Science topic

Cancer Cells - Science topic

Explore the latest questions and answers in Cancer Cells, and find Cancer Cells experts.
Questions related to Cancer Cells
  • asked a question related to Cancer Cells
Question
2 answers
I have been working with MCF-7 recently. But I think these cells are different from the MCF-7 cells that I worked with a few years ago. Furthermore, even I don't know what these round cell on the culture are? Are these dividing MCF-7 cells? Sometimes these round cells become very numerous (especially in high confluency) and sometimes they become few. I took some photos from my cells under the microscope at different magnifications and different cell confluences.
Does anyone have any experience with these cells? what are these round cells? Are these MCF-7 cells good for working?
Thank you in advance.
Relevant answer
Answer
Dear Rizky Clarinta Putri,
Thank you for your answer. The round cells don't move. But I think those cells were high passage cells, So I replaced them with other MCF-7 cells. These new MCF-7 is Ok.
  • asked a question related to Cancer Cells
Question
2 answers
"I am currently researching the applications of nanotechnology in the medical field, specifically for cancer treatment. I am interested in understanding the various methods being explored to use nanomaterials for targeting cancer cells more effectively, minimizing side effects, and improving treatment outcomes. Could anyone provide insights or recent studies that explore these advancements, or suggest relevant papers and resources on this topic?"
  • asked a question related to Cancer Cells
Question
3 answers
I am working with blood and A549 cancer cell. My experiment design is where I will stain the cancer cells with CellTracker CMTPX, and also stain the blood (lysed blood sample with a few residual RBC) with hoechst separately and then spike the stained blood sample with the stained cancer cells.
There are no issues with the dye used for cancer cell but the hoechst always stains the cancer cells a dull blue as well after I spike the blood with cancer cells. I have tried to wash the stained blood cells 5 times in PBS to prevent this stain contamination but it keeps happening. Is there any way to prevent this?
Relevant answer
Answer
I think the stained blood sample has residual Hoescht and then since Hoechst is cell-permeable the cancer cells stain with residual Hoechst. A few follow-up questions to help.
What is the concentration you are using?
What is the purpose of staining with Hoechst? If it is just to stain the DNA (does not matter if it is dead or alive), then I do not see an issue with the blue color.
Hoechst is cell-permeable. If you are using it on live cells, you would want to use something else that is not permeable without fixation or if the cell is dead, such as Propodium Iodine (PI). What color is your CellTracker? Make sure that the PI and CellTracker excitations do not overlap.
  • asked a question related to Cancer Cells
Question
2 answers
I have 3stock 1. MDA MB 231, 2. 4T1 and 3. MCF7 so which process should I follow for getting better cancer stem cell in these stocks. Hanging drop method, long exposure in very low concentration of regorafenib drug or rapid passage?
Relevant answer
Answer
The choice of method depends on the type of research question. Each system has its own advantages and disadvantages, and you must carefully consider which system is best suited for your application.
Hanging drop method is the most common method. In the hanging-drop method, cells in the culture medium suspension are placed on the underside of petri dish lids. The cells accumulate at the tip of the drop, spontaneously aggregate, and form spheroids. However, this method is limited by the difficulty in exchanging the culture medium, thereby preventing cells from being cultured for more than 3 days. Furthermore, cells within the spheroids are subject to diffusive oxygen and nutrition supply. Additionally, it is critical to prevent the necrotic damage to the cells at the core of the spheroid and to control the size and composition of spheroids.
On the other hand, if you use sphere-formation assay, it will provide useful tool to assess the stem cell population residing in cancer cell lines. The advantage of using the sphere-formation in vitro assay is to isolate, propagate, purify, and amplify specific population of cancer stem cells. It enables studying stem cells at different stages of their formation (at different generations) and detecting markers of their signaling pathways. It also solely depends on the functional intrinsic property of stem cells in forming a complex structure in a 3D environment, namely self-renewal ability and differentiation potential.
In the sphere formation assay, cancer stem cells are isolated using anchorage-independent sphere culture. Cancer stem cells can grow on ultra-low attachment plates that are coated with a layer to inhibit the attachment of cells. When cells are grown in serum-free and non-adherent conditions, cancer stem cells can survive and clonally expand to form spheres, whereas differentiated tumor cells undergo apoptosis due to their anchorage dependence.
Below attached links will be helpful.
Best.
  • asked a question related to Cancer Cells
Question
2 answers
Is it somehow related to tumour microenvironment or some other reason behind this.
Relevant answer
Answer
Hello Ananya,
The reason is as follows.
High T cell densities may mediate effective killing. High T cell densities enable high contact frequencies and multiple T cell–tumor cell encounters. This may increase the likelihood of multiple T cell simultaneously attacking a single target cell (cancer cell). In addition, cytotoxic cytokine release, including interferon-γ and tumor necrosis factor, is positively associated with high density T cell infiltration.
High T cell density which can be called swarming enables efficient apoptosis induction by favoring serial perforin-dependent hits, preferentially by different T cells whereas the ineffective killing at low T cell density largely relies upon single encounters.
A successful T cell effector function correlates with high local T cell densities. If a 1:1 ratio for T cell : cancer cell is taken,
the T cells will form predominantly short-lived interactions with target cells (cancer cells) which rarely result in direct apoptosis induction.
The individually ineffective T cell contacts induce sublethal damage, which becomes integrated over time in the target cell until
apoptosis is induced. Through such a multi-hit mechanism, T cell induce tumor cell death when density is high, whereas tumor cells repair sublethal damage and survive when T cell density is low. This mechanism of “additive cytotoxicity” explains how individually ineffective interactions become effective at high T cell density.
Thus, in apoptosis assay, the T cells have to be higher in density than the target cells ( cancer cells).
Best.
  • asked a question related to Cancer Cells
Question
3 answers
I have to extract DNA from 1.5 ml of fresh cancer cells (fibroblasts cultured in MEM), but before that i need to count the cells. I need to do both on the same day. My question is how long can i store cells in MEM in an eppendorf tube while i count cells on the side? Can i place the eppendorf back in incubator for the time being?
Note: I will be using 1.5 ml cell culture for DNA extraction and 100 micro litre from it will be used to count cells.
Relevant answer
Answer
Hello Sabawoon Nisar,
Please use 100ul from the cell suspension for counting and centrifuge the remaining culture (1.4ml) at 700-800 rpm to pellet down the cells. Discard the supernatant and store the cell pellet in ice until you start the DNA extraction protocol. Cell counting should not take more than 10 mins.
Best.
  • asked a question related to Cancer Cells
Question
1 answer
I'm not talking tumor cells. I'm talking individual cells that are either cancerous or pre-cancerous. Does light refract inside of them differently than their healthy counterparts? Could we measure this somehow? I'm thinking some sort of epi or endothelial tissue cancer cell, such as the cervix lining.
Relevant answer
Answer
If the refractive index of cancer cells and their healthy counterparts are different, then that would change the intensity of light refracted and reflected. The cancer cells have a larger refractive index because while cancer is occurring in a cell, the cell reproduces many thicker organelles and cytoplasmic structures. This would make the cell denser than the normal cell, thus reflecting more light than the normal cell.
For instance, the normal cell and skin cancer cell refractive index are 1.36 and 1.38, respectively, while the normal blood cell and cancer blood cell refractive index are 1.376 and 1.39 respectively.
The optical probe technique based on light-scattering spectroscopy may be used to detect pre-cancerous and early cancerous changes in cell-rich epithelia. From the stage of dysplasia to carcinoma in situ, cancer cells alter the epithelial-cell architecture, with the nuclei enlarged, crowded and hyperchromatic. These warning signs of the size distribution and chromatin content of epithelial-cell nuclei can be measured by light-scattering spectroscopy as an indicator of pre-invasive neoplasia.
There is a growing interest in extracting the microscopic properties of cancerous and normal cells by means of their measured optical parameters due to the differences of microstructure between these two types of cells.
The article attached below may be helpful.
Best.
  • asked a question related to Cancer Cells
Question
6 answers
After receiving MCF-7 cells from NCCS (India), I supplemented the flask with growth media (DMEM in 1% penicillin & streptomycin with 10% FBS). I then passaged MCF-7 cells. After 3 days, the cells look as per images attached. Both the images are taken from the same 25cc flask.
Do the cells show the expected morphology ?
Thank you in advance for your replies !
Relevant answer
Answer
This looks like a mixture of MCF-7 cells that have and have not attached to the flask. Non-attached cells will be rounded up and attached cells will be flatter. As they become more confluent, attached MCF-7 cells will take on a ”fried egg” appearance. I suggest that you let the cells grow to increased confluence to see how they behave. There are lots of pics of these cells in the old literature. Keep in mind that these cells express estrogen receptor and are estrogen sensitive. Unstripped serum and phenol red containing DMEM contain estradiol and/or other estrogenic chemicals that stimulate cell proliferation to varying levels. You can test the cells for these responses, which are inhibited by anti-estrogens (tamoxifen, Raloxifene). Hope this helps.
