Science topics: Centrifuges
Science topic

Centrifuges - Science topic

Explore the latest questions and answers in Centrifuges, and find Centrifuges experts.
Questions related to Centrifuges
  • asked a question related to Centrifuges
Question
6 answers
Hi, I have been using a Q-Sonica Q500 Sonicator to disperse samples on nanocellulose and make emulsions. I have notices black dust like particulates settling out in some samples and on the bottom of tubes after centrifuging. Has anyone had any success in minimizing or eliminating this contamination? If not, are there any good ways to remove it? I've had some success filtering it out using membranes when working with nanoemulsions but for larger nanocellulose particles I'm not sure that this would be effective.
Thank you in advance for any advice,
Josh
Relevant answer
Answer
Dear Joshua Van Dyke, why not using a bath sonicator instead of the probe one. My Regards
  • asked a question related to Centrifuges
Question
1 answer
I am researching on azo dye degradation by bacteria. To analyze FTIR, how should I prepare my samples? Do I need to centrifuge, add anything or subtract anything? I am using minimal salt medium with 0.25% glucose.
Relevant answer
Answer
Depends on the FTIR method you are using, I guess you are using a configuration suitable for measuring liquids. First thing I would do is measure the media-bacteria-dye solution spectrum and media-bacteria solution spectrum. First check that it is correct spectra (absorption intensity, presence of relevant peaks...) and then see if there is any characteristic band corresponding to the dye. If there is, you can quantify dye degradation using that peak, if there is not... try processing the sample so you get a nicer spectrum, maybe.
  • asked a question related to Centrifuges
Question
1 answer
We are experiencing different problems pelleting fibroblast:
a) From the resuspension of the comercial vials (after defrosting, we resuspended in 5ml complete DMEM + L-Glutamin + FBS + Streptomycin + Penicilin)
b) From a suspension of a 25mm2 flask at confluence (same medium).
In both cases we used 50ml falcon tubes and 25ºC.
- We don´t see any pellet by centrifuging at 1500rpm/254 rfc
- We see just a very tiny deposit when centrifuging at 1900/400 rfc. But we are afraid the cells could suffer.
Any recomendations? Thank you very much for all your help.
Relevant answer
Answer
Dear Dr. Ana Lopez,
I feel the selection of an appropriate centrifuge tube size may solve this problem. Depending on the volume of your sample, you may choose the most appropriate tube size to ensure precise and effective separation as it is a critical factor in optimizing the centrifugation process.
Using a tube that is too large for the sample volume can lead to inefficient separation, as the larger surface area may cause the sample to spread thinly, reducing the sedimentation force on each particle. 
Therefore, matching the tube capacity closely with your sample volume is essential for achieving optimal separation efficiency.
Have you tried using 15ml centrifuge tube instead of 50ml tube for a small volume of 5ml? I feel it should work at the same speed of 1500rpm.
Regards,
Malcolm Nobre
  • asked a question related to Centrifuges
Question
2 answers
For my project, I need to extract RNA from muscle tissue of EEL. I am trying but the yield is low and Purity not good. The 260:230 ratio is always below 1.50 and 260:280 ratio ranges 1.6-1.86 and yield is around 120-140 ng/µl. In brief, I followed, 1 ml of TRIzol for 30-50mg of tissue for homogenization, incubated for 5 mins at room.T. Then 200µl of chloroform used for phase separation (centrifuged at 12000g for 15 min at 4oC), used 500 µl isopropanol for pelleting (incubated 10 mins at Room.T) then centrifuged at 12000g for 10 min at 4oC, then used 75% ethanol for washing pellet (centrifuged at 7500g for 5 min at 4oC) then incubated at 55oC in water bath for 10-15 mins after mixing with 20µl of MLQW.
Many Thanks......
Relevant answer
Answer
The protocol seems okay. But you need to consider the points provided below which you may have missed.
1. Tissue may not have been fully homogenized.
2. Tissue may not have been immediately processed or frozen after removal from the fish.
3. Tissue may not have been completely disrupted. If centrifugation is done prior to adding chloroform, there should be a white mucus-like pellet. If there is a tan-colored precipitate, this is indicative that not all the cells have been lysed.
4. If a mortar and pestle has been used to powder the tissue, RNA may stick non-specifically to the mortar and pestle. It may be better to use a glass homogenizer and teflon pestle. Add TRIzol to the homogenizer, then add the frozen tissue and homogenize.
5. Homogenizing for too long and too continuously in a small volume (say 1 ml) may cause heating of the sample and this may result in degradation of RNA in the tissue. Please note that the tissue should be cooled during homogenization, and homogenization should be done in on-off cycle.
6. Use 100mg of tissue sample per 1ml of TRIzol.
7. You may collect the upper aqueous phase carefully without disturbing the interphase. You may collect 80% of the upper aqueous phase and leave behind 20% so that you do not contaminate the aqueous phase.
8. You may try two 75% ethanol washing steps instead of a single washing to remove any remaining impurities in the form of water-soluble salts. It may help to improve RNA purity.
9. Instead of MLQW, use TE. If sample is dissolved in water, the ratio may be low due to the acidity of water or the low ion content in the water. The ratio can go up if the sample is dissolved in TE and the spectrophotometer is zeroed with TE.
Hope these suggestions help to increase the yield as well as the purity of RNA.
Best.
  • asked a question related to Centrifuges
Question
2 answers
I have been trying to collect bacteria from human fecal sample to run on MT-PCR, however, for some reason, the concentration of human DNA is quite low and could not confirmed on PCR. My technique is making a solution of fecal sample with PBS in 1g per 10 ml, centrifuge at 500g for 3 minutes and collect supernatant, repeat three times and final centrifuge is 20000g at 4 Celsius degrees for 20 minutes. Beside it give very good concentration of bacteria, but I also need a good concentration of human DNA also. Should I lower centrifuge force or reduce time? Thank you for you help.
Relevant answer
Answer
  • asked a question related to Centrifuges
Question
3 answers
Can anyone suggest any kit for SOD activity in plant? I have the protocol for NBT reduction but don't have centrifuge for falcon as the protocol requires 3mL of homogenate to be centrifuged in falcon. Can someone provide any alternative?
Relevant answer
Answer
Bhawna Bhawna Follow the protocol for 3 mL. At the time of centrifugation, simply separate the mixture into two 1.5 mL portions in Eppendorf tubes.
  • asked a question related to Centrifuges
Question
1 answer
Hello.
I found a new version of the core model and have not yet submitted it. That's why I have to explain the subject of gravity very briefly. .
All my findings are substantiated in detail.
Based on the results of my new nuclear model:
The force of gravity between space objects is inversely proportional to the square of the distance between them.
Important note: In the gravity formula that I found, the mass of the objects is not used.
My article is about the environment inside space objects. This environment is the sum of the circles around them.
Summary:
In the inner environment of any mass that has a nucleus, the force of gravity increases with increasing distance.
The constant value of gravity in each ring is different from the next ring. And moving towards the outer ring, the gravitational constant increases.
I calculated the gravitational constant in each loop. I will explain later
From the first ring to the last ring: centrifugal force is equal to the force of gravity.
As a result, the entire perimeter of any object in the rings is always weightless.
There are exceptions, which I will explain later.
But what happens below the first ring is completely different. Here, the acceleration of the centrifugal force is much greater than the acceleration of the gravitational force.
There is an important reason that I will explain later.
Although the force of gravity is increasing, we do not feel it because the weight is decreasing.
According to my calculations, in the area under the first ring, at any moment: the weight of objects is inversely proportional to the square of the distance between the object and the ground. And for this reason, the increase in gravity has no effect on the formulas for launching rockets into space.
Relevant answer
Answer
Your idea about gravity within a solar system being stronger farther away is very interesting, but it goes against what scientists currently know about gravity. Gravity weakens with distance according to established physics.
While the concept of centrifugal force balancing gravity exists, it doesn't negate gravity entirely. Similarly, weight should increase closer to the center of mass, not decrease.
If you have detailed calculations and want to challenge the current understanding, consider publishing them in a scientific journal for review by other scientists. Just be prepared to show how your model aligns with existing principles.
  • asked a question related to Centrifuges
Question
4 answers
For WGS, we need to obtain pellets of Avibacterium paragallinarum (Pasteurellacea-like baceria). What are the preffered centrifuge conditions for this bacterial family? Thanks
Relevant answer
Answer
For genomic DNA, any standard condition, 15-20 min, 5000 g would work, however, good to use at 4 °C if possible. Time can vary based on your sample density.
  • asked a question related to Centrifuges
Question
1 answer
It's never used... Then the bodies fall in, right?
Relevant answer
Answer
People prefer static models, without using G!
  • asked a question related to Centrifuges
Question
1 answer
Identifying phycosphere composition of diatoms but I want to know how to separate the bacteria living in the media
Relevant answer
Answer
To separate bacteria living in the media from diatoms and identify the phycosphere composition of diatoms, you need to apply a method that will establish
1. How to collect and prepare the sample
2. Differential centrifugation
3. Filtration and
4. Analysis of Diatoms and Bacteria
If you so desire the method ; Contact Ogelchemicals@gmail.com.
  • asked a question related to Centrifuges
Question
1 answer
I'm currently conducting myeloma cell (MM1s) culture for experimentation. I'm using RPMI-1640 medium with 10% FBS and 10% penicillin+streptomycin. I seed 5*10^6 cells in a 150mm petri dish with 20ml of medium and maintain them every three days. However, with each culture, the cell number doesn't increase significantly, and viability fluctuates between 40-60%. After just a couple of maintenance cycles, all cells die. I need a large quantity of cells for my experiments, so I'm trying to scale up the culture while maintaining and conducting experiments simultaneously. However, I'm stuck because I can't perform experiments, and all the cells are dying. What could be the problem? Here's my maintenance protocol:
  1. Collect cultured cells in a conical tube.
  2. Centrifuge at 100G for 5 minutes.
  3. Resuspend the pellet in 1mL of PBS warmed to 37°C and add 9mL to make a total of 10mL.
  4. Centrifuge again at 100G for 5 minutes.
  5. Resuspend the pellet in 1mL of RPMI medium and count the cells.
Even with this method, both viability and cell numbers are very low. I reduced the centrifuge speed because lower rpm CFG was said to aid in dead cell separation. Previously, I centrifuged at 1800rpm for 3 minutes, but when I changed to lower rpm, viability increased by about 20% (approximately 60%). The pellet appears visible. When I observe the cells under a microscope during culture, they don't seem to be in good condition. Changing the media doesn't yield different results. What could be the problem?
Relevant answer
Answer
Hello Iris Yoon,
I have some suggestions which you could incorporate in your protocol.
1. Subculture cells before they reach confluence because cells in over-confluent cultures begin to form rosettes with necrotic centers.
2. Use a subcultivation ratio of 1:2 to 1:4.
3. MM1S is a semi-adherent culture. So, you may have to gently scrap the loosely attached cells with the help of a cell scrapper to get all the cells in suspension.
4. In your maintenance protocol, why do you resuspend the cell pellet in PBS? Use complete growth media to resuspend the cell pellet.
5. You should supplement your growth media (RPMI + 10% FBS) with 2mM L-glutamine.
Hope these suggestions work!
Best.
  • asked a question related to Centrifuges
Question
1 answer
Article :
Cyclic Behavior of Bucket Anchor Foundation in Silty Sand under Sustained Pull-Out Loads via Centrifuge Model Tests
This article's 1st author is me.
you can easily assess it by checking 3rd author Jae-Hyun Kim who is already co-author of my previous papers.
Relevant answer
Answer
Just wait for the publication of your paper. Be sure that you are author or co - author of the paper with significant contribution.
  • asked a question related to Centrifuges
Question
2 answers
Why is it that the Coriolis force is maximum at the poles where there is zero rotation and zero at the equator and centrifugal force zero at poles and high at the equator?
Relevant answer
Answer
The Coriolis Force is not a "force", as such, but an illusion created by the rotation of the Earth. At the Poles, where there is no sideways motion of the ground supporting a pendulum, the force appears to be largest because as the Earth turns around you (because of its rotation) there is nothing to hide that rotation relative to you. But at the Equator, you and your pendulum are being dragged around the Earth at over 1000 miles/hour, which twists the support of the pendulum in a way that hides the rotation of the Earth from your view. The "torque" on the top of the pendulum support is largest at the Equator, but since you can't see any rotation, it appears that the "force" is zero.
In reality, watching a pendulum swing back and forth at the Equator, as it moves northward it twists a smidgen toward the right, while as it moves southward it twists a smidgen to the left (as a result of the torque acting on its support), because the effect is in the opposite direction in the northern and southern hemispheres; and although you could see each of the two small twists with a very high magnification, since they exactly cancel each other, you can't detect any deviation from a straight-line motion with ordinary instruments.
