structures and turbid media.
- Bacterial contamination: cultures usually appear cloudy (i.e., turbid), sometimes with a thin film on the surface. Also, sudden drops in the pH of the culture medium are detected. Under a low-power optical microscope, the bacteria appear as tiny, moving granules between the cells, and observation under a high-power microscope can resolve the shapes of individual bacteria.
-Mycoplasma contamination: mycoplasma can alter the behaviour and metabolism of the host cells in the culture. The best way of detecting mycoplasma contamination is by testing the cultures periodically using DAPI staining, fluorescent staining (e.g., Hoechst 33258), microbiological assays, PCR, ELISA, PCR, immunostaining or autoradiography, 
Finally, if you consider this answer appropriate, please upvote it using the green up arrow click.
Best regards,
  • asked a question related to Tumor Cell Culture
Question
6 answers
If so what factors do they need?
Relevant answer
Answer
Hi,
Yes, they need a lot actually.  EGF, Hydrocortisone, cholera toxin, Horse donor serum and insulin. I put these inside DMEM/F12 mixture and they survive and grow well under these conditions. 
Best,
Yunus
  • asked a question related to Tumor Cell Culture
Question
2 answers
We need a clear protocol to induce liver cancer in rats, in a shorter time and less expensive. 
Thank you in advance for help
  • asked a question related to Tumor Cell Culture
Question
3 answers
We are interested in imaging co-cultures of tumor cells and immune cells, but would like to culture them within the same dish. Is this possible? Or is there a fundamental reason all the co-culture papers use transwells?
Relevant answer
Answer
The major reason why tumor cell and immune cells are cultured in the same dish with the transwell chamber is that both types of cells are influenced each other. For example, immune cells secrete CXCL12, which binds to CXCR4 expressed in cancer cells. To investigate the paracrine secretion of the cytokines which play crucial roles for the crosstalk bewtween tumor-entrained immune cells and tumor cells, the usage of the transwell chamber to prevent the direct cell-to-cell contact seems to be essential.  
  • asked a question related to Tumor Cell Culture
Question
1 answer
I want to do an experiment where I take conditioned media from one plate of cells and and add it to a plate of cells with a gene knockout and see if there are changes in phenotype.  Can someone run through a protocol on how to do this.  Do I add serum in the plate while I am conditioning the media but no serum to the plate of cells for which I will be adding the conditioned media to or no serum at all? These are lung tumor cells which normally take RPMI with 10% FBS.
Relevant answer
Answer
Conditioned medium is likely to include some kinds of cytokines or growth factors which render the phenotype of the recipient cells, so that you do better use RPMI without 10% FBS. After all, serum is rich in not only nutrition and amino acids but also growth factors and cytokines!!! The presence of the FBS in the medium of recipient cells may mask the real influence of the conditioned medium.
  • asked a question related to Tumor Cell Culture
Question
7 answers
I want to find the migration difference between transfected and non-transfected tumor cell lines, I'm considering three brand migration assays: Falcon, Corning and BD Biosciences. For migration assays, the membrane pore should be 8um. For Corning, there are different membrane types, like polyester (PET), polycarbonate (PC) and collagen-coated (PTFE). Besides, it seems there are some relationship among Falon, Corning and BD Biosciences, it makes me confused. Briefly, who can tell me the differences among these different membrane. It would be appreciated very much?
Relevant answer
Answer
My response is coming in late but I see that many RG members were / are still interested by this question.
We tried to partly respond to this question by some research or review articles here attached.
Best regards
Robert
  • asked a question related to Tumor Cell Culture
Question
6 answers
I recently ran an experiment where I divided a mouse tumor into several wells of a 96-well plate. Each well was stained with a different panel of antibodies. In only one of the wells, I got the strange pattern you see on the far right, where my events formed a distinct diagonal line with a slope much smaller than expected. If I gate on the events, they are abnormal in the other plots as well. Any help you can give would be great, thank you!! 
Relevant answer
Answer
Hi Nikki,
Did you run the samples in the order of your displayed results (left-good; middle-bad; right-worst)?
Mouse primary samples are very 'dirty/sticky' to the FACS machine. My feeling is the events you recorded for the third sample (right) are accumulated dirt rather than true events. Sometimes, I do get the results you showed when I run a lot of samples. In that case, I will run cleaning solution/FACS buffer until no 'events' appear on plot before I proceed to the next sample. If you gate the events for the third sample on FSC-A vs. SSC-A, they probably appear at the corner of the plot or as a streak. The vital stain will also help to distinguish if they are true living cells.
  • asked a question related to Tumor Cell Culture
Question
4 answers
I work on different cancer cell lines (breast, liver, colon) , contamination occurs either the flask becomes turbid or fungus grows on the inside off the flask, i clean the incubator with chlorine and 70% alcohol, i use antimycotic and filter the complete media how shall i get rid of that aggressive contamination please?