  • asked a question related to Cancer Cells
Question
1 answer
Drug resistance is a significant challenge in cancer treatment, and this question explores how genetic modifications might overcome this barrier.
Relevant answer
Answer
I am not an expert in this field, but I am very interested and have researched to find an answer. I received some assistance from tlooto.com for this response. Could you please review the response below to see if it is correct?
Genetic engineering can be used to prevent drug resistance in cancer cells by targeting and modifying specific genes associated with resistance mechanisms. Techniques such as CRISPR/Cas9 can knock out or edit genes related to drug efflux pumps, DNA repair enzymes, or survival pathways, reducing the cells' ability to resist treatment [1][3]. Additionally, targeting cancer stem cells (CSCs), which are often implicated in drug resistance, can enhance therapy efficacy. CRISPR/Cas9 can be utilized to target critical genes involved in CSC maintenance and drug resistance [1][4]. Combining gene editing with traditional therapies can address both drug resistance and tumor recurrence [2][5].
Reference
[1] Saha, T., & Lukong, K. E. (2022). Breast Cancer Stem-Like Cells in Drug Resistance: A Review of Mechanisms and Novel Therapeutic Strategies to Overcome Drug Resistance. Frontiers in Oncology, 12.
[2] Chen, Y., & Zhang, Y. (2018). Application of the CRISPR/Cas9 System to Drug Resistance in Breast Cancer. Advanced Science, 5.
[3] Dashtaki, M. E., & Ghasemi, S. (2022). CRISPR/Cas9-Based Gene Therapies for Fighting Drug Resistance Mediated by Cancer Stem Cellsc.. Current gene therapy.
[4] Dhanyamraju, P. K., Schell, T., Amin, S., & Robertson, G. (2022). Drug-tolerant persister cells in cancer therapy resistance.. Cancer research.
[5] Housman, G., Byler, S., Heerboth, S., Lapinska, K., Longacre, M., Snyder, N., & Sarkar, S. (2014). Drug Resistance in Cancer: An Overview. Cancers, 6, 1769 - 1792.
  • asked a question related to Cancer Cells
Question
4 answers
Hi,
I am working on culturing Jurkat cells with different types of cancer cells to see their ability to kill cancer cells in different conditions.
It's my first time doing viability tests in adherent cells and I am using MTT assay. I am having a problem with the control well containing only Jurkat cells also has the signal even after I wash it twice with PBS before adding MTT solution. I could try washing it more but I notice that some of my cancer cells start to detach after the second wash
Any suggestions for what I could do to improve my experiment? or I should use other types of viability tests? I've read publications that use crystal violet or CCK-8 assay but I am not so sure if it will even fix my problem
Best regard,
Relevant answer
Answer
A few thoughts on MTT and similar assays. Cell viability is a critical quality attribute that is measured throughout cell biology studies. This is most critical in GMP manufacture of cell therapeutics. Please see "Assessment and comparison of viability assays for cellular products" published in Cytotherapy 26 (2024) 201 209. While MTT assays are often published as viability assays, one can argue as below that they do not address this cellular parameter. You might find the above reference helpful.
The MTT (3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide) assay is based on converting MTT into formazan crystals by living cells, which determines mitochondrial activity. Since, for most cell populations, the total mitochondrial activity is related to the number of viable cells, this assay is used to measure in vitro cytotoxic effects of drugs on cell lines or primary patient cells. Note that it is not a measure of cytotoxicity but rather an assessment of cell proliferation. Further, it does not analyze the percent or frequency of viable cells as a trypan blue or PI analysis would provide.
The XTT assay cannot measure cytotoxicity but rather measures cell proliferation or a lack of cell proliferation. XTT/MTT assays are used to measure cell proliferation in response to growth factors, cytokines and nutrients. They can also be used to measure the activity of growth-inhibiting agents such as inhibitory antibodies i.e. a measure of cytostatsis. As such, it does not analyze the percent or frequency of viable cells, such as obtained with a trypan blue or PI analysis. Nor does it provide an analysis of cytotoxicity, such as that measured with a 51Cr release assay.
These assays can measure a decrease in proliferation (cytostasis), but labeling this a loss of viability or cytotoxicity would be incorrect.
  • asked a question related to Cancer Cells
Question
9 answers
I would like to investigate the effect of bacterial secretome on cancer cells.
How do you prepare conditioned media from bacteria??
conditions- time points - media
I am looking for the best way to prepare the media in order to recapitulate the physiological conditions in vivo.
Thank you all.
Relevant answer
Answer
Ioanna Nikdima I do not have expertise in this field but the attached research article used the bacterial media (LB) for the preparation of conditioned media. Please check the related papers in this field.
  • asked a question related to Cancer Cells
Question
5 answers
I am inexperienced in conducting in vitro studies, an area outside my area of expertise. I have been training in basic cell culture technique for the past 2 months. Currently, I am evaluating the synthesized compounds at two different concentrations (10, 100 uM). I have noticed that some compounds exhibit higher % inhibition at lower concentrations compared to higher concentrations. This raises the question: "Is this scenario feasible?" If so, what could be the underlying reason?
Please enlighten me in this regard.
Relevant answer
Answer
I am not an expert on this phenomenon, but I have seen it myself with my own data. Let's discuss in more detail using some figures. These figures are obtained from my own paper that can be accessed from links below.
Let's look at the figure with the name 1.jpg. When looking at panel A, B, and C that represent continuous monitoring of cell viability throughout 72 hr of observations, you will notice that the compound applied at the highest concentration (red line) does not always have the lowest normalized cell index as a measure of cell viability. This is in part probably contributed by the rapid killing action of the said compound that did not allow enough time to be completely devoid of cell attachment. For your information, the machine I used here relies on cell attachment to estimate the overall health of adherent cells. In this case, however, you might say that the difference is minor, so we can safely assume that the cell viability values are probably at the same level or range.
Now, let's see the figure with the name 2.jpg. When looking at panel B with the label "Monolayer", you will notice substantial concentration-independent effect, e.g., with compound PI at 3.13 ug/mL, the growth inhibition is > several other higher concentrations. In this assay, I used an ATP-measuring luciferase assay for 3D culture. I did not think that the reagent was that incompatible for monolayer because for 3D culture, it is modified to have an enhanced penetration and lysis, which should not differentiate much between monolayer and 3D culture. However, as you can tell, it is more concentration-dependent when looking at the Spheroid (3D) model.
So, from these observations, you can probably conclude that the difference response you saw might be contributed by:
  • type of cell viability assay, i.e., mitochondrial activity, cell attachment, protein content, ATP content, etc.
  • growing mode of cells, i.e., monolayer, spheroid
  • mechanism of actions of compound that occurs at a very specific range of concentrations
  • differential range of concentrations inducing cytostatic (stopping growth without killing) or cytotoxic (killing)
The combination of these factors is likely the reasons for this phenomenon. I would say it is not uncommon; this type of data is just less interesting and people mostly could not explain exactly its occurrence.
  • asked a question related to Cancer Cells
Question
3 answers
The cancer cells (NCI-H1975) & (HCC827) NSCLC is not growing, we have rule out in incubator factor, the media. What else can effect the cell growth?
Relevant answer
In our lab, we observed that NCI-H1975 cells sometimes exhibit low growth rates. To address this, I recommend using a medium containing 20% Fetal Bovine Serum and changing the medium every two days. This approach should enhance cell growth. Additionally, ensure to monitor the pH of your medium and test for mycoplasma contamination as previously advised.
  • asked a question related to Cancer Cells
Question
9 answers
I have no idea why my orthotopic mouse models have failed.
I have used gastric cancer cell lines, and I injected the cells into a mouse stomach.
Here is my procedure and a result photo as a failed example.
1. Detach gastric cancer cells with 0.25% trypsin, then wash the cells with PBS
2. Centrifuge for 3 min, 1200 rpm at room temperature
3. Suction the trypsin and PBS, then put 1 mL of PBS
4. Get 1 x 10^8 cells in 1 mL of PBS, which is an example, counted by a cell counter
5. Transfer 100 uL of cell suspension to get 5 x 10^6 cells for 2 mice
4. Centrifuge for 0.5 min
5. Remove the PBS and put 15 uL of Matrigel and 15 uL of PBS
6. Suspend the 30 uL of gastric cancer cells and put it into a 0.5 mL insulin syringe
7. Keep the cells in the ice
8. Anesthetize a syngeneic mouse (4-6 weeks old) with 2% Isoflurane
9. Open the abdominal cavity
10. Take out the stomach then inject the cells, and try to inject them into the serosa; however, I sometimes inject the cells into the muscle layer.
11. If bleeding occurs after cell injection, the injection has failed
12. Suture the peritoneal cavity
13. Remove the stomach after a month, and dissect the stomach
Mice have never died, and they do not have any cancer organs even if I injected gastric cancer cell lines in their stomach.
Metastasis has also never been discovered.