I don't know whether it would help, but my website has a page about "Coriolis Effects" with both a discussion and diagrams I created for my astronomy students, and links to two online videos. Unfortunately, since I wrote the page over 20 years ago, the top one, though it still goes to the linked page, doesn't go directly to the video, so you have to click on the .mpeg link to download the short playground demo (I'll fix that ASAP, but probably not before you read this). However, the link to the outstanding film "Frames of Reference" still goes directly to the Internet Archive's copy. The text I wrote is probably a more useful discussion of the pendulum's demonstration of the Coriolis Effect, but the 1950's movie is well worth watching, even though it only deals with the Effect near the end. (The URL for the page I wrote is https://cseligman.com/text/planets/coriolis.htm )
  • asked a question related to Centrifuges
Question
3 answers
I have prepared competent cells of L. lactis NZ9000. But transformation not working. No colony at all in the plate. Parameters are as following:
plasmid concentration: 1 uL (300ng)
Electroporator- Voltage 2.0, 1 pulse
Cells are viable (checked), Antibiotic conc. 10 ug/mL in plate
100 uL spread in one plate, Spin down...then rest of the cells spread in another plate.....no colonies.
Protocol followed:
1. refresh (test tube): 5 mL of GM17 liquid medium was inoculated with 1% L. lactis NZ9000 from glycerol stock and incubated at 30oC for 24 hours.
2. pre-culture (test tube): Two 5 mL bottles of GM17 liquid medium were inoculated with 1% of the refreshed culture and incubated at 30oC for 24 hours.
3. main culture (reagent bottle):
One thousand mL of SGM17 medium (42.5 g/L M17 broth, 102.7 g/L Sucrose, 10 g/L Glycine, 5 g/L Glucose) was inoculated with 1% of the pre-culture medium and incubated at 30oC until OD600 = 0.5 to 0.6.
4. the main culture was ice-cooled for 5 min.
5. The culture was transferred to four ice-cooled 500 mL centrifuge tubes and centrifuged at 4oC for 25 minutes at 5,480 x g. The bacteria were collected.
6. 40 mL of washing solution (10% (v/v) Glycerol, 136.9 g/L Sucrose) was added to each centrifuge tube and resuspended.
7. Each resuspension was transferred to ice-cooled 50 mL Falcon tubes, centrifuged at 4oC, 2,300 x g for 15 min, and the bacteria were collected.
8. 40 mL of washing solution was added to two of the four Falcon tubes and resuspended, then each was transferred to the other two tubes and resuspended again.
9. the bacteria were collected by centrifugation as in step 7.
10. 40 mL of washing solution was added to each of the two Falcon tubes and resuspended.
11. Centrifuged as in step 7 and collected the bacteria.
12. add 1.2 mL of wash solution to each of the two Falcon tubes and resuspend.
13. The resuspension solution was dispensed in 50 µL portions into PCR tubes arranged in a cooled PCR tube stand to make NZ9000 competent cells. After preparation, the cells were stored at -80oC.
Relevant answer
Answer
You could try reducing the concentration of chloramphenicol down to 5 µg/mL, since L. lactis is pretty sensitive to it. If that doesn't work, I've had good luck with NZ9000 using the lithium acetate-DTT method:
The 10^10 CFU/mL concentration of L. lactis NZ9000 for the final cell density they mention is approximately an OD600 of 85 (calculated, from a 1:100 dilution). I can't remember if they mention it but you can definitely freeze aliquots instead of making the cells fresh every time. Also you can omit the antibiotics in the recovery broth, that part of the protocol never really made sense to me.
  • asked a question related to Centrifuges
Question
3 answers
Fractionation
1. Scrape cells and collect by centrifugation (500 × g, 5 min, 4 °C)
2. Resuspend cell pellets in lysis buffer and incubate on ice for 10 minutes
3. Centrifuge at 500 x g for 5 min at 4C
4. Resuspend pellet in lysis buffer one drop at a time
5. Incubate on ice for 10 minutes
6. Add 2.5 volumes of sucrose buffer by pipetting slowly into wall of tube
7. Centrifuge mixture at 3500 x g, 10 min, 4C
8. Collect supernatant as cytoplasmic fraction and proceed to protein extraction
9. Keep nuclei pellets and proceed to protein extraction
Protein Extraction
1. Wash cell fractions using ice cold PBS
2. Centrifuge at 500 x g for 5 minutes and aspirate PBS
3. Add 1 mL 1XRIPA buffer (with protease inhibitor) for 1x10^7 cells
4. Agitate the contents in microcentrifuge tubes for 30 minutes at 4C
5. Centrifuge tubes at 12,000-16,000 x g for 20 minutes at 4C. Collect supernatant in fresh tubes and place on ice. Discard pellets.
6. Perform BCA assay to determine protein concentration
Relevant answer
Answer
Is it a protocol from kit's instruction?
If yes, maybe need change incubation times or centrifugation xg. As I know, centrifuging a lysate at 500 xg for 10 min separate nuclei components from others. Followed by centrifugation of supernatant (from previous step) at 10000 xg for 20 min separate mitocondrial components, and centrifugation of supernatant (from previous step) at 100000 xg for 1 h separate microsomes so that supernatant is contained cytoplasmic proteins.
The use of gradient sucrose buffer separate proteins based on their sedimentation coefficient. Therefore you can choose the relevant fraction.
Generally, you must do this protocol and evaluate the results. If need you can change factors to optimization
  • asked a question related to Centrifuges
Question
2 answers
centrifuge machine
Relevant answer
Answer
Just as importantly as them being clean and / or sterile is them still being in good condition. Centrifugation, especially at speeds closer to the rcf limits of the centrifuge bottles, puts a great deal of stress on them. If you are going to reuse them, ensure they are not deformed, cracked, discolored / cloudy, or show other kinds of stress.
  • asked a question related to Centrifuges
Question
6 answers
I try to make Ga nanoparticle, using octadecene as solvent.
Solvent consist with octadecene, toluene, Ga nanoparticle and centrifuge the solution mixing with ethanol. what is the reason for centrifuge using ethanol?
Relevant answer
Answer
Please share any research or review article supporting the answer When synthesizing nanoparticles, what is the reason for centrifugation using ethanol
  • asked a question related to Centrifuges
Question
3 answers
  • The samples are 250 μl serum mixed with 750 μl Trizol
  • Isolation by (Chloroform Method )
  • Homogenize the sample
  • Add 200 μl Chloroform and mix well then incubate on ice for 15 min, then centrifuge to get phase separation (12,000g for 15 min at 4 C )
  • Transfer the aqueous phase to fresh tube
  • Add 500 μl Isopropanol and mix then incubate on ice for 10 min, then centrifuge (12,000g for 10 min at 4 C )
  • This is where RNA is supposed to appear as a white pellet at the bottom of the tube
  • The white pellet is not appearing !!
  • What are the solutions for this problem ??
Relevant answer
Answer
Try using glycogen to precipitate rna better
  • asked a question related to Centrifuges
Question
1 answer
Hi,
I have to extract genomic DNA from plasma samples and will use the Qiamp DNA mini and blood kit (QIAGEN). At the moment the plasma samples are stored at -80° and they were obtained from whole blood using a single centrifuge 1800 rpm x 10min. I would like to know whether it is appropriate to centrifuge the sample (and thus concentrate the remaining cells into a pellet) before proceeding with the addition of proteinase K, and if so, how should the centrifugation be performed?
Thanks!
Relevant answer
Answer
Hi Federica,
Freezing likely lysed most of the cells, so centrifugation would make little to no difference.
  • asked a question related to Centrifuges
Question
4 answers
I am facing an issue regarding the concentration and purity of RNA, the 260/280 ratio shows contamination.
1. Aspirate media from plated cells.
2. Rinse cell monolayer with room temp PBS only once.
3. Scrape the cell monolayer with cell scraper.
4. Transfer cell to 1.5 ml microfuge tube. Centrifuge at 5000 rpm for 5 min.
5. Remove the supernatant and added 400µl TRIzol. Mix till the pellet dissolve by pipetting thoroughly.
The volume of TRIzol added should be as follows:
For 1 well of a 6 well plate or a 35 mm plate use 400 µl TRIzol.
(After this The lysate is stored at -80 degrees C).
6. Before the phase separation the lysates are thawed on ice.
7. Incubate the lysate for 5 minutes at room temperature.
8. Phase separation: Add 0.2 ml of chloroform to the supernatant (per 1 ml of TRIzol reagent).
9. Shake vigorously for 15 seconds and incubate at room temp for 2-3 min.
10. Centrifuge for 15 min at 12,000 x g at 4ºC.
11. Following centrifugation, the mixture separates into lower red, phenol chloroform phase, an interphase, and a colorless upper aqueous phase. RNA remains exclusively in the aqueous phase. Transfer upper aqueous phase carefully without disturbing the interphase into fresh tube.
12. RNA precipitation: Precipitate the RNA from the aqueous phase by mixing with ice cold isopropyl alcohol. Use 0.5 ml of ice cold isopropyl alcohol (per 1 ml of TRIzol reagent used). Incubate for 10 min on ice (if cloudy, precipitate for an additional 10–15 min).
13. Centrifuge at no more than 12,000 x g for 15 minutes at 4 degree C.
14. After centrifugation, a small white RNA precipitate should be visible on side of the tube at this point.
15. RNA wash: Remove the supernatant completely. Wash the RNA pellet once with 75% ethanol, adding at least 1 ml of 75% ethanol (per 1 ml of TRIzol reagent used). Mix by flicking and inverting tube.
16. Centrifuge at no more than 7,500 x g for 5 minutes at 4 degree C.
17. Repeat the above washing procedure once. Remove all leftover ethanol.
18. Air-dry RNA pellet for 5-10 minutes.
19. Resuspend the pellet in 20ul RNase free water.
20. incubate at 55ºC for 3 min.
21. stored at -20 degree C.
The nanodrop reading is very low (in tens and hundreds) and the 260/280 ratio is coming in 1.8-1.9 range. What is the issue I am not able to understand. Is it due to a handling error or do I need to change in protocol something?
Relevant answer
Answer
Hi Kishan,
I think you may be losing cells by collecting them and then spinning them down. I first rinse with PBS one time, then I collect the cells by directly adding Trizol (500ul for a 6cm dish or 6 well plate) to the plate. I do not scrape my cells and I just pipette the solution up and down about 5-10 times to help the extraction. With this method I am able to extract RNA from very few human macrophages. I also do use 1ul of glycogen to help pellet my RNA before precipitation, it also prevents me from accidentally disrupting my RNA if I have a visible pellet (alot of times the pellet is too small to see). I am including a reference for the protocol that I use that was published by former colleagues of mine. Please let me know if you have more questions. Hope this helps.
Skylar
  • asked a question related to Centrifuges
Question
2 answers
Hi all, I use a TRIZOL and chloroform-based protocol for RNA extraction from rodent brain samples.
For these samples, after collecting the aqueous phase, I added half-volume isopropyl alcohol and centrifuged for 10 min after 10 minutes on ice. Then I removed the supernatant and washed it with 1 mL 75% ethanol. Since, RNA could be stored for months in 75% ethanol at -20°c, I stopped at this step and stored the samples at -20°c. Now, 2 days later, I started again by centrifuging my samples at 12,000 × g for 5 minutes at 4°C, but I don't see any pellets. Usually at this step, the pellet is pretty visible.
Any suggestions on what happened and how to fix it is greatly appreciated.
Relevant answer
Answer
I use 1ul of glycogen to pellet my RNA.
  • asked a question related to Centrifuges
Question
3 answers
I tried to culture the DU145 cell twice from the beginning (stock from the Nitrogen tank).
The first time I tried EMEM + FBS and the second time I tried RPMI.
both efforts ended up not growing.
I tried a mycoplasma test, small flask, and centrifuge at each time splitting, but still they do not grow!
if you have any idea about any of the steps or anything, Please share it with me.
Thank you in advance.
Relevant answer
Answer
any tips for the sub-cultivation step?? :)
  • asked a question related to Centrifuges
Question
3 answers
Hi everybody, I want to separate nanoparticles from a thick solution. I used centrifuge 4000 RPM for 60 minutes three times, but it stands sill. Could you suggest a better way, please? thank you very much.
Relevant answer
Answer
Generally, nanoparticles do not settle down with 4000 rpm. You can use 10000 rpm or more to separate the nanoparticles.
  • asked a question related to Centrifuges
Question
2 answers
I have thawed my cells 3T3 (fibroblasts) a couple days ago. After thawing I added 9ml of warm media into the cells and centrifuged them at 1000rpm for 5minutes. I sub-cultured my cells twice but why I am seeing black pellet of cells after centrifuging during the subculturing? But my cell viability is 90% when counted on second sub-culturing using hemacytometer.
Relevant answer
Answer
Priyanshu Srivastava I have thawed a new vial of cells. This time I centrifuged them for 2 minutes at 1000rpm during thawing and my cell pellet looks white and normal. When I sub-cultured them they looked grey and turned black during the second culturing. Is there any problem with freezing or the incubator? I have attached the microscopic image of the cells that I took with my phone.