Relevant answer
Answer
Dear Salma,
You may increase the amount of the antibiotic in you culture media but you need to be careful many cells do not like this, though. On the other hand, If you have a chance of course UV procedure is really help you to get rid of many germs inside from your hood. If your hood close to any air flow, try to decrease this as much as you can. 
Have you ever checked the flow rate inside through your hood. Your filter may blocked and then it is most probably can not work properly. I strongly recommend you to check these.
Best,
Yunus
  • asked a question related to Tumor Cell Culture
Question
6 answers
I work with B16-F10 melanoma cells, and I have a huge problem with infections. I think, that this infection is caused by some kind of yeast, but I'm not sure. Have anybody an idea? Thank you!
Relevant answer
Answer
Hi Anna, only just found this question.  Yes, the trouble is that the pictures are not in focus.  If you are still having the problem, is it possible for you to provide additional pictures that are in focus, also higher magnification maybe?  so that we can at least see the cells and compare their size with the floating material?  It does look like yeast, as far as the focus and low magnification permit a guess.  Do the clumps get bigger with time?
If it is indeed yeast, your best option is to start again, as B16F10 cells are easy to get hold of, and maybe you have some frozen.  Then just be very careful with your culture technique.  Your own hands are the most likely source, e.g. eating bread or fruit before going in the lab.  Wash well, spray everything with 70% ethanol, etc.
  • asked a question related to Tumor Cell Culture
Question
3 answers
i have treated a tumor cell line using a plant extract and terminated the treatment at different times (eg: 1 hour, 2 hours, 4 hrs and so on) and estimated some enzyme activities and observed that the enzymes decreased up to a certain time, increased again and then decreased again....  how do i interpret this and what exactly is happening??
thank you
Relevant answer
Answer
Hi Lairinzuali,
Well, the funny point is that your plant extract makes the enzyme activity you follow versus time to decrease then to increase, then to decrease again at a longer time of incubation.
A possible explanation is that, as suggested by Robert, a first component in your plant extract "quickly" inhibit the enzyme whom activity you look at. But this component does not act for long (why's that ... its stability??? do you heat the incubation mixture?). Then, another component, acting much less "quickly" begins to act at a longer time and, this time, being more stable, decrease largely yhe atcivity of the enzyme you look at.
A possible further explanation is that the second decrease, that appearing after the inetrmediate re-incrase of the enzyme you follow, is due to a component that is produced during your incubation by some enzyme of the tumor cells transforming a compound of your plant extract to a derivative that is toxic to the enzyme activity you look at. This is OF COURSE very interesting, this hypothesis paths the way to inetresting experments and results.
Best regards
Philippe
  • asked a question related to Tumor Cell Culture
Question
3 answers
Hi!
I trying to block bcl-2 in bcl-2-overexpressing cell culture model. In my conditions ABT take effect at 2 mkM - cells turns apoptosis and dead. But ABT-199 active at nanomoles. What concentration you use and which way to confirm bcl-2 inhibition?
Thanks!
Relevant answer
Answer
Hi Serguei,
I agree with Kamil answer. You need to determine IC50 in your cell culture using some assay to measure citotoxicity. In some cells IC50 for ABT-199 concentration can vary between micro and nanomolar. You must to do the citotoxic assay to know what concentration is optimal for your cells.
 Good luck,
Joan
  • asked a question related to Tumor Cell Culture
Question
9 answers
I have prepared tumour spheroids with U87 MG cells and I am trying to get fluorescence images using confocal microscope. However when I placed tumour spheroids between cover slips and microscope slides, I think they get squeezed and I lose the 3D shape of the spheroids. Do you have any suggestion to prevent this? Thank you.
Relevant answer
Answer
Here is a paper describing a live cell imaging system that was developed by some of my coworkers. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3826880/   Our lab also provides foundry services to customize such devices. http://www.vanderbilt.edu/mfc/
With microfluidic devices, you are able to image your cellular system in a controlled environment that few other platforms are able to achieve. 
  • asked a question related to Tumor Cell Culture
Question
2 answers
These cell lines along with available xenografts and other factors along with other biomarkers ( if you have any it would be appreciated)  will be used to build a preclinical model for high risk Wilms tumors.  The development of assays specific to this model will result in the ability to do novel cell line and animal testing which could potentially lead to clinical trials with new compounds for children with these disease.
Relevant answer
Answer
sorry no
  • asked a question related to Tumor Cell Culture
Question
3 answers
i want to count gamma h2ax foci in the tumoral cells,which software is suitable?
Relevant answer
Answer
There are many (expensive) softwares that can detect nuclei and count particles in it. But if you plan to do this once and move on, you can use Image J which is free. There is a fair bit of information on how to use Image J for foci counting on the internet and some have written macros for this purpose you can use!
We use the ScanR software which comes with a high throughput microscope.
Good luck
Pegah
  • asked a question related to Tumor Cell Culture
Question
4 answers
i want to choose the concentration of the drug that has the most efficacy, is flow cytometry of cell cycle a suitable test or not?