This experiment has been conducted with the naked eye without a microscope.
I have been frustrated. What is my problem? Would you do me a favor, please?
Relevant answer
Answer
Hello, I prefer to resuspend with a micropipette.
  • asked a question related to Cancer Cells
Question
1 answer
I want to perform WST-1 test with alginate based 3D-colorectal cancer cells. I wonder about before performing WST-1 test, ıs it necessary to remove alginate spheres from the cells (to carry it from 3D to 2D 96 well-plate)? or can ı perform the test with 3D alginate spheres? Thank you for your contribution.
Relevant answer
Answer
You can perform the WST-1 test directly on alginate-based 3D colorectal cancer cell cultures without removing the alginate spheres. The WST-1 reagent can penetrate the 3D structure, allowing the assay to measure cell viability within the 3D environment. Here are some tips:
1. **Ensure Adequate Reagent Penetration**: Mix the WST-1 reagent thoroughly with the medium to ensure it can diffuse into the alginate matrix.
2. **Incubation Time**: Allow sufficient incubation time for the reagent to react with cells within the 3D structure.
3. **Readout**: Measure absorbance as per standard WST-1 protocols.
This approach allows you to maintain the 3D culture conditions and assess cell viability more accurately in the 3D context.
  • asked a question related to Cancer Cells
Question
2 answers
Cell Cytotoxicity
Relevant answer
Answer
Thank you Malcolm Nobre
  • asked a question related to Cancer Cells
Question
1 answer
I am interested in analyzing the effects of some immunotherapy on the number of mitochondria in cancer cells as well as in immune cells. I would like also to isolate mitochondria to use them for further analysis far from other cellular compartments.
Relevant answer
Answer
Mitochondria are highly dynamic organelles undergoing coordinated cycles of fission and fusion, referred as ‘mitochondrial dynamics’, to maintain their shape, distribution and size. The number of mitochondria in a cell depends on cell activity and type. For instance, mitochondria multiply when the energy needs of a cell increase. Therefore, power-hungry cells have more mitochondria than cells with lower energy needs.
Mitochondria can take up as much as 25% of the cell volume. Cells contain approximately 1000 to 2500 mitochondria. It would be difficult to count the exact number of mitochondria in the cells. Even if you try to isolate mitochondria from cells, the count will not reflect the exact number present in the cell because you would lose quite a number of them during the isolation process.
Attached below are a few articles for your reference.
So, instead of the count I would suggest you work on the mitochondrial mass of cells which is evaluated by means of one of the following methods:
1. the biochemical method based on the assay of the matrix specific enzyme CS,
2. the genetic method based on the measurement of the mtDNA copy number, or
3. by evaluating the mass of typical mitochondrial proteins as translocase outer membrane (TOM) subunits through immunoblotting.
For more information regarding the above (mitochondrial mass) you may want to refer to the article attached below.
The second part of your question.
Isolation of mitochondria from single cell suspension.
1. Having obtained single cell suspension, wash them with PBS once and centrifuge at 300 × g for 3 min
2. Discard the supernatant and resuspend with 5 volumes of extraction buffer.
Extraction buffer recipe:
20 mM HEPES-KOH, pH 7.5,
0.25 M sucrose,
10 mM KCl,
1.5 mM MgCl2,
1 mM EDTA,
1 mM EGTA,
1 mM dithiothreitol,
0.1 mM PMSF
3. The cellular suspension is homogenized with a Teflon-glass homogenizer with 10–20 up-and-down passes of the pestle. Use of a teflon-glass homogenizer is recommended as it can lead to increased yield. Homogenization and all the subsequent steps of the protocol must be performed at 4°C to minimize the activation of phospholipases and proteases. Avoid excessive homogenization since it can cause damage to the mitochondrial membrane and trigger release of mitochondrial components.
4. The homogenate is then centrifuged at 750 × g for 10 min.
5. The resulting supernatant is transferred to a pre-chilled centrifuge tube and stored on ice, and the pellet is resuspended in extraction buffer, homogenized and centrifuged as for steps 2–4.
6. The supernatants obtained from the two low-speed spins are pooled and then centrifuged at 10,000× g for 15 min. Crude mitochondria, which are recovered in the pellet, are resuspended in extraction buffer (approx. 20–25 μL).
7. You may check the purity and the enrichment of mitochondria from whole cells by Western blot analyses, measuring cytosolic (e.g. actin), nuclear (e.g. lamine A/C), and mitochondrial (e.g. OXPHOS subunits and porin) marker proteins.
8. The integrity of the mitochondrial fraction can be tested by quantitation of an intermembrane space protein (e.g. cytochrome c) or a matrix protein (e.g. cyclophilin D) by immunoblot. Alternatively, you could measure mitochondrial Ca2+ buffering capacity to assess the integrity of isolated mitochondria.
Most of the methods to isolate mitochondria rely on differential centrifugation, a two-step centrifugation carried out at low speed to remove intact cells, cell and tissue debris, and nuclei from whole cell extracts followed by high-speed centrifugation to concentrate mitochondria and separate them from other organelles. If you require a high degree of purity, density gradient centrifugation or affinity purification of the organelle are used to further purify mitochondria or to separate different populations of the organelle.
The paper attached below will be helpful.
Regards,
Malcolm Nobre
  • asked a question related to Cancer Cells
Question
2 answers
Hi everyone,
I need help and yuoue experience!!!
What kind of cell culture contamination is it?
Video attached: The cells are lung cancer cells that are thawed from -80°C fridge and we known that are probably alla dead, but we see this strange, non identify object, that move and change shape.
I never seen this kind of bacteria before. I always seen the classical sand contaminaion with torbid medium.
Thank you for any information or suggestions
Valeria
Relevant answer
Answer
It looks like cell debris in Brownian motion.
  • asked a question related to Cancer Cells
Question
5 answers
The cell images of MDA-MB-231 shown illustrate the effects of different drug treatments on cancer cell migration. There are four drug treatment groups: A, B, C, and D. Each group has three treatment time points: 0 hours, 24 hours, and 48 hours. The 0-hour time point is actually the group without any treatment, serving as a control.
My questions are:
1. When taking microscopic photos of the cells, is it acceptable to put the 0-hour images from each group (I, II, III, and IV) in the same folder?
2. If, during the process of assembling the figures, the images of groups II and IV (Groups II and IV are both untreated control groups) are accidentally swapped, will this affect the scientific conclusions?
3. Additionally, is it possible to omit three of the four images from groups I, II, III, and IV, keeping only one image as a common control for groups A, B, C, and D?
Thanks
Relevant answer
Answer
Choose controls that are relevant to the experiment. For cell migration studies, this might include negative controls (untreated cells) and positive controls (cells treated with a known migratory agent).
  • asked a question related to Cancer Cells
Question
3 answers
Resistant cancer cells
Relevant answer
Answer
The development of drug-resistant cancer cell line can take from around 3 to 18 months. If you are interested in developing resistance cancer cells in the laboratory, then you may want to refer to the article attached below.
This article summarizes the major methodological approaches for developing drug-resistant cell lines in vitro. Key decisions to be made prior to starting resistant cell line development; the choice of parent cell line, dose of selecting agent, treatment interval, and optimizing the dose of drug for the parent cell line, all of these have been discussed here.
However, if you wish to obtain drug resistance cancer cells, then as mentioned by Sabine Strehl , you may refer to the link above. RCCL has a collection of drug-resistance cancer cell lines from different cancer entities. The drugs used include all major classes of cytotoxic anti-cancer drugs as well as targeted agents.
Good Luck!
  • asked a question related to Cancer Cells
Question
1 answer
I have loaded mesoporous silica nanoparticles with a drug and attached them to antibodies but after conjugation with antibodies, the nanoparticles loaded with the drug lose their functionality to kill cancer cells, as if the presence of antibodies block the release of the drug. Anyone has faced the same problem or any idea how to solve it?
Relevant answer
Answer
Dear Hanieh
1. You'd better to measure the amount of loaded drug after antibody conjugation. Maybe buffers or conditions like pH used for antibody binding caused a burst drug release.
2- If the binding of the antibody is accompanied by unwanted crosslinking, it may negatively affect the efficiency of the system.
3- The hypothesis that you proposed may be true, for example, the binding of a large amount of antibody, in addition to the fact that due to steric hindrance, it cannot be effective in binding to the target cell, but it can interfere with the release of the drug. This is possible both through the creation of steric hindrance and the possibility of interaction between the drug and the antibody.
  • asked a question related to Cancer Cells
Question
3 answers
I'm planning a small research project involving conditioned medium from cancer cells on a human cell line (specifically KGN cells). I intend to expose the cancer cells to this medium for 72 hours and then use it on the KGN cells. However, I'm concerned about finding the best control condition. We know that cells consume nutrients from the medium, and using conditioned medium versus fresh medium might not be optimal.
I would greatly appreciate hearing your opinions and suggestions on this matter.