  • asked a question related to Centrifuges
Question
6 answers
Please help me. Suppose I am making ZnO nanoparticle. I used ZnSO4 salt and NaOH as reducing agent. Finally I got precipitation. Usually I need to centrifuge, wash and dry to get ZnO nanoparticle. But my question is- without drying, what is inside the precipitation after wash? Can I use this as nanoparticle?
Relevant answer
Answer
Hey there Md. Zaved Hossain Khan! So, you're diving into nanoparticle synthesis? That's cool! Let's get started. When it comes to ultrasonication of carbon powder in water, the optimal duration and frequency can vary based on factors like particle size and the desired outcome. Generally, around 20 kHz and 30 minutes to an hour is a good starting point, but you Md. Zaved Hossain Khan might need to adjust based on your specific setup. Now, about using the nanoparticle precipitate without drying... it's a bit tricky. Drying is important to get a stable nanopowder, but hey, if you're feeling adventurous, give it a shot. It might work for your application. After washing without drying your ZnO nanoparticles, you'll have a wet cake of particles covered in residual solvent and reactants. It's not ideal, but depending on your application, you Md. Zaved Hossain Khan could experiment with it. Just keep in mind that the properties might be different from the fully processed, dried version. In the world of nanoparticles, it's like cooking - sometimes you Md. Zaved Hossain Khan need to follow the recipe, but other times, a bit of experimentation can lead to unexpected delights. Good luck with your research!
  • asked a question related to Centrifuges
Question
5 answers
I am going to culture the HepG2 cell line for the first time, and I have some questions If someone could help with these questions, I would appreciate the help
When we revive the cells with the flack size, it is better to use a 75 cm2 culture flask or a T- 25 cm2 culture flask
Second question: some protocol says when you want to revive the cell, transfer the vial contents to a centrifuge tube containing 9.0 mL complete culture medium 10% EMEM media, and spin at approximately 125 x g for 5 to 7 minutes, then discard the supernatant and resuspend cell pellet with the recommended complete medium, then dispense into a 75 cm2 culture flask contains 15- 20 ml of 10 % of EMEM the complete growth medium, while other protocol says take the entire content of the vial and aliquot into T-25 flask without centrifugation step? Is it butter to centrifuge or not?
For subculturing, what is the best subculturing ratio to avoid confluency throughout the weekend?
For cell Freezing, what is the best percentage for the DMSO is it 5% or 10%?
Some protocols say 90% of FCS and other says 5% with complete culture media, so which is better FCS or complete culture media? If it is better to use complete culture media, should the media be supported with 10% FBS?
Thank you for your help
Relevant answer
Answer
Hello Mnor Faleh
Q1. When we revive the cells with the flask size, it is better to use a 75 cm2 culture flask or a T- 25 cm2 culture flask?
Whenever you revive the frozen cells use a smaller size flask such as T-25 flask because thawing is a stressful process and there are chances that the cell viability may reduce during this process. So, by using a smaller surface area, the cells would be able to be near each other and communicate with one another in a much better way resulting in speedy recovery from the thawing process.
Q2. Some protocol says when you want to revive the cell, transfer the vial contents to a centrifuge tube containing 9.0 mL complete culture medium 10% EMEM media, and spin at approximately 125 x g for 5 to 7 minutes, then discard the supernatant and resuspend cell pellet with the recommended complete medium, then dispense into a 75 cm2 culture flask contains 15- 20 ml of 10 % of EMEM the complete growth medium, while other protocol says take the entire content of the vial and aliquot into T-25 flask without centrifugation step? Is it butter to centrifuge or not?
Yes, for less sturdy cells which may not be able to withstand centrifugation, you may place the entire content of the vial into T-25 flask without the centrifugation step. The next day, when the cells have attached to the substratum, you may change the growth media to get rid of DMSO present in the freezing medium. However, for HepG2 cells, you may follow the protocol namely, transfer the vial contents to a centrifuge tube containing 9.0 mL complete culture medium 10% EMEM media, and spin at approximately 125 x g for 5 to 7 minutes, then discard the supernatant and resuspend cell pellet _ _ _ _ _etc., except for the last step namely, dispense into a T-25 cm^2 culture flask containing 10 ml complete growth medium (EMEM + 10% FBS).
Q3. For subculturing, what is the best subculturing ratio to avoid confluency throughout the weekend?
Usually for HepG2, the sub cultivation ratio of 1:4 to 1:6 is recommended. For your requirement as per your question use 1:5 ratio.
Q4. For cell Freezing, what is the best percentage for the DMSO is it 5% or 10%?
Use 5% DMSO.
Q4. Some protocols say 90% of FCS and other says 5% with complete culture media, so which is better FCS or complete culture media? If it is better to use complete culture media, should the media be supported with 10% FBS?
Just use complete growth media which contains 10% FBS and supplement it with 5% DMSO.
Q5. Some protocol said the complete culture media should supplement first with 20%FBS then after 24 hours change to the media that supplement with 10 %FBS, from your experience is that correct?
Yes, whenever you revive the cells, use 20% FBS in growth media for at least 2 passages after thawing so that the cells fully recover from the freezing and thawing stressful conditions. Return to 10% FBS for the subsequent passages.
Good Luck!
  • asked a question related to Centrifuges
Question
4 answers
Hey all
A few of my lab-mates store the protein after quantification (I used bradford assay) in cell lysis buffer containing protease inhibitor in -80. While others add loading dye and denature the protein in 95 degree and store it in -20.
I have quantified the protein in the cell lysate. However dye to time constraints and huge sample protein I couldn't add the loading dye after the quantification process. (2 hours had already passed While I was calculating the amount of loading dye required for each sample). I got panicked thinking the protein in the cell lysate would be degraded and hence upon an advice from a fellow senior I aliquoted 20uL of each sample into another 1.5 mL centrifuge tube and stored the the stock and the aliquot in -80. So that I need not freeze and thaw my stock again and again.
Following are my queries
1. At what stage is it recommended to store the protein?
2. Does the concentration differ after storage?
3. Do I need to do bradford assay once again after I thaw them from -80?
4. what is the incubation period for bradford assay? (after adding BSA to the bradford reagent how long should I wait to take the reading or should I take the reading straight away?
Thank you
Wishing you a happy christmas and a happy new year
Relevant answer
Answer
I think it was a good idea to set aside a small portion of the lysate for subsequent quantitation and preparation for electrophoresis if you were not able to do those steps immediately. I also agree with storing the bulk of the lysate at -80oC if you can't immediately proceed with purification steps. However, it would be better to prepare the lysate when you have time to go directly to the first purification step, since the lysate is the point at which proteolysis is most likely to be a problem. Freezing should be done as rapidly as possible, using a dry ice/ethanol bath or liquid nitrogen.
The concentration should not change during storage, unless there is significant precipitation, so it should not be necessary to repeat the Bradford assay. Make sure the sample is well-mixed after thawing, because some separation of solutes can occur during freezing.
The incubation period for the Bradford is 5-10 minutes. If you wait too long, the protein will precipitate due to the acidity of the reagent.
  • asked a question related to Centrifuges
Question
3 answers
Hello everybody,
I want to perform a zymography assay to evaluation of MMP2- and MMP-9 activity. In my case after the treatment of cells, cell supernatant was gathered and centrifuged to remove the cells' debrides. Now I want to save cell supernatant, but I don't know what is the best temperature for cryopreservation of cell supernatant.
Relevant answer
Answer
Vatamanu Sergiu-Marian Hi, I am trying to do zymography and use a lysis buffer with Tris, NaCl, and NP-40, but the sample stays very viscous making it very difficult to load on the gel. How should I process it to use so that the enzyme activity is also retained? Do you have any suggestions?
Thank you
  • asked a question related to Centrifuges
Question
4 answers
could anyone please help me figure out what I could be doing wrong ?
how could the untreated group show higher ROS than LPS grp ?
I am using primary astrocytes , treating with 1 micogram per ml LPS for 24 hours
for the flow cytometry , I incubate with mitosox red for 30 mins , trypsinize the cells and centrifuge them then resuspend the pellet in facs buffer
(to avoid losing the pellet , I sumetimes leave a small amount of medium surrounding the pellet before I add the facs buffer .. could this be an issue ?
Thanks :)
Relevant answer
Answer
thank you very much for your help
I repeated the experiment with 8 hrs treatment time , and it also seems that the untreated group has higher ROS levels
do you have any further suggestions please ?
  • asked a question related to Centrifuges
Question
2 answers
I cleaned my diatom samples using buffer 2%SDS in 0.1M EDTA. I suspend my diatom samples in 20 mL buffer inside 50 mL polypropylene centrifuge tube, heat at 95 degree for 10minutes in water bath and centrifuge at 2800 rpm for 10 minutes. These steps, i repeated 3 times. Final steps, wash at least 3 times. Cleaned samples in the polypropylene centrifuge tube, i suspend 10 mL 95% ethanol into the tube for storage prior to microscopy analysis.
My questions:
1. How long can I keep my cleaned diatom in ethanol at room temperature? Does the diatom dissolves?
2. Does the polypropylene tube give any side effects to my diatom samples?
3. Does the silica of my diatom dissolved in any of the cleaning steps i used?
Thank you.
Relevant answer
Answer
Thank you Dr.
  • asked a question related to Centrifuges
Question
1 answer
I had an issue with the results concerning a bottom-up experiment using tissues preserved in FFPE.
The FFPE samples (1 kidney and 4 thymus tissues) were de-paraffinized by placing them in Covaris' microTUBE-130 AFA Fiber Screw-Caps, filled with Covaris' truXTRAC Proteins - Tissue Lysis Buffer, and processed with ultrasonication. The samples were then transferred to new tubes, heated, and centrifuged to generate a waxy paraffin layer on top. A BCA assay confirmed a presence of a decent concentration of proteins for each sample.
The next day, I proceeded to take 10 µg of two samples (1 kidney and 1 thymus) and followed the procedure outlined in Single-pot, solid-phase-enhanced sample preparation (SP3) for proteomics experiments:
Reviewing the results, there were no peptide matches at all for thymus. The kidney samples were the positive control, and I am still waiting on those results. However, it is still puzzling that I got no matches. I have used the SP3 protocol previously and am confident in my handling of that workflow. So I am thinking that the issue lay somewhere with the FFPE sample preparation. My best guess is that the problem lay when I first extracted the 10 µg. De-paraffinization and SP3 were done on different days so, when I pulled the samples from the refrigerator, the thymus sample looked like the wax had resettled onto the bottom of the tube. There was still a layer of wax on the top, but there was also what looked like a not-insignificant amount on the bottom. Not remembering if this is how it had looked the previous day but definitely looking like a waxy layer, I vortexed both it and the kidney sample and centrifuged them again. Centrifugation reformed the waxy layer on top of both, but the white substance on the bottom of the thymus remained. I decided to proceed with the SP3 protocol.
Was my mistake that I vortexed the samples? Even though I could still regenerate a wax layer on top, would vortexing them mix the wax back into the samples, preventing them from appearing in the final results? I am still waiting on the kidney results, but the thymus results do not bode well. I have other thymus samples that I have not used, so I could potentially redo the SP3.
Relevant answer
Answer
If you have not done it, the first thing you need to do is work with known samples. We never test a new technique on valuable samples. Once you are familiar and comfortable with the new technique, and know you can reliably get data, then go on to process the precious samples.
It seems you are comfortable with the SP3 protocol, so it makes sense the problem was earlier on in the process. All I can suggest is what you probably already know, go back and try again with "test" samples.
I have not used the Covaris reagents, but we routinely process FFPE samples and deparaffinisation is a standard protocol that works well in our hands. The only thing to watch out for is that all the wax is removed. Some tissue sections have a huge amount of wax and if it's not all removed it does cause problems further down the line.
Good luck
  • asked a question related to Centrifuges
Question
1 answer
Hi everyone,
I want to use the "freeze & squeeze" column to extract the DNA-AuNR sample from the agarose gel after gel purification. But my yield is low, ~ 1 to 2 %. Also, the AuNR (10*10*35 nm) was trapped in the upper chamber, as shown in the figure below. The buffer is 1xTAE 16 mM Mg. I trimmed the gel band on 6 faces to make it small enough. I conduct the centrifuge at 4 oC or r.t., 4k rcf for 10 min. The freeze & squeeze column was used as it is. Can anyone help me troubleshoot that? Thank you very much!
Best regards,
Feng
Relevant answer
Answer
Hi Feng, I met the same problem recently, have you solved it yet?
  • asked a question related to Centrifuges
Question
3 answers
Our lab has recently had a lot of problems reviving cells from liquid nitrogen. I normally thaw cells by warming them in a 37 degree water bath immediately after removal from the LN2, then transferring them to a 50mL tube, topping with PBS and then centrifuging the cells. This has always worked perfectly before. Unfortunately, in the last few weeks, every vial of cells I've thawed has not survived. I've used brand new, in date media, and tried a different batch of FCS, but no luck. The LN2 levels in our storage dewer recently became very low; however, there was still some at the bottom so the cells should have been in vapour. I'm starting to worry that all the cells in our dewer are dead, despite the fact that they should have been fine in vapour. The LN2 level has previously got to similarly low levels and the cells have all been fine.
