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
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.
"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?"
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?
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?
Is it somehow related to tumour microenvironment or some other reason behind this.
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.
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.
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 !
Drug resistance is a significant challenge in cancer treatment, and this question explores how genetic modifications might overcome this barrier.
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,
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.
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.
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?
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?
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.
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.
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
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
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?
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.
Is there a convienient way to mark cancer cells that are undergoing mitosis?
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!
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
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?
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.
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.
I welcome your comments.
References:
[1] https://lnkd.in/gRkczjVf
[2] https://lnkd.in/gPQxpbCW
[3] https://lnkd.in/gWDpfnXk
[4] https://lnkd.in/ghipmQnp
[5] https://lnkd.in/g7JcAQmN
[6] https://lnkd.in/gbMZmn_Q
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.
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).
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.
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!
Can I select normal cells that are different from the cancer cells I am studying to test for toxicity?
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?
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
If "YES", how long it take to grow and if "NO" then why?
How long cancer cell can survive in trypsin and complete media (1:6) solution in room temperature (bsc)?
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?
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?
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
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?
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?
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.
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?
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?
can we use gold nanorods for Photothermal Therapy can any one guide me
Submission Deadline: 30 September 2024
For more detail: https://www.techscience.com/or/special_detail/tumor-metabolism
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.
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?
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 ?
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.
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
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
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?
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?
specially biomarker secreted by the K-562 CANCER CELL
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.
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.
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?
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?
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?
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.
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.
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
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?
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?
I am specifically interested in breast cancer cells but numbers for any cancer type would be very helpul. Also what is their typical diameter?