Relevant answer
Answer
Incubate the culture medium in cell-free dishes or flasks. This will allow for heat, evapoation and culture substatum contact changes that would occur in the conditioned medium. Dilute with fresh medium in the same ratio as your conditioned medium.
  • asked a question related to Cancer Cells
Question
4 answers
Is there a convienient way to mark cancer cells that are undergoing mitosis?
Relevant answer
Answer
Many thanks, that is very helpful! Luisa Matos do Canto
  • asked a question related to Cancer Cells
Question
4 answers
If you have several hundred cancer cell types, you have a rich resource for comparative analysis and experimentation. What experiments (innovative) would you do? Any thoughts are greatly appreciated!
  • asked a question related to Cancer Cells
Question
4 answers
Hello Friends
I have been trying to do scratch wound closure assay in semi-adherent cancer cells, whenever I put the scratch, either a whole layer of cells comes off and there are many large aggregates of cells floating, and the scratch is not neat. I am trying to assess migration potential in these semi adherent cells in vitro. Please suggest alternatives or possible modifications to have a solid and clean scratch. Thanks
Relevant answer
Answer
Dear Dr. Khursheed Ali
You may follow the protocol modified by the investigators in the below attached article. This protocol has been developed as a low-cost handmade method and will prove useful for semi-adherent cell lines. Refer to the protocol given in Figs 1, 2 and 3. The assay is called pipette tip gap closure migration assay especially meant to study cell migration or wound healing in semi-adherent cell lines and may be used as an alternative to the other migration assay protocols.
Regards,
Malcolm Nobre
  • asked a question related to Cancer Cells
Question
1 answer
Hi all,
My gene causes the cancer cells to grow very slowly when it is overexpressed. Is there a method to make the cells move faster?
Relevant answer
Answer
Your gene may be stressing your cells. This may be because the gene is so overly expressed it's consuming too much of the cells' resources, leading to stress and slow growth. The gene itself may be toxic or problematic in some way too. Not knowing the cell line or the gene, it's hard to say what may be happening.
  • asked a question related to Cancer Cells
Question
3 answers
Previously I did some surface marker expression (CD40, CD86) of RAW macrophages and DC2.4 cells using flow cytometry. For RAW cells, results were not satisfactory. Now, I am optimizing apoptosis assay with a pancreatic cancer cell.
Relevant answer
Answer
Designing a panel in flow cytometry means selecting your fluorochrome-conjugated antibodies so that their signal won't overlap while taking account of your cytometer specification and allowing you to distinguish your important cell populations.
  • asked a question related to Cancer Cells
Question
4 answers
Telomere length has been recognized as one of the best biomarkers of aging, indicating its importance in understanding the aging process. However, the evidence suggesting telomere length as a definitive biomarker of aging in humans is not conclusive and remains equivocal. Staying in harsh environments like space stations can lead to telomere lengthening as an adaptive response to stress and radiation exposure. Research on astronauts and individuals in high background radiation areas has revealed interesting findings regarding telomere dynamics in such conditions. Stressors such as space radiation and microgravity can trigger telomere lengthening, which typically reverses upon returning to Earth. This phenomenon is influenced by factors like radiation dose, dose-rate, and radiation type, underscoring the complexity of telomere modifications in extreme environments. Similarly, residing in another challenging setting, such as an underwater compound in a Florida lagoon, may also result in telomere lengthening due to increased pressure and exposure to environmental stressors. The compound's atmospheric pressure is 70% higher than at the surface, impacting bodily functions such as urination and metabolism. Researchers like Dr. Joseph Dituri are investigating the prolonged effects of this heightened pressure on the human body, as it could potentially offer insights into reversing the aging process and extending lifespan. The elevated pressure is thought to boost stem cell proliferation, telomere length, and collagen production, potentially slowing down or reversing aging effects. Nonetheless, it's crucial to recognize that continuous telomere replenishment is a hallmark of immortal cells like cancer cells, necessitating further research to grasp the possible consequences of these transformations.
Relevant answer
Answer
Thank you for your input. I completely agree with you that the idea of "reversing aging" is still mostly theoretical and debatable. Nevertheless, our understanding allows us to reverse specific signs of aging, like alterations in telomere length.
  • asked a question related to Cancer Cells
Question
7 answers
please
Relevant answer
Answer
To achieve a final concentration of 0.1% DMSO while maintaining the desired concentration of the plant extract, you can follow these steps:
  1. Calculate the Volume of DMSO Needed:Since you have a stock solution of 600 mg/mL of plant extract in 100% DMSO, you need to calculate how much of this stock solution to use to achieve the desired concentration. Let's denote:C1 = Concentration of plant extract in stock solution (600 mg/mL) V1 = Volume of stock solution to use C2 = Desired concentration of plant extract in final solution V2 = Total volume of final solution
  2. Use the Dilution Formula:The dilution formula is: C1V1 = C2V2Since you want to keep the DMSO concentration at 0.1%, the volume of DMSO you use will be V1.Plug in the values:C1 = 600 mg/mL C2 = Concentration of plant extract in final solution (you haven't specified, let's denote it as X mg/mL) V2 = Total volume of final solution (let's assume 1 mL for simplicity) You want the final DMSO concentration to be 0.1%, so the volume of DMSO (V1) can be calculated using:V1 = (0.1/100) * V2
  3. Calculate the Volume of Stock Solution Needed:Now that you have the volume of DMSO (V1) and you know the concentration of plant extract in the stock solution (C1), you can calculate the volume of stock solution needed using the dilution formula:V1 * C1 = C2 * V2
  4. Prepare the Final Solution:Once you've calculated the volume of stock solution needed, you can dilute it with the appropriate amount of DMSO to achieve the desired concentration.
Here's a summary of the steps:
  • Calculate V1 using the desired final DMSO concentration.
  • Use the dilution formula to find the volume of stock solution (V1) needed.
  • Prepare the final solution by diluting the calculated volume of stock solution with DMSO to achieve the desired concentration.
  • asked a question related to Cancer Cells
Question
4 answers
When culturing 22rv1 cells, I often encounter sudden cell death phenomena, and it is difficult to find a clear reason. When culturing cells for 4-5 passages, the cells suddenly begin to grow slowly, tend to aggregate, and gradually die. Can colleagues with similar experiences help me solve my confusion? I also culture other tumor cells, but have never encountered similar issues before.The following are images of problematic cells.
Relevant answer
Answer
Arvind Kumar Shukla First of all, thank you very much for your suggestions. However, to be frank, I find your advice too general. It's difficult to address my specific issue based on your response, as all the problems encountered with the cells could be explained using your suggestions.
  • asked a question related to Cancer Cells
Question
1 answer
Dear all, I hope you are well!
I would like to know how to calculate the optimal dose of human breast cancer cells to inject into female Wistar rats for testing the anticancer effect of a plant in vivo.
Also, I am slightly confused about using ethanolic extract (especially as ethanolic extract has shown anti-inflammatory activity and no toxicity in mice and is anticancerous in vitro) versus aqueous extract (based on traditional use).
Relevant answer
Answer
Optimizing the dose of human breast cancer cells for in vivo testing in female Wistar rats involves considerations such as tumor establishment, growth kinetics, and animal welfare. Here's a general approach to optimizing the cell dose for in vivo experiments:
  1. Establishment of Tumor Model:Select an appropriate human breast cancer cell line that is relevant to the research question and has been characterized for its tumorigenic potential in animal models. Determine the route of cell inoculation for tumor establishment. Common routes include subcutaneous (SC), orthotopic (in mammary fat pad), or metastatic (intravenous, intracardiac) injection, depending on the experimental goals.
  2. Pilot Studies:Conduct pilot studies to assess the tumorigenic potential and growth kinetics of the selected breast cancer cell line in female Wistar rats. Inoculate rats with different cell doses (e.g., 1 × 10^6, 5 × 10^6, 1 × 10^7 cells per rat) and monitor tumor growth over time by palpation or imaging techniques such as caliper measurements or bioluminescence imaging. Assess tumor take rate, latency period, growth rate, and final tumor size for each cell dose.
  3. Tumor Growth Kinetics:Analyze the growth kinetics of tumors derived from different cell doses to determine the optimal cell dose for consistent tumor growth within a reasonable timeframe. Calculate tumor volume doubling time and assess the correlation between cell dose and tumor growth rate.
  4. Ethical Considerations and Animal Welfare:Consider ethical considerations and animal welfare when determining the optimal cell dose. Avoid using excessively high cell doses that may lead to rapid tumor growth, excessive tumor burden, or compromised animal welfare. Ensure that the selected cell dose allows for the establishment of tumors that are biologically and clinically relevant to human breast cancer.
  5. Statistical Analysis:Perform statistical analysis to compare tumor growth characteristics (e.g., tumor volume, tumor weight) between different cell doses and determine statistically significant differences. Use appropriate statistical tests such as t-tests or ANOVA followed by post-hoc tests for multiple comparisons.