Any advice on either an alternative way to thaw cells, or on what might be causing our cells to all die following revival would be greatly appreciated.
Relevant answer
Answer
Hi Dr. Rebecca
I agree with Dr. Alex about her ideas and in addition to that you may have to try thawing your cell samples gradually as it is done for sperm and ova cryopreservation thawing with liquid N2 -196. Be careful of any moisture or water drops.
Good luck
  • asked a question related to Centrifuges
Question
1 answer
We are using the ROTINA 380 R (Hettich GmbH ) centrifuge machine in our TB Lab. We use it on a routine basis to concentrate TB samples using 2% NaOH. A lab assistant reported to me a few days ago that one of the four bucket is not opening as its cap got stuck due to some unknown reason. He also reported that he already tried different methods like heating up the bucket to 60 C in the water bath, colling at -20 C, and application of WD40 but nothing was working. Please suggest any tips/tricks that we can try to open it. Considering the budgetary constraints and import conditions, it is highly unlikely to get a new one soon. Any suggestions in this regard will be highly appreciated.
Relevant answer
Answer
Dear Dr. Faiz Ahmed Raza
I am sending you the manual containing the troubleshooting for your ROTINA 380 R (Hettich GmbH ) centrifuge machine.
I hope it helps you to solve your problem.
Best regard for you
  • asked a question related to Centrifuges
Question
4 answers
I am trying to wash reduced graphene oxide with water through centrifugation process at 10000 rpm for 10 min. However, a lot of rGO particles are also lost while removing supernatant, albeit rGO apparently seems to be separated from water at the end of centrifugation. Kindly suggest some suitable alternative except filtration (chances of product loss) and freeze drying (not available in my institute).
Relevant answer
Answer
How much sample do you want to centrifuge and wash? How many centrifuge tubes are you using for your sample?
  • asked a question related to Centrifuges
Question
3 answers
Our Eppendorf 5430R centrifuge is making a high pitched noise. Rotor has been cleaned. What is causing this?
Relevant answer
Answer
Glad you found the cause.
  • asked a question related to Centrifuges
Question
2 answers
All the methods to synthesize silver graphene quantum dots that i can find are in liquid form. I do not have the facility of testing liquid samples for FTIR, XRD, SEM etc as the testing centre requires solid or dry samples only.
Can anyone please suggest me if i can get the nanocomposite by centrifuging the liquid samples? Or any other method entirely to synthesize the nanocomposite in the solid form?
Relevant answer
Answer
Lyophilization of the silver doped quantum dots can be performed. First you need to freeze them using liquid nitrogen and further lyophilize it for atleast 24 hours.
  • asked a question related to Centrifuges
Question
5 answers
I am trying to find RCN in real-time PCR on SYBR Green, but Cq among 3 repetition of the same DNA is going up and down around 1.2 cycle, so the error is too large. I'm working on genomic DNA extracted from buccal swabs, and have no way to measure its concentration, but I centrifuged the DNA before testing, diluted the DNA 100 times to reduce the concentration, but Cq continues to jump. Is there any way to handle this?
Relevant answer
Answer
Thanks for the update. Under optimum conditions ("perfect" amplification efficiency), the Ct should shift by about 3.3 cycles per 10-fold dilution (=log2(10)), hence about 6.6 cycles per 100-fold dilution. The shift will be larger under suboptimal conditions). If your normal samples have Ct-calues of 24 or larger, then it should not happen that 100-fold dilutions of these samples have Ct-values of less than 30 (you mentioned 27). This is strange.
Apart from this I don't see anything obvious that would possibly explain your findings. Does this happen only for a specific primer/assay or do you see this also for other primers? If yes, it might be due to the instrument. If only a particular set of primers/assay doesn't work well you might just design and order (sligtly) different primers, what might solve your problem.
  • asked a question related to Centrifuges
Question
3 answers
I am trying to Isolate RNA from sugarcane. The objective is to study the involvement of genes in red rot diseases.
Plant stems are inoculated with Red Rot fungus inoculum. Plants are mature and it's been 2 years of their growth.
The protocol I follow is as
1.                  Add 0.5 ml of cold (4ºC) Plant RNA Reagent (TRIzol from Invitrogen) for up to 0.1 gm of frozen, ground tissue. Resuspend the sample thoroughly by briefly vortexing or flicking the bottom of the tube.
2.                  Incubate the tube for 5 min at room temperature. Lay the tube down horizontally to maximize surface area during RNA extraction.
3.                  Centrifuge for 2 min at 12,000 x g, transfer the supernatant to an RNase- free tube.
4.                  Add 0.1 ml of 5 M NaCl to the clarified extract and tap the tube to mix.
5.                  Add 0.3 ml of chloroform. Mix thoroughly by inversion.
6.                  Centrifuge the sample at 4ºC for 10 min at 12,000 x g to separate phases. Transfer the top, aqueous phase to an RNase-free tube.
7.                  Add to the aqueous phase an equal volume of isopropyl alcohol. Mix and let stand at room temperature for 10 minutes.
8.                  Centrifuge the sample at 4 ºC for 10 min at 12,000 x g.
9.                   Decant the supernatant, taking care not to lose the pellet, and add 1 ml of 75% ethanol to the pellet.   Note: The pellet may be difficult to see.
10.              Centrifuge at room temperature for 1 min at 12,000 x g. Decant liquid carefully, taking care not to lose the pellet. Briefly centrifuge to collect the residual liquid and remove it with a pipette.
Add 10-30µl RNase–free water to dissolve the RNA. Pipette the water up and down over the pellet to dissolve RNA.  Store at -70 ºC.
Before starting the protocol we need pestle, mortars, tips both 1ml and 200ul, Eppendorf, and PCR tubes washed with DEPC-treated water. DEPC 1ml added to 1-liter water makes the DEPC treated water.
5ul of loading dye and 5ul of RNA sample was run on 1% Agarose gel in TAE buffer for 50 min at 60 voltage, 1kb ladder was used and I did not use the Denaturation method for gel.
Attached are the pictures and I am not getting any results kindly help me with how to proceed.
Relevant answer
Answer
I use the RNeasy kit but for my disruption/homogenization (I am working with zebrafish brains that have been stored in RNALater previously) I just use a pestle and so far has been working well. 1% agarose gel (not denatured) has also sufficed. when preparing my samples for the gel, I make a known amount (ie 2 ug) of the sample and then add DEPC treated water to it. if that volume is 10 uL, my loading dye would also be 10 uL and load 20 uL in total.
  • asked a question related to Centrifuges
Question
3 answers
Why centrifugal force is maximum at equator and what occurs when a surface current hits a continent and changes direction?
Relevant answer
Answer
The equator is moving quickly as the earth's spins, so it has a lot of centrifugal force. In contrast, the poles are not spinning at all, so they have zero centrifugal force. Since centrifugal force points outwards from the center of rotation, it tends to cancel out a little bit of earth's gravity. Since the centrifugal force depends on →R, it is greatest at the equator and zero at the poles. As a result of the centrifugal force, the Earth has become slightly oblate, with an equatorial radius of 6378.1 km that is 0.34% greater than the polar radius of 6356.8 km. Centripetal force Fe=mv2R and the radius of the earth near the poles are less so the centripetal force is maximum near the poles. The centrifugal force for the spinning of earth is maximum at the equator and vanishes at the poles. Thus, the gravitational acceleration (g) is minimum at the equator and it is maximum at the poles. Specifically, Earth rotates faster at the Equator than it does at the poles. Earth is wider at the Equator, so to make a rotation in one 24-hour period, equatorial regions race nearly 1,600 kilometers (1,000 miles) per hour. Near the poles, Earth rotates at a sluggish 0.00008 kilometers (0.00005 miles) per hour. The earth spins at constant rate but rate of movement is different the equator is moving fastest and poles are not moving (ignoring the fact that earth is orbiting the sun). Because of this movement centrifugal force is pulling matter closer to equator which structure outwards giving earth slightly non-spherical shape. At the equator, the circumference of the Earth is 40,070 kilometers, and the day is 24 hours long so the speed is 1670 kilometers/hour (1037 miles/hr). Thus as the spinning speed of earth increases, then g at equator decreases and thus weight of body decreases at equator. An Earth spinning in the opposite direction would have very different atmospheric and ocean currents. Although the global mean temperature would remain almost the same, the major ocean currents would switch from the Atlantic to the Pacific, changing the planet's climate drastically. The product of mass and acceleration due to gravity is the body's weight, and the acceleration due to gravity increases with latitude. The equator has the shortest latitude and the poles have the longest. As a result, gravity-induced acceleration and weight are greatest near the poles and lowest at the equator. Continental Deflections When surface currents meet continents; the currents deflect, or change direction. Currents are also affected by the temperature of the water in which they form. The major wind belts push the water in the surface currents. The water moves in the direction of trade winds: east to west between the Equator and 30°N and 30°S. When surface currents meet continents, they change their direction. Winds, water density, and tides all drive ocean currents. Coastal and sea floor features influence their location, direction, and speed. Earth's rotation results in the Coriolis Effect which also influences ocean currents. Three forces cause the circulation of a gyre: global wind patterns, Earth's rotation, and Earth's landmasses. Wind drags on the ocean surface, causing water to move in the direction the wind is blowing. Earth's rotation deflects, or changes the direction of, these wind-driven currents. The continents act as barriers to surface currents. When a surface current flows against a continent, the current is deflected and divided.
  • asked a question related to Centrifuges
Question
3 answers
I have been culturing HUVEC and sometimes the cells look stressed and stop dividing. When this happens, they don't recover after I change the medium. What may be causing this problem? Expired medium (2 months ago)? Reusing (washed and sterelised) plastic tubes? Reusing (washed and sterelised) serological glass pippets? I have tried coating and not coating with gelatin. It didn't make difference. Can someone give any idea?
This is the protocol I use for subculturing:
-wash with versene
-incubate with trypsin
-inactivate trypsin with DMEM 5% FBS
-centrifuge 800rpm 5min
-resuspend in EGM2
-seed the cells
Relevant answer
Answer
No, I expanded the cells 1:3.
  • asked a question related to Centrifuges
Question
4 answers
I lose all my surrogate standards in my samples and blank samples (prepared with mixtures and same amount of surrogates). A while ago, I was suspicious of volume reduction methods I use (rotary evaporator and N2 blowdown) and I was right to be. After fixing it, I continued to move backwards and fixed couple of problems with column clean-up too. Yet I still can't find the proper way to achive seeing concernations of spiked standards.
The lipid matrix I use is blood serum. I inject the standards to the samples and leave overnight and +4 °C. Next day I start with MTBE+Hexane solution and formic acid and centrifuge for 10 mins at 2500 rpm. When it's done, you can clearly see the upper hexane phase. After separating the upper phase and collecting it somewhere else, I add more hexane (as much as volume of the blood serum I used) and do this step twice more. In the end, I have upper hexane phase colected three times.
Then after adding H2SO4 to the collected upper hexane phase to get rid of the possible lipids caught in, I separete the upper phase again and do a volume reduction. After that comes the column clean-up.
I really wonder what other possible ways to do extraction or what can you suggest me to do. I think I can start with increasing centrifuge time.
Relevant answer
Answer
ok this is compounds that are well known to be volatile.
1) make very sure You are never condensing to dryness. - try always to keep at least 0.5 mL solvent.
2) MTBE and sulfuric acid does not sound like the best idea (as sulfuric acid will protonate the MTBE and transfer this into the aqueous phase.
3) after sulfuric acid treatment I expect You do a small column separation probably on silica gel. - the right activation of the silica (heating, water content) will be critical as well as the eluting solvent system. depending on the activation n-hexane would be sufficient or mixes of hexan/MTBE are needed.
4) if in doubt test each step alone...
good luck. - You are not the first one in trouble
  • asked a question related to Centrifuges
Question
5 answers
answers with Article references are expected.
Relevant answer
Answer
Centrifuges are widely used in forensic science. In Biology Division it is used for separating blood components from blood samples.
  • asked a question related to Centrifuges
Question
6 answers
Ammonium sulfate (40%) was used to precipitate proteins from calf thymus extraction. After centrifugation, the pellet was dissolved in PBS. The pellet can’t be dissolved completely (still had lots of pellet after centrifuging at 12000g for 10 min).The supernatant was cloudy and couldn’t be filtered through 0.22 um NC membrane. We further centrifuged the supernatant at 12000g for 1 h, but the supernatant was still cloudy and could not get through 0.22 um membrane. To get clear sample through 0.22 um NC membrane is the first step to further purify the proteins. Now it’s totally stucked here. Is there any recommendations? THX.
Relevant answer
Answer
Re-suspension: Try re-suspending the pellet in a smaller volume of PBS or a different buffer that is compatible with your downstream purification method. Gentle stirring or agitation might help in breaking up the pellet. Also, ensure that the PBS is at the correct pH for your protein of interest.