Relevant answer
Answer
Hi Fereshte,
I have not worked with PARP inhibitor before. But, flow cytometry is probably the best method to measure cell cycle quantitatively. A DNA staining will allow you to visualize the cell cycle profile. Options are as following:
1. PI + RNase
2. Vybrant violet
3. Hoechst 33342
You can also combine DNA and BrdU staining to confidently distinguish S phase cells.
  • asked a question related to Tumor Cell Culture
Question
4 answers
Hello everyone,
our problem is non formation of tumors in xenograft expt. We are using female nude mice.
We injected HO15.19 (rat1 a) cells expressing Myc as follows:
1. First female nude mouse: we injected HO15.19 (rat1 a) cells expressing WT Myc : 2 million cells+matrigel into one flank and 3 million cells in PBS into another flank.
2. Second female mouse: we injected HO15.19 (rat1 a) cells expressing mutant Myc : 1 million cells in matrigel into one flank and 5 million cells in PBS on another flank.
It has been 22 days and we still see no trace of tumors. Can anyone help troubleshoot the problem. I am a novice in the field and had not done tumorigenesis studies before?
Relevant answer
Answer
Hi Amirali, thank you for the advice. We don't have NOD/SCID mice in our animal facility. We have to use NUDE mice. I mixed the cell pellet well before injection. However I will check the needle issue as suggested by you and Robert walter.
  • asked a question related to Tumor Cell Culture
Question
4 answers
I try to figure out some easy method to transfer spheroids from hanging drops from a petri dish lid into 96-well plates. So I had the idea to generate the tumor spheroids directly on the lid of a 96 well-plate directly instead of generating them on petri dish lids and to centrifuge the plate after spheroid initiation. Does that work to centrifuge the spheroids from the lid directly into the well? 
Relevant answer
Answer
Thanks for your rapid answers! I think I will try to initiate spheroid growth directly in the wells now. Hope that works, because every cell line behaves totally different in 3D-culture
  • asked a question related to Tumor Cell Culture
Question
3 answers
I generated them with hanging-drops and transferred them into 48 well-plates for further cultivation. But after 8 days they didn't look really solid and compact anymore. And when I tried to transfer them into a falcon tube for other experiments, they actually dissolved during pipetting.
Relevant answer
Answer
Thank you vor your answer Isabelle and Gabriel.
I tried out another cell line, which works now. I got compact spheroids, which were 'easy' to transfer and did not dissolve.
  • asked a question related to Tumor Cell Culture
Question
5 answers
I am culturing liver cancer cell line in serum free tumour sphere medium (DMEM/F12 + EGF + bFGF) and culturing them for at least 7 days to enrich for cancer stem cells. Do I need to change the medium everyday?
I have a concern that this will disturb the sphere formation as I will need to centrifuge and I am afraid that during the resuspension in fresh media will affect the spheres. I am culturing them in 6-well ultra low plate by the way.
Relevant answer
Answer
Hello,
You really don't need to change the medium every day, but it depends on the amount of spheres that are current in the well. We've cultured cancer stem cell spheroids in Petri dishes, the centrifugation did not affect their stability or behaviour. And the addition of EGF and bFGF is unnecessary.
  • asked a question related to Tumor Cell Culture
Question
2 answers
I aim to perform a study about circulating tumor cells in colorectal cancer and to culture CTC s
Relevant answer
Answer
after a step of enrichment to remove other blood cells I've just learned of a machine that sort rare event based on surface markers. DEPArray of Silicon Biosystem
  • asked a question related to Tumor Cell Culture
Question
3 answers
How can culture cells from fresh tissue of breast cancer?
Relevant answer
Answer
From my experience I know that the long trypsinization destroys the cells. I propose 15 min trypsinization no longer but repeated several times. After such time, decant the supernatant, add a few drops of serum and put in the fridge. To the remaining tissue add trypsin and repeat the procedure. The supernatants you can mingle to the same Erlenmeyer flask. If the tissue is very hard, after a thorough mechanical fragmentation you should use collagenase, but also no longer 20 minutes and proceed like in the case of trypsin. Unfortunately Serum does not deactivate the collagenase and you must quickly decant the supernatant, add a lot of medium and centrifuge. I suggest 2 times. A tedious task, but I do not know collagenase inhibitor..
  • asked a question related to Tumor Cell Culture
Question
10 answers
Hello everyone,
Does anyone know how I can isolate and transfer a spheroid from one plate to another one? 
how can I wash a spheroid with PBS (e.g. Can i use centrifugation method? and how can I cut the spheroids to see their cross-sections?
Thank you.
Relevant answer
Answer
I also use MDA. They grow up perfect in Matrigel instead of agarose. No attachment in well is observed.
-Marilena-
  • asked a question related to Tumor Cell Culture
Question
6 answers
Looking at tumoursphere formation in a paper which uses LM2 (triple-negative) and MCF7 (oestrogen-receptor positive) cell lines. Control comparisons show a much lower tumoursphere formation - is this because the culture medium did not contain any oestrogen supplementation? No experience culturing cells / whether this is something you can accept as an inherent difference between the cells?
Relevant answer