  6. Reproducibility and Consistency:Ensure reproducibility and consistency of tumor growth across multiple experiments by repeating the experiments with the selected optimal cell dose. Validate the findings by comparing tumor growth characteristics between independent experiments.
  7. Validation of Tumor Model:Validate the established tumor model by histological analysis, molecular profiling, or functional assays to confirm its similarity to human breast cancer and its suitability for preclinical studies.
By following these steps and considering factors such as tumor growth kinetics, animal welfare, and experimental reproducibility, you can optimize the dose of human breast cancer cells for in vivo testing in female Wistar rats. This optimization process is essential for generating reliable and relevant data for preclinical research and drug development studies.
  • asked a question related to Cancer Cells
Question
3 answers
Hello,
I have 2 questions regarding cancer mRNA vaccines. When synthetic mRNA vaccine for cancer is introduced into the body, our expectation is that the immune system will be activated against the tumor cells and exhibit a response to eliminate them.
However, the question arises as to whether, after translation and the emergence of tumor antigens on the surface of the target cells, primarily dendritic cells (DCs), it leads to the activation of cytotoxic T lymphocytes (CTL) and other immune cells that only eliminate those specific cells rather than the millions of tumor cells that have already caused cancer in the body tissues.
Despite many studies, I cannot comprehend the philosophy behind mRNA cancer vaccines in the face of this challenge.
Furthermore, I have another question in the same context. Assuming that the activated immune cells are intended to eliminate cancer cells, they face a formidable barrier called the tumor microenvironment and mechanisms by which tumors evade the immune system. Ultimately, these factors somehow inhibit the immune system.
The question is, how can the activated immune cells by mRNA vaccines overcome this microenvironment barrier and reach cancer cells? Especially considering that in many research samples, inhibitors of the tumor microenvironment are not simultaneously used with mRNA vaccines.
If you have information on the answers to these two questions, I would greatly appreciate your guidance.
Thank you.
Relevant answer
Answer
The immune system can recognize and respond to foreign mRNA through various mechanisms, particularly when mRNA is introduced into cells via delivery systems such as lipid nanoparticles (LNPs) or viral vectors. Here's how the immune system can become activated against mRNA and potentially eliminate it:
  1. Recognition by Pattern Recognition Receptors (PRRs):mRNA molecules may be recognized as foreign by pattern recognition receptors (PRRs) expressed by immune cells. PRRs, such as Toll-like receptors (TLRs) and retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs), can detect viral RNA or other nucleic acids with pathogen-associated molecular patterns (PAMPs). Activation of PRRs by foreign mRNA can trigger intracellular signaling pathways that lead to the production of pro-inflammatory cytokines, type I interferons (IFNs), and other immune mediators.
  2. Activation of Innate Immune Responses:Recognition of foreign mRNA by PRRs can lead to the activation of innate immune responses, including the production of inflammatory cytokines and chemokines. Innate immune cells such as dendritic cells (DCs) play a crucial role in sensing and responding to foreign mRNA. DCs can internalize mRNA-containing particles, process the mRNA, and present mRNA-derived antigens to T cells, leading to the activation of adaptive immune responses.
  3. Induction of Adaptive Immune Responses:Presentation of mRNA-derived antigens by antigen-presenting cells (APCs) can stimulate the activation and differentiation of CD4+ T helper cells and CD8+ cytotoxic T cells specific to mRNA-derived epitopes. Activated T cells can recognize and eliminate cells expressing foreign mRNA, leading to the destruction of mRNA-transfected cells.
  4. RNA Interference (RNAi) Pathway:Endogenous cellular mechanisms such as the RNA interference (RNAi) pathway can also contribute to the recognition and degradation of foreign mRNA. Small interfering RNAs (siRNAs) or microRNAs (miRNAs) generated from foreign mRNA sequences can guide the RNA-induced silencing complex (RISC) to target and degrade mRNA molecules, leading to their elimination.
  5. Activation of Antiviral Defense Mechanisms:Activation of immune responses against foreign mRNA may involve the activation of antiviral defense mechanisms aimed at eliminating viral RNA. These mechanisms can include the upregulation of antiviral proteins such as protein kinase R (PKR) and 2',5'-oligoadenylate synthetase (OAS), which inhibit viral replication and promote mRNA degradation.
Overall, the immune system can become activated against foreign mRNA through various recognition pathways, leading to the induction of innate and adaptive immune responses aimed at eliminating the foreign mRNA and preventing potential harm to the host organism.
  • asked a question related to Cancer Cells
Question
1 answer
I'm attempting to stain cancer cells in suspension from a 12-well plate for my research project. Could anyone provide guidance on the most effective staining protocols and techniques for ensuring accurate and reliable results? Any insights or recommendations on suitable staining dyes, concentrations, fixation methods, and imaging procedures would be greatly appreciated. Thank you in advance for your assistance!
Relevant answer
Answer
What do you intend to stain: cell surface markers, an intercellular protein, organelles, nuclei, nucleoli, ... ? Without knowing any details of your experimental set-up it is impossible to give you any advice; do some research on your own and then ask such more specific questions.
  • asked a question related to Cancer Cells
Question
2 answers
Can I select normal cells that are different from the cancer cells I am studying to test for toxicity?
Relevant answer
Answer
Hello Sun Sun,
It is necessary because it will help to determine the therapeutic index of your investigational product meaning the relative effectiveness of your compound in causing cytotoxic effect on cancer cells compared to inducing cell death in normal cells.
You may calculate the therapeutic index as the ratio of 50% cytotoxic concentration (CC50) to the inhibitory concentration of 50% (IC50).
Therapeutic index: TI = CC50/IC50
The greater the TI value, the more effective is the drug against cancer cells with least cytotoxic effect on normal cells (reduced side-effects).
CC50 is defined as the median cellular cytotoxic concentration that results in the death of 50% of the normal cells while IC50 is defined as the concentration of the candidate compound needed to inhibit a biological process or response by 50% and is used as a measure of drug potency.
Usually, normal cells that belong to the same tissue of origin as the cancer is selected for cytotoxicity assay. But it is not always necessary. You may select any normal cell, the most commonly used normal cells are the fibroblasts.
Best.
  • asked a question related to Cancer Cells
Question
6 answers
I want to graft the cancer cells i.e, MCF7 on cam of egg. To do this I need to have some scaffold. In a paper i read about matrigel, is there any other alternative of matrigel or can we graft the cells directly onto the cam without any scaffold?
Relevant answer
Answer
Yes. Any tissue that produces angiogenic factors will graft well.
  • asked a question related to Cancer Cells
Question
2 answers
Hello.
I have a question about these two behaviors, the Warburg and Crabtree effects. I am a master's student and my teacher said to me that Warburg occurs in cancer cells and Crabtree in yeast, basically that the Warburg effect on cancer is analogous to the Crabtree effect on yeast. However, I see some scientific articles that say that cancer cells have both. In yeast, I understand that high concentrations of glucose inhibit oxidative respiration. I am confused. Can someone explain this to me? Cancer cells only exhibit the Warburg effect or also exhibit the crabtree effect?
Thanks
Relevant answer
Answer
The “Warburg effect” is defined as an increase in the rate of glucose uptake and preferential production of lactate, even in the presence of oxygen. The normal differentiated cells will rely primarily on mitochondrial oxidative phosphorylation to generate the energy needed for cellular processes. But cancer cells instead will rely on aerobic glycolysis, a phenomenon which is termed as “Warburg effect”.
Aerobic glycolysis is an inefficient way to generate ATP, however, the advantage it confers on cancer cells has been unclear. But there is a thought stating that cancer cells acquire and metabolize nutrients in a manner conducive to proliferation rather than efficient ATP production, and the associated mutations acquired by cancer cells enable them to function in this manner.
In some cancer cells, sometimes, depending on the absence or the presence of glucose and the environmental conditions, the cancer cells can reversibly switch between fermentation and oxidative metabolism. This short-term and reversible event is referred to as the “Crabtree effect”. This reversible shift might be an advantage to cancer cells, as it would allow them to adapt their metabolism to the rather heterogeneous microenvironments in malignant solid overgrowths.
So, cancer cells show both the effects, depending on the environmental conditions, they can switch reversibly.
Your teacher is right when he/she said that the "Crabtree" occurs in yeast. Some yeast species such as S. cerevisiae use fermentation even in the presence of oxygen when glucose concentrations are sufficiently high. The use of fermentation in the presence of oxygen and at high glucose concentrations is referred to as the “Crabtree effect”. Yeasts that display a “Crabtree effect” are Crabtree-positive while yeasts that do not display a “Crabtree effect” are Crabtree-negative.
You may want to refer to the article attached below for more information on “Crabtree effect” in yeast.
So, when high concentrations of glucose or fructose are added to the culture medium, respiration is frequently inhibited. This phenomenon of “Crabtree effect” is observed in numerous cell types, particularly you will find this effect in proliferating cells, which would include not only tumor cells but also yeast.
Hope this clears your doubt!
Best.