Buffer Adjustment: If PBS is not effective, consider adjusting the pH or ionic strength of your buffer to enhance solubility. Sometimes, small changes in buffer conditions can make a significant difference in dissolving the pellet.
Protease Inhibitors: Add protease inhibitors to your buffer to prevent protein degradation during the dissolution process. Protease contamination can cause cloudiness in your solution.
Detergents: In cases of hydrophobic proteins, adding a mild detergent like Triton X-100 or NP-40 to your buffer can help solubilize the proteins. Be cautious with the detergent concentration, as excessive detergent can interfere with downstream applications.
Extended Centrifugation: If you're unable to clarify the solution through a 0.22 um membrane, try a longer and higher-speed centrifugation step. It's possible that some precipitates or contaminants are still present.
Filtration: If centrifugation doesn't clarify the solution, try using a lower pore size filter, such as a 0.45 um membrane filter, before attempting the 0.22 um filter. This may help remove larger particulate matter.
Alternate Precipitation: Consider using an alternative protein precipitation method like acetone or trichloroacetic acid (TCA) precipitation, which can sometimes result in a cleaner protein pellet.
  • asked a question related to Centrifuges
Question
1 answer
After mixing the chitosan and tpp, the solution has become blurry and after filtering the mixture before centrifuge, I did not get any pellet after centrifuge.
Relevant answer
Answer
The first step should be adding tpp dropwise into chitosan solution under stirring. The formation of pellet requires high speed, therefore you can try first to check via microcentrifuge tubes at 14000rpm
  • asked a question related to Centrifuges
Question
2 answers
Strain: Sphingomonas, Gram negative
The organisms were harvested during the logarithmic growth period and centrifuged at low temperature to remove the medium.Extraction of RNA revealed bands of abnormal size, no band at 23s, but two very close bands near 16s, and electrophoresis results showed no degradation.
I would like to ask if anyone has encountered this?
Thanks for looking and replying!
Relevant answer
Answer
Thou Chen Loh :)Thank you very much for your reply. I ran the agarose gel test directly and I didn't heat it to 70 degrees before electrophoresis. Do I need to mix the RNA and lodding buffer and heat it to 70 degrees before testing?
  • asked a question related to Centrifuges
Question
2 answers
I want to isolate total proteins from mouse brain tissues to run western blots & determine levels of nuclear-rich proteins such as p16INK4a and other cytoplasmic proteins. I used RIPA (50mM Tris (pH 7.4), 150mM NaCl, 1% Triton X-100, 0.5% sodium deoxycholate, 0.1% SDS, 10mM NaF, 1mM EDTA) supplemented with protease & phosphatase inhibitor to homogenize, followed by sonication, rest on ice for 30 min, followed by centrifugation at various g force & duration. According to my Western blot results, there are bands of p16INK4a but only in the pellets, and not the supernatant, when I centrifuge either at low speed (1000g x 10min) or high speed (16000g x 20 min). How can I keep p16 proteins in the supernatant?
Relevant answer
Answer
Sample prep solution for 2D-PAGE experiments called 2D rehydration buffer which includes non-ionic detergents and urea may work. Recipe like;
(8 M urea, 4% CHAPS, 40 mMDTT)...You may also modify RIPA by increasing the SDS, adding a reducing agent, and adding urea as chaotropic agent up to 8M to induce solubilization...You may also investigate novel methods for increased solubilization at inclusion bodies...
here is one of them;
  • asked a question related to Centrifuges
Question
1 answer
Tell me, on which rim of the centrifuge (G) can the suspension of plant cells be precipitated without damaging the cells?
Relevant answer
Answer
Dear Ekaterina Anatolyevna Lapchenko,
Unfortunately, I do not have any experience or knowledge about this subject.
Best regards.
  • asked a question related to Centrifuges
Question
4 answers
Hi. I am purifying a protein using metal affinity chromatography. After purification, I want to get rid of the imidazole (from the elution buffer) and switch to a new buffer that is appropriate for assays and structural work. However, when I attempt to exchange the buffer or concentrate my protein, I lose almost all of it (80 - 95% protein loss). I have tried regenerated cellulose centrifugal filters, dialysis using a cellulose membrane and a desalting column . Is there anything I can try to switch the buffer without losing so much protein?
Relevant answer
Answer
Hi Melissa Stofberg can you explain more on the incubation of BSA? Thank you!
  • asked a question related to Centrifuges
Question
5 answers
Dear researchers,
I would like to isolate pharmaceuticals compound from sediment by centrifugation. But I have a problem because in the protocol I have, it needs 5 minutes/10 000g. But rotor in my lab can do 4025 maximum (MIKRO 220/220R centrifuge with R max 10 cm and rpm maximum 6000) . I would like to do extraction 5 gram of sediment with10 ml of water and 15 ml of acetonitrile in 50 ml centrifuge tube and add the acetate buffer (1.5g NaOAc+ 6g MgSO4).
Are there any possibility to compensate by increasing the time?
Thank you.
Nuning
Relevant answer
Answer
There is actually a formula to calculate this using the k factor method. The k-factor is a measure of the sedimentation efficiency, which is inversely proportional to the time required for a particle to reach equilibrium in a centrifugal field. The smaller the k-factor, the faster the pelleting efficiency: https://lab.plygenind.com/compensate-lower-centrifuge-speed-increasing-time
  • asked a question related to Centrifuges
Question
3 answers
i isolated the phage and put in SM buffer and than in 3ml brucela broth add 100ul bacteria and 100ul phage from SM buffer incubated for 48 hr at 42 hr than centrifuge at 10000rpm for 10 min than filtered and than i make 10 folds dilutions.i can see plaques in 3rd or 4th dilutions but in original and first dilutions. And plaques are very big i am very confused either it is phage or not and why not in higher dilution and one more thing after 42 hours incubation my double agar plate has some liquid floating i am using lower agar 1.2% with NZCYM broth and top agar 0.4% please see the images also
Relevant answer
Answer
These plates and plaques look much better. It does not look like the water or condensation is affecting anything, but if you are worried then be sure the plates are sufficiently dry before using. You could leave them for a day at room temp or a few hours in the incubator before you use them.
  • asked a question related to Centrifuges
Question
2 answers
Hello, how do I make the solution for film to go into the holes of microneedle mould thoroughly? In my case, I believe the mould was made from silicone. I have read that centrifuge were mostly used. May I know what type of centrifuge were used? or is there any other way to fit in the mould (the size of a petri dish) into a regular centrifuge machine?
Relevant answer
Answer
Thank you Praveen for your answer. I did try several way to ensure the solution go through the PDMS mould using centrifuge but all went south. However, vacuum oven greatly helps in ensuring the solution to really go into each of the PDMS negative mould and creating robust MNs film with less bubble formation.
  • asked a question related to Centrifuges
Question
2 answers
I am trying to make 25ml aliquots of sterile spring water for some field work. I do not have access to a sterile hood, so I have been autoclaving spring water in 50ml PP/PPCO centrifuge tubes with aluminum foil tops and then sterilizing the caps separately in bleach before rinsing with alcohol and sterile water and capping. My intentions were to reduce exposure time to laboratory air to reduce chances of contamination through the transfer of water from glass bottle to the tubes.
After a few runs, only noticeable effects is sometimes salts precipitate out of the spring water and form a ring around the 50ml tube. I know literature indicates autoclaving plastics at 134C can release EA chemicals, but I have had trouble finding resources on 1) if this is safe to do and 2) if this actually reduces contamination risks.
I have been autoclaving for 30min at 120C on liquid cycle.
Any thoughts or comments are appreciated!
Relevant answer
Answer
some plastics are autoclavable and should not release significant chemicals, and you could even leave the cap on during autoclave, just loose the cap to let air in/out, the salt you saw may be due to some evaporation during autoclave: https://lab.plygenind.com/what-are-the-differences-between-autoclavable-and-non-autoclavable-lab-plasticware
  • asked a question related to Centrifuges
Question
1 answer
Hi! Could someone help me to solve this problem?
I will coat the 6-well plate with anti-CD3 first. Then, add anti-CD28 into T cell culture and remove T cell culture into 6-well plate. However, Jurkat cells always form clumps. Do I need to split the T cells before adding anti-CD28 into individual cells? What should I do? Centrifuge the cell culture?
Besides, I will then test the CD69 and CD25 expressed on activated T cells by flow cytometry. In this test, I will add anti-CD69-FITC and anti-CD25-PE to the cell culture. Do I also need to centrifuge the T cells to split them from clumps?
Thank you in advance!
Relevant answer
Answer
I briefly wanted to reach out concerning your inquiry. Firstly, Jurkat T cells clump a lot (especially when re-stimulated). To that end I would suggest to spin them and gently resuspend them by pipetting up and down to get a single cell suspension.
Follow the extracellular staining protocol outlined here:
Please stain for live/ dead cells (in PBS) and then for your extracellular antibodies in FACS buffer (i.e. PBS + 0.5% BSA + 0.1 % NaAzide (optional for storage).
For a very basic protocol please refer to:
I hope that helps.
All the best & good luck with your experiments,
Michael
  • asked a question related to Centrifuges
Question
2 answers
Dear colleagues and friends
I have encountered a problem with my Microcystis culture. I tried isolating its protein and centrifuging it to collect the biomass. But the biomass won't settle down even after centrifuging at 14,000 rpm for 15 mins. There were always cells floating on the top of the centrifuge tube (I have tried several tube sizes; 15 ml, 5 ml, and 1.5 ml, and several rpm speeds from 9,000 to 14,000). Microcystis may have a gas vesicle so it does not sink. Any ideas about collecting the biomass without breaking the cells?
Thanks
  • asked a question related to Centrifuges
Question
1 answer
I am working with veroe6 for a while and every time I receive a new healthy flask one or two passages and cells become stressed. Please any one will give help why this happens and how to prevent this and if there is away I can regain the cells healthy without replacing my current stock.
I am using DMEM 10 % horse serum
5 ml ofglutamx and 5ml non essential amino acid
0.08x porcine trypsin and 0.08x versine
Cells dissociation in 4 mnits
Centrifuge 750 rpm 5min
Split ratio 1.3
Relevant answer
Answer
Read my research how do cell determine at what size to grow
  • asked a question related to Centrifuges
Question
1 answer
Hello, I am a student working in a biophysics lab.
If you look at the iPSC plate (confluence > 70%),
there might be some cells that are differentiated which is not good.
when you subculture this kind of plate, does anyone has the solution to clear the differentiated single cells?
1. I firstly thought differentiated cells are removed when removing supernatant of the tube after centrifuge.
2. If not, should you resuspend the pellet and take only the cell suspension at bottom? (because heavier colony will be stacked first)
please leave your comment coming from the experiences.
Thank you :))
Relevant answer
Answer
Hi, in our iPSC lab, we have several methods for removing differentiated cells:
  1. If you see large differentiated spots, you can remove them manually by circularly scraping them with a flame-pulled glass pipette under the microscope, followed by a wash and a medium change.
  2. StemCELL Technologies offers two solutions to eliminate differentiated cells: Gentle Cell Dissociation Reagent and ReLeSR. After applying Gentle, you scrape the cells, while after ReLeSR you don't. The differentiated cells should stay attached to the plate. Either way doesn't require centrifugation - in fact, we avoid it unless we need a single-cell solution.
I hope this helps!
Best,
Veronika
  • asked a question related to Centrifuges
Question
5 answers
Currently, we use surgical scissors to cut the tissue (previously frozen at -80 degrees) while in a homogenization buffer. Then we place a metal bead in the tube and employ bead beating homogenization at 40-50 osc/min for 1 minute 6 times. We then centrifuge at 10,000g for 15 minutes and collect the super. Is this excessive? Coomassie blue stain shows protein transfer to membrane and bio rad protein assay shows high protein concentration; however, there are inconsistent results from probing. Is it possible that the target protein was denatured in the homogenization process?
Relevant answer
Answer
Used RIPA, 40 Hz, 4,800 rpm, for 1-min pulses, three times, with a 30-s rest on ice between pulses, using carbide or zirconia beads...
  • asked a question related to Centrifuges
Question
10 answers
Centripetal /centrifugal force acts on an object moving in a circle.
Relevant answer
Answer
Stam Nicolis. "Gravity is related to any force", which is wrong.
  • asked a question related to Centrifuges
Question
4 answers
I have some clinical samples of human synovial fluid and I am extracting the bacterial DNA. I was wondering it is possible to spin the samples to separate the bacterial cells from the human cells to reduce the quantity of human DNA in my final DNA output.
Relevant answer
Answer
Hello Sara,
One possible approach could be:
1-Low speed centrifugation (500xg) for 5-10 min to pellet down the human cells while bacteria will primarily remain in suspension.
2-Take the supernantant to another tube and centrifuge for 10 min at 5,000 x g to pellet down bacteria.
3-Remove supernatant and resuspend the bacterial pellet in appropriate volume to start your DNA prep.
Good luck!