  • asked a question related to Cancer Cells
Question
4 answers
If "YES", how long it take to grow and if "NO" then why?
Relevant answer
Answer
Hi Abhijeet, I was also interested in this topic. I found this publication describing a s.c. MCF7 mouse model:
Growth of MCF-7 breast cancer cells and efficacy of anti-angiogenic agents in a hydroxyethyl chitosan/glycidyl methacrylate hydrogel - PubMed (nih.gov).
In addition a German CRO is offering MCF-7 as s.c. model:
MCF7: Subcutaneous breast cancer xenograft tumor model (reactionbiology.com)
  • asked a question related to Cancer Cells
Question
8 answers
How long cancer cell can survive in trypsin and complete media (1:6) solution in room temperature (bsc)?
Relevant answer
Answer
They are fine, need not to worry, by tomorrow morning, they will be excellent
  • asked a question related to Cancer Cells
Question
4 answers
Hi
I used the MTT assay for drug-inhibiting cancer cells
Then, I used GraphPad prism to calculate the IC50 of this drug following these equations
- log (inhibitor)vs response (three parameters)
- log (inhibitor)vs response--variable slope  (four parameters)
- log (inhibitor)vs normalized response
- log (inhibitor)vs normalized response --variable slope
However, the result of each equation is not equal, especially both log(inhibitor) vs response (3 and 4 parameters) IC50 of both equations is significantly less than the concentration at 50% cell viability. When compared with log (inhibitor)vs normalized response IC50 of this equation is related to concentration at 50% cell viability.
As mentioned above, I wonder what is the suitable equation to calculate IC50 for further experiment such as apoptosis analysis?
Relevant answer
Answer
Hello Chidchanok Chawiwithaya Of course, there are ideas about the inappropriateness of R square in non-linear regression models.
However, compare the R square of each model with each other. Whichever was better and higher has a higher chance to be selected as the optimal model.
Of course, I think you can test other non-linear models, especially S (5PL) and special models.
  • asked a question related to Cancer Cells
Question
2 answers
When I calculate the IC50 of natural products on cancer cell and normal cells between concentration of 0.3125 and 40ug/ml using MTS, the inhibition curve is sigmoidal on cancer cells not on normal cells. I found GraphPad software require the the inhibition curve is sigmoidal, and some papers suggests using Hill equation. Does anyone have suggestions?
Relevant answer
Answer
Hello Bin Sun In my opinion, Graphpad software is very appropriate in this matter.
Be sure to try all the different types of dose-response equations.
  • asked a question related to Cancer Cells
Question
1 answer
Like in Lung cancer how b cells are getting affecting, so just want to how and what are the complications B cells are facing regarding different type of cancerous cells
Relevant answer
Answer
Yess sir
  • asked a question related to Cancer Cells
Question
2 answers
How do the intricate molecular and microenvironmental dynamics within the lungs create a preferential niche for the metastasis of various cancer types, and what specific molecular mechanisms govern this phenomenon, considering the diverse heterogeneity of cancer cells and the complex interplay with the pulmonary microenvironment?
Relevant answer
Answer
You may want to refer to the article attached below.
Best.
  • asked a question related to Cancer Cells
Question
4 answers
I am little bit confused to understand whether all the niches where cancer cells remain dormant are antimetastatic niches? How will we consider the accumulation in the bones?
Relevant answer
Answer
I am not sure if all niches with dormant tumor cells are anti-metastatic. There can be a lack of critical growth factors, including oxygen and nutrients, as well as immune regulation of tumor cell numbers within disseminate micro-metastases. Here is another of a host of reviews looking into this.
  • asked a question related to Cancer Cells
Question
4 answers
I am currently performing co-culture experiments with murine CD8 T cells isolated from mouse spleens and with skin cancer cells (PDV) in a 24-well plate. I first isolate the CD8 T cells from spleen which are around 70% alive at time of isolation, mix them together with CD3/CD28 T Cell activation beads to activate the CD8 T cells, and plate them 2x10^5 T cells on top of 2x10^5 skin cancer cells that were plated the night before. The cancer cells are treated with mitomycin C prior to plating the T cells in order to halt cancer cell proliferation. Then I let them incubate in a tissue culture incubator with standard parameters (37 degrees Celsius and 5% CO2) for 5 days. In addition to the wells where I co-culture the T cells with the plate cancer cells, I also plate some of the T cells with the activation beads in an empty well to act as a control. I then assess the viability of the T cells by staining with a live dead stain and running flow cytometry. However the results of the flow cytometry show show that most of my T cells are dead (<40% viability). The most problematic issue is that the T cell-only wells come up only 1% viability which render the rest of the experiment null since I would not have a T cell control to compare the T cell viability from the T cell/cancer cell co-culture wells.
The interesting observation is that the T cells co-cultured with cancer cells have greater viability compared to the control T cells cultured by themselves which makes me think that the cancer cells are stimulating the T cells and improving T cell viability that way. I was wondering if there are any adjustments I could make to improve the viability of my T cells especially for the T cells in the T cell only control wells?
Some adjustments I have made already:
1. Increase the speed of the T cell isolation from the spleen to improve baseline viability prior to seeding on the cancer cells
2. Utilize wide bore tips when pipetting T cells to decrease the shear force on the T cells
Thank you for any suggestions and apologies for the length of this question.
Relevant answer
Answer
Dear Matthew,
Your work seems interesting. Which type of cancer are you investigating? The results can be dependant on that too. What are the goals of your study?
Sincerely,
Amar, MSc in MLT, Researcher interested in breast cancer
  • asked a question related to Cancer Cells
Question
2 answers
We would like to obtain mitochondria from cancer cell lysates. However, we do not have a clear protocol. Additionally, we do not have the kit to enable mitochondria isolation. Do you have any protocol or article recommendations that you could suggest to us?
Relevant answer
Answer
Mohamed Khashan Thank you for replying ☺
  • asked a question related to Cancer Cells
Question
9 answers
I have been experiencing difficulty growing SKBr3 cells. They grow for a day or two then round up, float off the surface, and die. I use McCoy's media with 10% serum. I suspect it may have something to do with the surface I grow them on. Any suggestions? How do you grow SKBr3s?
Relevant answer
Answer
We cultured SKBR3 cells in DMEM low glucose medium with 12% FBS. it had very good morphology and get confluent with in 2 days after seeding. it is advised to split them before reaching above 80% confluency. For subculturing use 1x trypsin EDTA (300ul for T25 and 1ml For T75 flask, with incubation time 3 min)
  • asked a question related to Cancer Cells
Question
3 answers
can we use gold nanorods for Photothermal Therapy can any one guide me
Relevant answer
Answer
There is a lot of work related to this topic, but currently this method is still in the stage of research and not suitable for use.
See those recent reviews for example : https://doi.org/10.1039/D2RA00566B
Also as expressed by Mr Weippert, one of the question being currently asked is what becomes of the nanoparticles after the treatment (risks of long term toxicity).
Finally, gold salts, and gold nanoparticles are completely different objects from a chemistry point of view. I would wager their toxicity would be completly different both in mecanism and intensity.
  • asked a question related to Cancer Cells
Question
1 answer
Submission Deadline: 30 September 2024
Summary
Metabolism refers to the unique biochemical processes that sustain life in an organism. In cancer cells, predominant biological processes include glycolysis, reduced oxidative phosphorylation, promotion of apoptosis and cell death, and increased synthesis of metabolite intermediates essential for cell proliferation, migration, and death. These metabolic properties lead to changes in the tumor microenvironment.
Transcriptomics studies the total RNA (mRNAs and non-coding RNAs) transcribed from a specific cancer cell in a particular functional state. As the size of transcriptome datasets continues to increase regarding tumor biology, there is a growing demand for computational and analytical methods. At present, countless public datasets available online allow researchers to have a comprehensive view on aging and related disorders. Microarray or sequencing of mRNA, ncRNA, or m6A provides informative clues for delineating biological progresses. Multi-omics analysis, including genomics, proteomics, and matrix omics et al., helps providing a comprehensive view of different processes. Single-cell methods further make it attainable to chart genome, transcriptome, and proteome at single-cell resolution. Furthermore, the advancement in algorithm boosts the reports of novel findings from existed data, and large public health databases, such as NHANES or Seer, further helps unveil the risk factors in real world.
The above data allows the possibility for the detection of tumor biology, novel targets, and evaluation of therapeutic effectiveness. This thematic collection aims to provide a comprehensive overview of the latest research advances in cancer metabolism through integrative analyses. We welcome research articles, reviews, perspectives, commentaries, and clinical trials that discuss both basic and translational research as well as therapeutic perspectives in cancer from the view of metabolism.
Relevant answer
Answer
We welcome research articles, reviews, perspectives, commentaries, and clinical trials that discuss both basic and translational research as well as therapeutic perspectives in cancer from the view of metabolism.
  • asked a question related to Cancer Cells
Question
3 answers
What must happen for normal cells in order to become cancer cells?