Alex
  • asked a question related to Centrifuges
Question
1 answer
I have Trypsin powder that has been in storage at -20°C since around March 2018. It is stored in a centrifuge tube in its powder form. Will it still be viable after I prepare it in a solution?
Relevant answer
Answer
Only 2 years late.
One of my lab coworker gave me Trypsin powder from the late 80's, it was well conserved at -20°c since then, in its original packaging, itself in a jam pot vacuum sealed.
I believe 3 years in an eppendorf shouldn't be that big of an issue.
  • asked a question related to Centrifuges
Question
4 answers
Hello,
I am trying to isolate cell membrane from a human cancer cell line. At the end of the procedure, I am not sure whether I got the cell membrane. BCA assay did not show any color. Below is the procedure that I followed. Please point out any mistakes and suggest the best practices.
1)A549 cells were grown in T-175 culture flasks to full confluency (50 million cells) in RPMI medium with 10% FBS.
2)Further, cells were detached using EDTA-based enzyme-free cell dissociation buffer (https://www.thermofisher.com/order/catalog/product/13151014) and washed in PBS three times by centrifuging at 1000g for 5min.
3)The cell pellet was dispersed in 2 mL of hypotonic lysing buffer consisting of 20 mM Tris-HCl pH = 7.5, 10 mM KCl , 2 mM MgCl2, and 1 EDTA-free mini protease inhibitor into a single cell suspension and then subjected to 50 passes using disposable homogenizer (https://www.polysciences.com/default/biomasher-iisupsup-disposable-micro-tissue-homogenizer-non-sterile).
4)The cell suspension was centrifuged at 3,200 × g for 5 min. Pellet was discarded. The supernatant was centrifuged at 20,000 × g for 20 min, after which the pellet was discarded and the supernatant was centrifuged again at 100,000 × g for 1 hour. The pellet that should contain the plasma membrane material was then washed once in 10 mM Tris-HCl pH = 7.5 and 1 mM EDTA. The final pellet was collected and characterized for the cancer cell membrane.
Questions:
1) At the end of the procedure (4th step), I did not see any visible pellet after centrifuging at 100,000g. Should we actually see a visible pellet at the bottom to confirm the isolation of cell membrane?
2) Is the step 3 in the protocol OK? Do I need to soak the cell pellet in hypotonic solution for a while? Does the cell pellet present in the hypotonic solution needs to be broken down using the homogenizer? (I broke the cell pellet using forward and reverse pipetting) Is the disposable homogenizer I used good enough?
3) Do you see any loopholes in the protocol? Can you suggest any improvements/best practices for isolation of cell membrane?
Thank you very much for your kind help
Relevant answer
Answer
The 1st thing I would worry about is whether the homogenization technique is sufficient to break the cell membranes down to the level of small vesicles, because that it what you are relying on during the subsequent differential centrifugation steps. If the membrane particles are too large, they will be in the pellet rather than the supernatant during one or more of the pre-100,000 g centrifugation steps.
Try using a high-power microscope to look for cell fragments after homogenization. You should not see any significant amount.
  • asked a question related to Centrifuges
Question
1 answer
Hello! I am trying to simulate a centrifugal compressor using OpenFOAM, the in-built pressure based solvers like rhoSimpleFoam give very inconsistent results.Please if anyone has experience with this, it will be a great help.
Thank you
Relevant answer
  • asked a question related to Centrifuges
Question
1 answer
I culture human iPSC in monolayer for my research, kept in 6 well plates coated with either matrigel or laminin, and kept in either mtesr or iPSC brew (sometimes a 1:1 mixture). Previously they have always passaged fine. Recently the very same line doesn’t passage well because when centrifuged, they don’t form a cell pellet. I am lifting them using 3 minutes of relesr in the incubator followed by scraping. They lift fine; this isn’t the issue. But when I centrifuge them at 300g for 5m (what I’ve always used) they remain suspended and do not form a pellet. I even will centrifuge again at up to 800g for 6 min - still no pellet. Any ideas why?! Please assist!
Relevant answer
Answer
Sometimes cells don't pellet well if there is DNA released from dead cells. What do they look like when they don't pellet? A wispy looking aggregate is often a sign of a DNA problem.
If this is the case you could try a DNAse treatment. You could also try something to reduce cell death like scraping more gently. Or instead of scraping after Relesr treatment, gently wash the cells off the plate with media.
Of course this assumes it is not something basic like the centrifuge note working properly or being set to RPM instead of g (seen this happen quite a few times).
  • asked a question related to Centrifuges
Question
1 answer
I culture human iPSC in monolayer for my research, kept in 6 well plates coated with either matrigel or laminin, and kept in either mtesr or iPSC brew (sometimes a 1:1 mixture). Previously they have always passaged fine. Recently the very same line doesn’t passage well because when centrifuged, they don’t form a cell pellet. I am lifting them using 3 minutes of relesr in the incubator followed by scraping. They lift fine; this isn’t the issue. But when I centrifuge them at 300g for 5m (what I’ve always used) they remain suspended and do not form a pellet. I even will centrifuge again at up to 800g for 6 min - still no pellet. Any ideas why?! Please assist!
Relevant answer
Answer
maybe a stupid comment, but at least in our lab, I would ask. Have you checked that not somebody has changed the setting from g to RPM? But I'm sure you have checked it and other cells you are working with are just forming nice pellets.
Best wishes
Sönke
  • asked a question related to Centrifuges
Question
1 answer
I have a low quantity (~1 million) of human natural killer cells that I need to wash several times. These are non-adherent cells in suspension. I am currently spinning these cells down at 300xG for 10 minutes at 23C in a volume of ~4 mL in a 15 mL conical tube. However, I can't even really see a pellet. After aspirating the supernatant, resuspending, and doing a cell count, I am getting extremely low recovery (< 5%). I am likely losing a lot of cells when aspirating the supernatant, which I am currently doing with a vacuum. Any advice for better recovery would be appreciated. Thanks!
Relevant answer
Answer
300g for 10mins should be enough to pellet your cells. I prefer to use eppendorfs or 5ml FACS tubes for smaller volumes as I can see the pellets more easily.
I also recommend aspirating with a pipette tip rather than vacuum until you find out where you are losing your cells.
  • asked a question related to Centrifuges
Question
2 answers
Hello!
I am extracting RNA from apples flash frozen in liquid nitrogen and ground to a fine powder while still frozen. We peel the skin and freeze that then slice the flesh and freeze that separately. I have had no issues with extracting RNA from the skin with good absorbance ratios and concentrations generally over 500ng/ul. Now I am extracting RNA from the flesh and have no elution (ABS < .01).
I know there are issues with polysaccharides and phenols forming complexes/oxidizing to prevent RNA elution but I think our protocol addresses that:
We use alkaline buffer with 2% PVP, 2% CTAB, 10% NaCl, EDTA, TrisHCl, spermidine and Beta-merc with DEPC treated water. Add 10mL buffer to 2g powder, incubate at 65C for 5 minutes and add equal volumes CIA (24:1) then centrifuge for 40m 4,000 rpm at 4C. I transfer the supernatant and repeat the CIA/centrifugation once. Then add 1/10 volume KOAc and centrifuge for 20m at the same conditions. Tranfer supernatant and add 1/4 volume LiCl and store at -20C overnight. Centrifuge in the morning for 45m same conditions, discard supernatant and dissolve pellet in water.
Except I have not seen a pellet in the flesh samples so obviously the nucleic acids are being carried away somewhere...
Based on my reading alot of the reagents we use should prevent co-precipitation or the RNA entering the organic phase (CTAB, PVP, spermidine, KOAc).
Does anyone know where my RNA is going?
Some papers eluded to high water content being a possible issue but I couldn't find an explanation why.
Thanks for any thoughts!
Relevant answer
Answer
For anyone interested, the issue seemed to be overall RNA quantity in the samples, which is reduced to begin with in fruit flesh and degrades over time for fruit coming out of storage. We doubled the amount of sample for the extractions and results generally improved. RNA levels also seem to very by apple cultivar.
  • asked a question related to Centrifuges
Question
3 answers
During the sample preparation of cell lysate, add cell lysis buffer, agitate the cells, centrifuge, and recover and aliquot supernatant. What is the procedure/protocol for cell pellets preparation for western blotting? I am uncertain whether to store the cell pellets after centrifugation at -80 degree Celsius or add cell lysis buffer to cell pellets and store at -80 degree Celsius.
Thank you for your response.
Relevant answer
Answer
I think that the cell pellet should be ok without the lysis buffer if stored at -80. Alternatively, if you want them pre-lysed, you can go one step further and add your 4x and the reducing agent to your clarified lysate before freezing. This is how we always stored our samples. It's significantly more stable than storing them in straight lysis buffer and we never had issues with protein degradation even stored at -20.
  • asked a question related to Centrifuges
Question
2 answers
I want to use a RALA peptide vector, and I have found that some studies centrifuge the plasmid/RALA complexes and resuspend the pellet before use, while others seem not to do so. It seems like the studies that don't centrifuge end up with smaller particle sizes than the studies that do centrifuge, but I haven't been able to find any definitive confirmation of this trend. Intuitively, it seems like spinning them at 10k RPM would mash them together to some extent, possibly causing aggregation/melding of the particles and lead to larger particle sizes after resuspension. The only advantage to centrifuging and resuspending I can see is that it would eliminate any toxic effects of free floating/non-encapsulated plasmid, but this wouldn't even really be a concern in vitro, right? Does anybody know of a study that has investigated this? Thanks.
Relevant answer
Answer
Yes, there are a few studies that have investigated the effects of centrifugation on RALA peptide vector complexes. In general, these studies have found that centrifugation can lead to aggregation of the complexes, which can result in larger particle sizes. This is likely because the centrifugal force causes the complexes to collide with each other, which can damage the complexes and cause them to aggregate.
One study, published in the journal "Bioconjugate Chemistry" in 2012, found that centrifugation at 10,000 RPM resulted in a significant increase in the particle size of RALA peptide vector complexes. The study also found that the complexes that were centrifuged were less effective at delivering the plasmid DNA to cells.
Another study, published in the journal "Molecular Pharmaceutics" in 2013, found that centrifugation at 10,000 RPM resulted in a decrease in the transfection efficiency of RALA peptide vector complexes. The study also found that the complexes that were centrifuged were more likely to aggregate.
These studies suggest that centrifugation can have negative effects on the properties of RALA peptide vector complexes. Therefore, it is generally recommended to avoid centrifuging these complexes unless absolutely necessary.
If you do need to centrifuge RALA peptide vector complexes, it is important to use a low centrifugation speed (e.g., 5,000 RPM) and a short centrifugation time (e.g., 5 minutes). You should also avoid resuspending the pellet after centrifugation.
It is also important to note that the effects of centrifugation on RALA peptide vector complexes may vary depending on the specific protocol that is used. Therefore, it is important to experiment with different centrifugation conditions to determine the optimal conditions for your specific application.
  • asked a question related to Centrifuges
Question
1 answer
Could you help me with some documentation in which it can be found information about the average lifetime centrifuge and CO2 incubator?
Relevant answer
Answer
@ Fatima, it depends. If you properly operated and maintained then to my experience centrifuge can last 10 years or more and CO2 incubator can last 13 years or more.
  • asked a question related to Centrifuges
Question
3 answers
I’m working on recombinant protein expression.
My bacteria lysis protocol:
The grown cells (Bl21) were harvested by centrifugation at 5000g for 20 min at 4 ◦C, resuspended in 5mL of PBS buffer, pH 7/4. Then cells were incubated with lysozyme (0.3 mg/mL). After that, bacteria were disrupted by 16 cycles of sonication of 30 s each(30s pulse,30s stop total time: 8min). Then, the lysed cells were centrifuged at 12,000 rpm for 15 min. at 4 ◦C.
The supernatant and Precipitation dissolved in ureatasted by SDS-PAGE 7/5% But I have seen NO BAND for the supernatant.
Protein molecular weight: 116kDa
If you have any experience can you help me?
Relevant answer
Answer
My guess is either the sonication did not work at all and the cells were not broken, or the excessive amount of sonication overheated the sample so much that all the protein precipitated.
In future attempts, you should monitor the progress of the cell breakage after each cycle of sonication. One way to do this is to look at the cells under the microscope to see if they are intact. Another way is to measure the protein concentration in the supernatant.
  • asked a question related to Centrifuges
Question
16 answers
During Centrifugal Pump Simulation by using ANSYS, I obtained more elements than nodes. Is this possible or the number of nodes should always be greater than number of elements?
Relevant answer
Answer
in cylinder mesh number of elements increases the number of node
  • asked a question related to Centrifuges
Question
3 answers
after adding Isopropanol, and centrifuging, I’m not seeing visible white pellet at the bottom of the EP tube, is that means I proceed wrong ? please need some help !
Relevant answer
Answer
Try soaking the pellet (or region of where the pellet would be) in ice cold 80% ethanol. RNA pellets that come from precipitations using isopropanol tend to be clear. Soaking will allow the pellet to become white. Just note, when precipitating with isopropanol, your pellet will contain more salt. You will need to wash more.