How normal cells convert to cancer cells?
What the point that must be passed for converting normal cell to cancer cell?
Relevant answer
Answer
In studies of formation of carcinomas, the most common form of solid tumors in humans, tumors of epithelial tissue origin, it is common to characterize the changes in terms of drivers and processes that produce epithelial to mesenchymal transition. Here are some review articles.
  • asked a question related to Cancer Cells
Question
3 answers
We are intended to expand the cancer cells directly from patient tumor tissues. We will going to use X-VIVO for this purpose. Can you recommend some medium or serum free medium that are better than X-VIVO ?
Relevant answer
Answer
Hello! I would like to ask you how is your primary ovarian cancer tumor cell culture? Can you provide a specific protocol or experience for me to learn from? Thanks!
  • asked a question related to Cancer Cells
Question
1 answer
I haven't been able to find access to the full body text of this article. I need it for my research, so please help.
Relevant answer
Answer
You could ask the authors to send you a pdf - try the "Request full-text" ..! ;-)
Best regards,
Christian
  • asked a question related to Cancer Cells
Question
1 answer
I have prepared thymoquinone alginate beads, and I want to investigate the cytotoxicity of my product against colon cancer cell.
Is there a recommended way to do that:
placing the beads in the 96 well plate directly or extracting thymoquinone from the beads first then applying on the cells
Relevant answer
Answer
There are a few options for testing the cytotoxicity of thymoquinone alginate beads against colon cancer cells:
1. Extract thymoquinone from the beads first, then apply the thymoquinone to the cells. This allows you to know the exact concentration of thymoquinone the cells are exposed to.
- Extract thymoquinone using a solvent like ethanol or methanol. Sonicate or vortex the beads to release the thymoquinone.
- Evaporate the solvent to collect the thymoquinone. Dissolve in DMSO or culture medium for cell treatment.
- Treat cancer cells (eg. HCT116, HT29) with thymoquinone at different concentrations (10 μM - 100 μM). Assess cell viability after 24-48 hrs.
2. Directly add the thymoquinone beads to cells:
- Place beads in 96-well plate, seed cells on top or around beads. The thymoquinone will diffuse out.
- This makes it harder to control the exact dose, but may better mimic delivery in vivo.
- Assess cell viability after 24-48 hrs. Compare to control beads without thymoquinone.
3. Extract media conditioned with beads, apply to cells:
- Incubate beads in media for 24 hrs, collect conditioned media.
- Apply conditioned media to cells. This will contain released thymoquinone.
- Assess cell viability after 24-48 hrs compared to unconditioned media.
So, extracting thymoquinone first allows better dose control, while direct bead addition mimics in vivo delivery.
  • asked a question related to Cancer Cells
Question
2 answers
Greetings, everyone!
I have printed 3D structure for engineered liver tissue and then implanted in a rat liver.
However, I used HepG2 and EA.hy926 for this tissue.
Both cells are cancer cell lines, and they are the current problem in my study.
Other researchers still used cancer cells for in vivo experiment, so I think I should say some sentences in discussion about my issue.
How can we discuss using cancer cell (especially HepG2) for an in vivo implantation/ transplantation experiment?
Thank you all in advance for your valuable insights and contributions to this vital discussion.
Warm regards,
Alex
Relevant answer
Answer
This is a fair point, and I agree that it is worth considering.
The first cell lines to be generated were often from cancerous tissues, as the ability to expand and divide indefinitely is a useful characteristic (both from the cancer, and the researcher's point of view).
The use of (cancerous) cell lines however has become widely accepted, and most people will not discuss this in their research.
An alternative to using cell lines is to use primary cells. The problem here however is that they are more difficult to get (depending on the tissue type of course), are generally more finicky to grow, can show significant heterogeneity, and are less well characterised.
In short, I agree with your opinion and believe it should be discussed more widely, but reviewers are unlikely to make a fuss if you don't mention it in your discussion.
Sam
  • asked a question related to Cancer Cells
Question
1 answer
I am conducting an experiment where I am putting cultured cancer cells (B-ALL line) in whole blood and running them through a microfluidic device. I stain the cells with DAPI and DiI (cell membrane dye) and then add them to whole blood. The issue that I am facing is that clumps develop in my microfluidic device when I run the sample. I have noticed that the amount of clumps and the time it takes for them to develop are related to the concentration of cancer cells I add in the blood. (The more cells I add, the bigger the clumps and the quicker they develop.) Does anyone have experience with spiking cancer cells in whole blood and recommend any changes? Should I fix the cancer cells before spiking?
Relevant answer
Answer
Greetings.
So, if i suggest you to do few things.
First; Your total concentration of blood+cells should not exceed than 1M cells/ml for appropriate distribution.
Secondly; Clumping is not just because of cancer cells (or their concentration) but PI cause aggregation. If you may use any other DNA dye, it will save you further. PI is known to be very sticky dye. even in Flowcytometry, people need to clean the probe b/t the sample to remove PI carryover.
Then, cancer cells tends to form aggregates (called blasts). So you need to add a bit more anticoagulant in the buffer.
Finally, if nothing works; use 20-30 uM nylon mesh to clear aggregates before running the sample in instrument.
I hope your query is answered.
Best wishes.
  • asked a question related to Cancer Cells
Question
3 answers
We are conducting an experiment to analyze the change of pERK level after transfection of GOI to cancer cells. However, even in the group that contains only the transfection reagent, the pERK level has significantly increased up to 24 hours compared to the group that has not been transfected and is gradually decreasing. If this is a predictable result, I wonder what pathways are involved in the increase. If not, do you know how to eliminate the effect of transfection reagents?
Relevant answer
Answer
Not only transfection, even just changing the medium, bringing in fresh serum will change ERK1/2 phosphorylation in cells. Just leaving them for more than 24h post medium change without any additional intervention, or combining this with a reduced serum media may help. Cannot rule out any special transfection reagent directly activating ERK, but it is most probably fresh growth factors and should normalise if given enough time. Good Luck!
  • asked a question related to Cancer Cells
Question
1 answer
specially biomarker secreted by the K-562 CANCER CELL
Relevant answer
Answer
Yes, biomarkers are secreted by cancer cells, or they may be also generated by other non-cancer cells or tissue in response to cancer. Biomarkers can be DNA, RNA, protein or metabolomic profiles that are specific to the tumor. You may look at the DNA sequence, gene fusions, measure RNA or protein levels.
For K-562, I would suggest you focus on the exosomes released by K-562 cells into the conditioned media. These exosomes show angiogenic activity in both in vitro and in vivo matrigel assays. They have a role to play in the physiologic organization of endothelial cells. The exosomes may interact with their target cells in three ways namely, binding to cell surface receptors, fusion with the plasma membrane, or internalization. The exosome content could contribute to endothelial cell stimulation.
So, the ability of exosomes released by K-562 to interact with and stimulate endothelial cells could possibly suggest that exosomes could be a new target for chronic myeloid leukemia therapy.
You may want to refer to the article attached below. It will be helpful.
Additionally, the phenotype of these cells include the immunological markers CD3 (−), CD13 (+), CD19 (−), CD34 (−), CD41 (+), CD42 (+), CD71 (+) and CD235a (+), and they carry the BCR/ABL fusion gene, which promotes cell growth, inhibits apoptosis and causes defects of DNA repair.
Please refer to the attached articles below for more information.
Best.
  • asked a question related to Cancer Cells
Question
1 answer
I'm using panc02 cells, mouse pancreatic cancer cell line.Beforehand, I had some issues without doing any fixation, so now I’m trying with fixed cells.
Relevant answer
Answer
IF you are measuring APOPTOSIS then it defeats the purpose of using the dual dyes.
* Acridine orange will stain both live and fixed cells. In live cells fluorescence is weaker than fixed cells because it is primarily accumulated by lysosomes in live cells, so staining of nucleus is weak, since under low concentrations it does not reach nuclei at high enough amounts. In fixed cells AO will stain ALL nuclei bright green (double-stranded) and nucleoli (RNA containing, single stranded) orange/orange-red. This brightness is strong.
** Ethidium bromide does not stain intact live cells. Only stains live cells with compromized membranes like dead or apoptotic cells. This is a bright orange-red fluorescence. In fixed cells the brightness and color remain somewhat the same and ALL the nuclei will still be orange.
So in live cells, living non-compromised cells will be a diffuse green (from AO) without very clear nuclei (depending on the concentration used), and apoptotic cells will SELECTIVELY be orange from EtBr.
IN FIXED CELLS, ALL CELLS will be equally green+orange (some shade of yellow).......and no way to distinguish them. So, no, there is no point in using fixed cells if you are trying to measure apoptosis.
  • asked a question related to Cancer Cells
Question
1 answer
Lentivirus-Mediated Gene Delivery is a common method for introducing therapeutic genes into cancer cells. One such gene is Tiam1, known for its potential impact on cancer cell behavior.