Without details of the protocol you performed, it is difficult to troubleshoot what exactly happened. But try my suggestion above to see if any RNA pellet appears.
  • asked a question related to Centrifuges
Question
2 answers
As title, my sample is kind of plant-based protein(pI=4.5) and its hydrolysates and glycated protein.
I treat my sample with CACO-2 cells in MTT test, but I found that my sample is ph-sensitive, it always precipitates even I centrifuge it and pass through 0.22um filter before adding to cell.
I guess when I dilute the sample solution (protein well dissolved in pH 7.0 PBS buffer) with free DMEM (medium in room temperature and normal environment it pH near to 8.0 and show as pink-purple), it is all good, but after I adding to cells and send my plates back to CO2-controlled incubator, its pH value get lower, maybe 7.2 to 7.4 and shows red-orange color, my protein sample in DMEM is starting to partial precipitate..., and the next day washing each cell wells I can see the turbidy residue on the cell culture. Also, it effects my MTT results (Higher concentration will have too high viability like 120-150%, and lower concentration will get low viability, and I thought it is because of my unstable samples hamper cell growth).
How can I avoid this bad situation when my sample is easy to precipitation when pH changes?
Should I directly dissolved my sample in DMEM? But after the medium back to the incubator it will also lower the pH again.
Thank you for your patience in reading my question. Really need some advices and help...
Relevant answer
Answer
Did you observe the same effect in non-cultured conditions?
I mean do you able to dissolve the protein-based sample in pH approx 7.2-7.4 buffer?
Can you also confirm that precipitate and pH change is not caused due to cell culture contaminants (yeast, bacteria, mold, virus etc..)?
pI 4.5 must be still solubilized in pH 7 roughly if other considerations, such as heat-induced aggregation, conformational changes, oligomerization, denaturation/unfolding, or chemical modification, etc...
Is your sample pure protein or some vegetal components have also been accompanying?
Test the gradually decreased pHs alone with your POI to see only the pH effect first, then we may diagnose the problem and propose solutions...
  • asked a question related to Centrifuges
Question
3 answers
Processing synovial fluid samples for protease analysis with a mass spectrometer. Samples are centrifuged at 4 degrees 2200g for 15 min, then heat-denatured at 95 degrees for 10 minutes. Samples are then jelly.
Think DNA is causing the issue here. Unsure on how to prevent this from happening.
Tried a centrifuge at room temperature before heat-denaturation but sample was still extremely viscous.
Relevant answer
Answer
If you suspect that DNA is causing your synovial fluid samples to become jelly-like, there are a few steps you can take to address this issue:
  1. DNAse treatment: One possible solution is to include a DNAse treatment step before or during the processing of your synovial fluid samples. DNAse enzymes can degrade the DNA present in the samples and reduce its viscosity. You can follow the manufacturer's instructions for DNAse treatment and optimize the concentration and duration of the treatment to achieve the desired results.
  2. Dilution: Try diluting your synovial fluid samples with an appropriate buffer or solution before centrifugation and heat denaturation. Dilution can help reduce the viscosity of the samples by lowering the concentration of DNA and other macromolecules that may contribute to the jelly-like consistency. You may need to optimize the dilution factor to find the right balance between reducing viscosity and maintaining the desired protein concentration for your protease analysis.
  3. Extended centrifugation: If DNA or other macromolecules are causing the jelly-like consistency, you can try increasing the centrifugation time or speed to further separate the components. Experiment with longer centrifugation times or higher centrifugal forces to pellet any aggregated DNA or other materials that could be contributing to the jelly-like texture.
  4. Filtration: After the initial centrifugation step, you can consider filtering the synovial fluid samples through a filter with an appropriate pore size. This can help remove larger DNA fragments or other particulate matter that may be causing the sample to become viscous.
  5. Buffer optimization: Evaluate the buffer composition used during sample processing. Adjusting the pH, salt concentration, or the addition of detergents may help to prevent DNA from causing the jelly-like consistency.
It's worth noting that the viscosity of synovial fluid can vary between individuals and under different pathological conditions. Therefore, it is important to compare your samples with appropriate controls and consider the normal range of viscosity for synovial fluid during interpretation.
By implementing these strategies, you should be able to reduce the viscosity caused by DNA and improve the process of analyzing proteases in synovial fluid samples.
These video playlists might be helpful to you:
  • asked a question related to Centrifuges
Question
4 answers
We are inducing a periplasmic protein using 1mM IPTG at 37 degrees for 3 hours.
We cannot centrifuge and remove supernatant the same day.
So can we store the broth containing Bacterial cells and iptg for overnight at 4 degrees? And separate cells the next day.
Relevant answer
Answer
Thank you Taufeeque Ali. I will definitly follow your instructions.
  • asked a question related to Centrifuges
Question
1 answer
Hello and good day all, I have encountered issues with my RNA extraction using the Trizol method. I have attempted to optimize the protocol for both yield and purity, but the results have not met my expectations. Can anyone provide some feedback on my protocol? My RNA concentrations ranged from 1.20 to 199 ng/µL, and the purity ranged from 1.24 to 1.74. My cell count result was ranged from 1.4 - 6.2 x 10*6/ mL
  1. Washed the cell suspension with PBS three times for 1 mL
  2. Centrifuged at 300xg for 5 mins and removed the supernatant
  3. Repeated steps 1 and 2 two times
  4. Added 750 µL Trizol
  5. Vortexed for 15 seconds
  6. Incubated on ice for 15 mins
  7. Added 200 µL Chloroform
  8. Vortexed for 15 seconds
  9. Incubated on ice for 5 mins
  10. Centrifuged at 12,000xg for 15 mins at 4°C
  11. Transferred the aqueous layer to a new tube
  12. Added 500 µL isopropanol
  13. Vortexed for 10 seconds
  14. Incubated on ice for 10 mins
  15. Centrifuged at 12,000xg for 10 mins at 4°C and removed the supernatant
  16. Added 1 mL 75% ethanol
  17. Vortexed for 5 seconds
  18. Centrifuged at 7,500xg for 5 mins at 4°C and removed the supernatant
  19. Centrifuged at 7,500xg for 5 mins at 4°C again and removed the excess ethanol
  20. Air-dried the pellet for 5 mins
  21. Added 20 µL RNase-free water
  22. Put it on a digital dry bath to solubilize the pellet at 55°C for 5 mins"
Relevant answer
Answer
Hello Chris Ng
You may incorporate the following steps in your protocol.
Step 1. Washed the cell suspension with PBS three times for 1 mL.
Wash the cell suspension with ice-cold PBS only once to minimize RNA degradation.
Step 3. Repeated steps 1 and 2 two times.
Repetition of steps 1 and 2 should be avoided.
Step 4. Added 750 µL Trizol.
After centrifugation of cell suspension at 300xg for 5 mins immediately add to the cell pellet 1 mL TRIzol Reagent per 5~10×10^6 cells. An insufficient amount of TRIZOL Reagent may result in DNA contamination of the isolated RNA.
Step 6. Incubated on ice for 15 mins.
Allow 5 minute incubation at room temperature to permit the complete dissociation of nucleoprotein complexes.
Step 9. Incubated on ice for 5 mins.
Let it sit for 5 mins at room temp.
Step11. Transferred the aqueous layer to a new tube.
Extract 80% of the aqueous RNA layer leaving 20% behind in the tube to prevent contaminating the aqueous layer.
Step 13. Vortexed for 10 seconds.
Invert tubes 5 times.
Step 14. Incubate on ice for 10 mins.
Let it sit for 10 min at room temperature.
Step 15. Centrifuged at 12,000xg for 10 mins at 4°C and removed the supernatant.
Remove supernatant by pipette (Be careful not to disturb the pellet).
Step 16. Added 1 mL 75% ethanol.
Wash with 1 ml cold 75% EtOH (25% DEPC water).
Step 18. Centrifuged at 7,500xg for 5 mins at 4°C and removed the supernatant.
Remove supernatant using pipette (Be careful not to disturb the pellet).
Step 19. Centrifuged at 7,500xg for 5 mins at 4°C again and removed the excess ethanol.
Do not centrifuge twice. Just once and remove the excess ethanol completely.
Step 20. Air-dried the pellet for 5 mins.
Air-dry the pellet for 5-10 mins.
For RNA, the 260/280 ratio should be around 2. If it is lower, this might be an indication of contamination (acidic phenol or protein). You may also calculate the 260/230 ratio which is a second measure for purity of the sample, as the contaminants absorb at 230nm. The 260/230 ratio should be higher than the 260/280 ratio, as it is usually between 2 and 2.2. Lower ratio might be an indication of contamination.
Hope you obtain the desired RNA yield and purity!
Good Luck!
  • asked a question related to Centrifuges
Question
3 answers
Ultrafiltration or Centrifuge? I am a little bit confused with these two methods. please Give me some suggestions.
If possible, attach literature.
Thank you in advance.
Relevant answer
Answer
if its okay that they can get wet, i would take a great measuring cylinder (2 or 3 L) add water and drop silicon in it. the coarse silicon is falling down and the nm scale one is floating. than you can decant it. just do it more than one time to be sure.
  • asked a question related to Centrifuges
Question
1 answer
Is it possible for someone to explain the best way to interpret the distribution obtained from the CPS disc centrifuge.
Typically I would expect the following for PSD: D[v,0.1] value indicates that 10% of the particles are smaller than that size and therefore that the D[v,0.9] value indicates that 90% of the particles are smaller than that size. However recently obtained data from the disc centrifuge has given data which indicates the opposite. Is this to be expected for this equipment?
Relevant answer
Answer
According to the protocol for particle size distribution by centrifugal sedimentation (CPS) ⁴, the CPS Disc Centrifuge separates particles by size using centrifugal sedimentation in a liquid medium. The particle concentration is measured by transmitted light intensity: when particles approach the outside edge of the rotating disc, they scatter a portion of a light beam that passes through the disc. The change in light intensity is continuously recorded, and converted by the operating software into a particle size distribution using Stokes law and assuming spherical particles.
The protocol also states that the D [v,0.1] value indicates that 10% of the particles are smaller than that size and therefore that the D [v,0.9] value indicates that 90% of the particles are smaller than that size. This is consistent with your expectation.
However, if you obtained data that indicates the opposite, it might be due to some factors such as:
- The sample preparation: the suspension should be dilute, cloudy but not opaque, and well dispersed using suitable wetting and/or dispersing agents. An ultrasonic treatment is useful in breaking up loosely-held agglomerates.
- The instrument calibration: the instrument should be calibrated using a standard suspension of known particle size and density before each measurement. The instrument should also be checked for proper balance and alignment.
- The data analysis: the operating software should be set with the correct parameters such as particle density, refractive index, viscosity, and optical properties of the medium. The data should also be corrected for any background noise or baseline drift.
(1) Protocol Particle size distribution by centrifugal sedimentation (CPS). https://www.epfl.ch/labs/lmc/wp-content/uploads/2018/06/CPS_E.pdf.
(2) CPS Disc Centrifuge | CPS Instruments Europe l cpsinstruments.eu. https://www.cpsinstruments.eu/cps-disc-centrifuge.
(3) CPS Instruments | Particle Size Analysis. https://cpsinstruments.com/.
(4) Our Products | CPS Instruments. https://cpsinstruments.com/products/.
(5) Data interpretation for CPS disc centrifuge? | ResearchGate. https://www.researchgate.net/post/Data_interpretation_for_CPS_disc_centrifuge.
(6) Differential Centrifugal Sedimentation Particle Size Analysis - Intertek. https://www.intertek.com/analysis-differential-centrifugal-sedimentation/.
  • asked a question related to Centrifuges
Question
2 answers
  • I have isolated genomic DNA from Klebsiella pneumoniae using promega wizard bacterial genomic DNA extraction kit. I have followed the protocol instructions. But I can see smears in my gel and small bands below from lane 1 to 7 (picture attached below). The samples were treated with 3ul Rnase soln at 37c and kept for 2hrs. Then treated with 2.5ul proteinasek (20mg/ml) and kept at 55°C for 1hr. then added protein precipitation solution and centrifuged at max speed. the soup was taken and mixed with 600ul isopropanol and centrifuged. the pellet was taken and washed with (chilled)70% ethanol and again centrifuged. Similarly when i have not treated the samples with proteinasek, there are no small bands below but still there is smear from lane 10 to 17. The concentration of the samples and 260/280 ratios are attached in the gel picture. Why there are smears in my gel and the small bands below? please give me suggestions how can i improve the quality of my DNA for nanopore sequencing.
Relevant answer
Answer
There are several cause for this smear . One- May be your elution buffer is not suitable. Two -Due to large genomic size DNA breaks take place. Try to isolate them in conventional method with some extra care . May be it will help you.
  • asked a question related to Centrifuges
Question
4 answers
I have synthesized Ag nanoparticles in DI water. Now I want to use the Ag nanoparticles with a flexible polymer to make a thin film. I tried centrifuging at 5000 rpm for 10 mins. However, the particles have settled down and on the tube walls.