Relevant answer
Answer
T lymphoma invasion and metastasis 1 (Tiam1) has been identified as an oncogene. The role of this gene in cellular migration, invasion, and metastasis may not be limited to T lymphoma. It has been reported to be important in promoting tumor progression in a variety of other cancers, such as breast cancer, colorectal cancer, and lung cancer.
Gene silencing would be an appropriate method to inhibit cell growth and invasive ability of cancer cells. For this purpose, one could use the RNA interference (RNAi), also known as gene silencing technology. Tiam1 silencing with lentiviral vector-mediated RNA interference (RNAi) technology would be appropriate.
However, there are many obstacles that one would come across. First and foremost, the phosphodiester bond of siRNA is vulnerable to RNases and phosphatases. Once it is systematically administered into circulation, endonucleases or exonucleases throughout the body will quickly degrade siRNA into fragments, thus preventing the accumulation of intact therapeutic siRNA in the intended tissue. Also, immune recognition in blood circulation, effective transmembrane trafficking, and escape from endosomes and lysosomes to the cytoplasm where antisense strands of siRNAs need to be loaded into RISCs are among others that need to be addressed.
Also, there is the issue of delivery systems and the off-target effects. Improving delivery to target sites and reducing the harmful effects on non-cancerous cells need to be looked into.
So, delivering siRNAs efficiently and safely to desired tissues and cells, and enhancing the performance of siRNAs with respect to their activity, stability, specificity must be investigated.
Regards,
Malcolm Nobre
  • asked a question related to Cancer Cells
Question
5 answers
Dear colleagues,
I am working on a research project to investigate the metastatic propagation of cancer cells, so I need to see the metastasis of these cells in-vivo. Can any help me to find an appropriate and also the least expensive method?
Relevant answer
Answer
Metastasis is a complex phenomenon hard to modelise. It requires intertwined sequence of events including intravasation, dissemination, extravasation, survival and growth. I am not sure that in vivo models fully recapitulate. However, if you want to model dissemination, you can inject tumour cells expressing an easily detectable marker such as GFP or luciferase.
  • asked a question related to Cancer Cells
Question
2 answers
Hi there!
I am working with hepatocellular carcinoma cells. I want to study oxygen consumption in these cells. Is there a kit or a procedure I can use other than the XF Cell Mito stress kit?
Relevant answer
Answer
Amadeo M Parissenti Thank you for your reply. I'm going to do research on that.
  • asked a question related to Cancer Cells
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 Cancer Cells
Question
4 answers
Even in the presence of large amount of oxygen and functionable mitochondria, cancer cells follow anaerobic respiration to generate the ATPs to meet it's energy demand. It sounds like they are wasting the energy comparing to the ATPs generated using aerobic respiration. But why the cancer cells are following this?
There are several possible reasons for this phenomenon which I am mentioning here,
1. It will generate large amount of ATPs within short time. Which will be comparatively same to the quantity of ATPs from aerobic respiration.
2. It does not require cell organelle like mitochondria. Irrespective of the organelle presence, anaerobic respiration occurs and generates energy.
3. It will generate ATPs irrespective of the available oxygen in the cell environment. So it can survive even the place where the blood vessels does not deliver oxygen.
4. Just like during exercise how the muscle cells perform the energy generation through anaerobic respiration in short time. Cancer cells do the same to divide rapidly. Further it create an acidic environment by accumulating pyruvate. This low pH suppress the body immune system to perform it's function effectively.
5. The important point what I consider is, bypassing the TCA cycle will reduce the feedback inhibition of glycolysis by TCA cycle intermediates such as citrate, etc. Even ATPs are allosteric inhibitors to glycolysis by inhibiting the Phospofructokinase-1 enzyme. But creating high energy demand through anerobic respiration, this can be neglected in the cancer cells.
Understanding the cancer biology is always ends in some phenomenon. I added several points about what I think about the reason behind the Warburg effect of cancer cells. I am interested to hear more phenomenon cancer cells do.
Add the ideas and facts about the Warburg effect of cancer cells to make this discussion more interesting.
Relevant answer
Answer
Warburg effect or aerobic glycolysis cannot be viewed from an energetic point of view. It requires a multimodal approach.
Lets start with a contrafactual analysis. What would happen if a malignant cell would not use aerobic glycolysis.
Proliferation reduces oxygen availability because angiogenesis is quite ineffective to compensate for the increased need of oxygen.
In second place, the production of reactive oxygen species would induce a very dangerous oxidative stress. Warburg effect prevents both.
The main purpose of the Warburg effect is not to provide energy to the cell but to create building blocks for synthesis of amino acids, purine and pyrimidine precursors, and antioxidative molecules.
The energy imbalance is compensated by two mechanisms:
1) Increase of the glycolytic flux with a glucose uptake that increases 10-17 folds.
2) The oxidative metabolism may be downregulated but it is not eliminated.
Furthermore, during Warburg effect, almost 50% of glucose can be metabolized through the OXPHOS pathway.
Tumor cells need to keep the Krebs cycle working.
Therefore, the idea that malignant cells use the glycolytic pathway exclusively without OXPHOS is a well established mistake since the Warburg days.
  • asked a question related to Cancer Cells
Question
2 answers
I am looking for a specific marker to evaluate low-differentiated human cancer cells in a murine model for immunofluorescence. Cytokeratin is not specific enough. Thank you for your help.
Relevant answer
Answer
Thank you, but the idea is based more to detect the human cells in mouse.
  • asked a question related to Cancer Cells
Question
5 answers
Hello,
I'm working on drug delivery in cancer cells. For that, I prepared slides of adherent cancer cells for confocal microscopy. I fixed the cells with 150 ul of 4% paraformaldehyde for 10 min and mounted the cover slip on a glass slide. Then I stored the slides at -20 degC. After one day I did imaging, I found that cells got flattened morphology and some granular structures were seen inside the cells that were totally different from their morphology. Imaging was also not good. I've some doubts regarding this:
1. Whether the incubation time with paraformaldehyde (10 min) was more than required or storage at -20 degC damaged the cells?
2. What should be the optimum time and volume of paraformaldehyde incubation?
3. At what temperature we can store the mounted slides and for how long?
Please guide me regarding this.
Thank you
Relevant answer
Answer
Hello,
I usually incubate my immunofluorescence specimens in a 4% paraformaldehyde solution for 15 minutes and I have never faced any problems following this prodedure (I use about 200 to 250 μl for each slide). I store fixed cells at 4 degrees Celsius in PBS solution containing sodium azide until staining. I've tried keeping them for about 8 weeks and it worked just fine. Just make sure they stay hydrated and sealed to avoid contamination.
Hope this helps,
Best regards,
George
  • asked a question related to Cancer Cells
Question
2 answers
My cells exhibit numerous round and brightly illuminated objects, yet the culture medium remains clear.
I conducted observations using a 20X magnification objective lens.
However, I'm uncertain whether this phenomenon is a result of cell overcrowding or microbial contamination.
Does anyone have any suggestions?
Relevant answer
Answer
Hello Ta Tsao
Cancer cells don’t have contact inhibition, which means that when they’re 100% confluent, they’ll squeeze together, and crowding will cause cells to reduce to half of their original size. If these cells aren’t split at the required time, they’ll start detaching from the substratum, finally leading to cell death.
In cell culture, many cells round up during mitosis, forming very refractile (bright) spheres that may float freely if the culture is physically disturbed. However, dead cells often round up and become detach from the substratum and are usually not refractile.
So, this phenomenon is basically a result of cell overcrowding.
Best.
  • asked a question related to Cancer Cells
Question
1 answer
I did today tissue culture for mcf7 cell line( breast cancer) and i check my work under microscope i saw this below is this cancer cell or just cells debris?
Relevant answer
Answer
Hi Shahad,
How many days have passed since you passaged these cells? While there are few healthy ones showing up in a different phase contrast, many of your cells are dying or do not look healthy unfortunately. MCF-7 cells do tend to grow slowly at times. Harsh passaging conditions should be avoided. You may try using a milder cell dissociation agent like Accutase instead of Trypsin during passaging.
  • asked a question related to Cancer Cells
Question
1 answer
I am specifically interested in breast cancer cells but numbers for any cancer type would be very helpul. Also what is their typical diameter?
Relevant answer
Answer
The cell membrane may have thousands of pores. It would be of interest to understand the nature of pores present in normal v/s cancer cells (in general). In cancer cells, the deficiency of lipoproteins in plasma membrane and lack of fatty acids in phospholipid make membrane more diluted, deformed and alleviated. This is why membrane of cancer cells is much softer, irregular, loose as compared to normal ones. So once deformed, the pore in the cancer cell membrane closes much more slowly than the pore in the normal cell membrane because the cancer cell membrane takes longer to relax to equilibrium than normal cell membrane. The pore area in the cancer cell membrane is larger than that in the normal cell membrane because the cancer cell membrane is softer than the normal cell membrane.
As the membrane of cancer cell become more soft and loose, the cell loses its minerals like calcium, potassium, magnesiu