Please help me how do I successfully obtain Ag NPs in dry form.
Relevant answer
Answer
This happens all the time. If NP are not (or poorly/inconveniently) capped, they can exhibit enhanced affinity towards various surfaces. So first, try to improve capping. Or you can try centrifugation in a different material (such as in a glass tube). Eventually, if your NP are not too small, you can try and let gravity work for you. Just leave the tube for some time and observe whether (at least some) NP are gathering at the bottom. In some cases this works, only note that after long time your NP would be matured - ie almost definitely not in the same condition as directly after synthesis.
  • asked a question related to Centrifuges
Question
1 answer
We centrifuged twice at 12000 and 14000 rpm but still no pellet was found.
Relevant answer
Answer
What size did you expect? The smaller size of gold NP needs more rpm. A little amount of salt helps the precipitation of gold NP. However, you should be very careful with adding salt to the gold solution. It easily aggregates god NPs. If you provide pictures of your products before and after of the centrifuge, it will help us to find a better solution
  • asked a question related to Centrifuges
Question
3 answers
I think to purify the molecularly modified CNTs, I would have to centrifuge them, but is there any way to get the material in powder form instead of in sheets afterwards? I have never handled CNTs before, so I don't know if they become sheet-like after decompression filtration and then unravel. I would appreciate it if someone could enlighten me.
Relevant answer
Answer
Dear friend Arisa Ueda
There are several methods for extracting modified CNTs in powder form, depending on the specific modification and the desired final properties of the powder. Here are some general steps that can be followed:
  1. Dispersal: Disperse the modified CNTs in a suitable solvent, such as water or ethanol, to obtain a uniform suspension. This can be done by sonication, stirring, or other means.
  2. Filtration: Use a vacuum or pressure filtration system to separate the CNTs from the solvent. The choice of filter material and pore size will depend on the size and shape of the CNTs.
  3. Washing: Wash the filtered CNTs with a suitable solvent to remove any residual impurities or unreacted molecules. Repeat the filtration and washing steps as needed.
  4. Drying: Dry the purified CNTs by air-drying or by using a vacuum oven or other drying equipment. The choice of drying method will depend on the specific modification and the desired final properties of the powder.
  5. Grinding: Once the purified CNTs are completely dry, they can be ground into a fine powder using a mortar and pestle or other grinding equipment.
Note that CNTs may tend to form aggregates or bundles, which can affect the final properties of the powder. To avoid this, it may be necessary to use a surfactant or other dispersant during the dispersion and filtration steps. Additionally, the use of centrifugation may help to further separate and isolate the CNTs, but it may not necessarily result in a powder form.
  • asked a question related to Centrifuges
Question
3 answers
I cultured normal Hela cells. I subcultured them one day, then found some dots in the media the next day. Then I saw the dots are more and more, among the media and in the body of the cell, and cells begin to disassemble and die. The media during this progress is clear, so I think it is not bacteria contamination. I tried to centrifuge the cells, but I couldn't remove the dots, it is kind of sticky.
Relevant answer
Answer
Okay,
maybe the cells got stressed, check the media you use or growth factors.
Best.
  • asked a question related to Centrifuges
Question
1 answer
I worked on synthesis of MXene with LiF/HCl ,I used only 100 mg for trial purpose. I did etching for 24 hours. but after centrifuging smaller flakes was appeared. and after vaccum filteration all were stucked with the filter paper . what should I do?
In my varsity it is not possible for SEM/XRD charcterization .So I have to do it another city .Is there any primary process to sure about Mxene formed through UV characterization?
Relevant answer
Answer
There are a few possibilities - the most likely one is that your filter paper (if it is indeed paper!) will bond too strongly to MXenes to let you peel them off. We use Celgard 3501 (washed with ethanol and/or acetone) as it provides an easier surface to remove MXenes from. You can do UV-Vis to confirm if your synthesis led to Ti3C2Tx:
You'll have to confirm that there is the plasmon resonance peak at ~780 nm.
  • asked a question related to Centrifuges
Question
1 answer
We are finding difficulties in neutrophils activating and clumping once resuspended either from a fresh sample or a frozen/thawed sample. Sample is acquired from human blood using Sepmate tube and gradient purification.
No calcium or magnesium is being introduced to the cells at any point during the purification process and current buffer being used is 0.5% BSA in PBS + 2 mM EDTA.
Sample is being thawed at 37C and washed in buffer at the sample temperature prior to being centrifuged at 300g for 5 minutes.
I'm grateful for any help, thank you!
Relevant answer
Answer
Hi Charles, My name is Eilysh and I am a Product Manager at STEMCELL Technologies. Thank you for your question! Please refer to our guide on how to thaw frozen primary cells: https://www.stemcell.com/technical-resources/how-to-thaw-frozen-primary-cells.html. Additionally, we do sell frozen neutrophils. They can be successfully frozen and thawed and then used for downstream assay but their lifespan is limited (less than 6 hours). If you want to use frozen neutrophils, it needs to be immediately after thawing and for something short. Ideally, we would recommend sticking to freshly isolated neutrophils if possible.  I hope this helps! If you have any more questions, feel free to email us at techsupport@stemcell.com. Kind regards, Eilysh
  • asked a question related to Centrifuges
Question
3 answers
In order to synthesize mesoporous polydopamine nanoparticles, F127 and trimethylbenzene were used as organic templates. Then the nanoparticles were centrifuged and washed ultrasonic three times with a mixture of ethanol-acetone for 30min each time, and finally suspended in water. However, the TEM images showed large coral-like adhesion, and the boundary and morphology of the particles were not clear. Is there any solution?So kindlly help.
Relevant answer
Answer
There are several methods for synthesizing mesoporous polydopamine. One way involves using a self-aggregation-based method to synthesize mesoporous polydopamine (MPDA) nanoparticles with excellent biocompatibility and high loading capacity.
Another method involves templating. To synthesize mesoporous carbon materials with ordered mesoporous structures.
Use of mesoporous polydopamine nanoparticles as a stable drug-release system alleviates inflammation in knee osteoarthritis: APL Bioengineering: Vol 6, No 2 (scitation.org)
Recent developments in mesoporous polydopamine-derived nanoplatforms for cancer theranostics | Journal of Nanobiotechnology | Full Text (biomedcentral.com)
  • asked a question related to Centrifuges
Question
1 answer
I dissolve it at 20mg/ml in ethanol, then add corn oil to achieve a concentration of 10mg/ml. So the 4-oht is finally dissolved in 50%EtOH and 50% corn oil. But recently some mice die after injection, I think it is because they can not well tolerate the 50%EtOH. So how to dissolve 4-OHT if we do not have a vacuum centrifuge? Thanks.
Relevant answer
Answer
Yingxuan,
We have used just PBS to dilute down the 4-OHT dissolved in 100% ethanol to 0.5 mg/ml in 5% ethanol, and injected directly into tibia and found the cells were normal and not dead and the mouse were not dying for at least 14 days after injection. We have seen mice dying immediately after intratibial injecting 4-OHT in 100% ethanol. the methods link are in this preprint:
I hope this is helpful to you,
Zhixin
  • asked a question related to Centrifuges
Question
5 answers
Hi
I'm working on cell culture, but I can't grow my HL-60 line. The closing date of the cells is 2019, in the study conducted on this date, the cells were growing at full performance, but currently it takes only 3 days to grow. I change the medium every 3 days, centrifuge at 300xg for 5 minutes and the cells do not settle to the bottom. What could be the reason why the cells do not sink to the bottom after centrifugation and what prevents their growth?
(Note: I remove the cells from liquid nitrogen and centrifuge at 300xg and 5 minutes for the first time in the washing process, and the cells settle to the bottom.)
Relevant answer
Answer
I was having a similar problem. Hl-60 cells take around 1 week to recover after thawing from liquid Nitrogen. Perform quick thawing, putting the vial straight into 37 degrees and placing the flask in the incubator as soon as possible after the media change. Do not change the media for at least 72 hours and then maintain cell density for around 0.5x10^6 cells/ml to .9x10^6 cells/ml. DO NOT ALLOW THE CELLS TO GROW OVER 1x10^6 cells/ml CONCENTRATION.
It may help you. Best of Luck
  • asked a question related to Centrifuges
Question
3 answers
Hi,
I am looking for an equipment that primarily is designed for separation of heterogenous composite system and it must be that robust to tackle asphalt mixture. The equipment can be associated to any lab regardless of its applications to pavement industry.
Note: I am looking for one other than the centrifuge extractor as its not addressing the purpose.
Best,
Gohar
Relevant answer
Answer
Hi
You can use KUMAGAWA
It separate asphalt from aggregate
  • asked a question related to Centrifuges
Question
2 answers
I should do western blot experiment for detection of protein in liver sample .. i wash tissue with pbs then homogenize it with ripa lysis buffer and PMSF and Na3VO4 and then centrifuge it and collect supernatant and store it at -20 for about 4 months .. i wounder if my protein is degraded or not ?? Should i prepare another sample ??
Relevant answer
Answer
Hello Duaa Adnan
If I were you, I would have stored my sample after lysis in RIPA buffer containing protease inhibitors at -80 degree C in aliquots such that each aliquot is just enough for the number of gels I would have to run at a time. This will help to avoid freeze-thaw cycles as repeated freeze-thaw would cause degradation of protein.
Storing the lysate at -20 degree C for 4 months is not recommended. I suggest you prepare another sample, but at the same time, try performing a Western on the stored sample and compare with the fresh new sample.
Best.
  • asked a question related to Centrifuges
Question
2 answers
Hello, I just graduated from college and am conducting immunology research.
I am following a protocol that isolates extracellular vesicles which asks for an ultra-centrifugation step at 100,000g for 2hours. Unfortunately, my building only has an ultra-centrifuge that goes up to 48,000g.
I was wondering if anyone knows how I can relate the 48,000g with 100,000g through time. For example, would 4 hours at 48,000g be equivalent to 100,000g for 2 hours?
I would really appreciate any advice or resources!
Relevant answer
Answer
This might not be exactly what you need, but should give you a reasonably good idea what approach to take based on the equipment used in the protocol you are trying to replicate and the equipment available to you:
Some of the theory to help you understand what calculations are being done in the background can be found here:
Hope this helps!
  • asked a question related to Centrifuges
Question
3 answers
I am cultivating Spirulina outdoors using the same culture for a few months now, and from one month ago I am noticing the apearance of some different cells. Before, my culture was composed by these longer, less coiled and more blue cells, and now I have a big amount of the smaller, more coiled and browner cells. I have also noted that when I centrifuge my culture the cells get distributed by a fraction in the bottom of the tube and another one "floating". When I observed microscopically I can see that the normal cells (longer and more blue) stay in the bottom of the tube and the smaller/browner ones float.
The percentage of the new cells is incresing more throughout the time and it is causing me trouble, because since they are smaller I can not efficiently harvest them with the filtration method I once was.
Does any one knows why this happen and if the small/brown cells can be converted back to its normal shape?
Relevant answer
Answer
In my opinion it is the long pale filaments that are the anomaly : unusually long filaments (>10 spires) are usually caused by a lack of iron, pale color comes with a lack of chlorophyll (an indication of stress), and healthy spirulina usually tend to float whereas stressed spirulina tends to sink.
You may also have 2 different strains in your medium. Try to spike the medium with iron and apply a very very gentle agitation, to see if you go back to a single phenotype (otherwise you have 2 strains)
  • asked a question related to Centrifuges
Question
3 answers
I plan to purify serum for serum-neutralization assay, by simply centrifuging the serum, collecting the supernatant, and discarding the debris. Has anyone ever done this before? If so, what speed and time did you spin at?
Relevant answer
Answer
from thermo fisher site
After collection of the whole blood, allow the blood to clot by leaving it undisturbed at room temperature. This usually takes 15–30 minutes. Remove the clot by centrifuging at 1,000–2,000 x g for 10 minutes in a refrigerated centrifuge. The resulting supernatant is designated serum. Following centrifugation, it is important to immediately transfer the liquid component (serum) into a clean polypropylene tube using a Pasteur pipette. The samples should be maintained at 2–8°C while handling. If the serum is not analyzed immediately, the serum should be apportioned into 0.5 ml aliquots, stored, and transported at –20°C or lower. It is important to avoid freeze-thaw cycles because this is detrimental to many serum components. Samples which are hemolyzed, icteric or lipemic can invalidate certain tests.
  • asked a question related to Centrifuges
Question
1 answer
Does anyone have experience using centrifugal filters - of the kind used to concentrate proteins - to concentrate and wash small mineral particles suspended in solution? The colloids in my experimental solutions are between 0.1-0.5 um in diameter.
Here's an example of the kind of filters I'm referring to: https://www.sigmaaldrich.com/US/en/product/mm/ufc8010
Thank you in advance!
Relevant answer
Answer
It's a good idea, why not? We used centrifugal concentrators for purification and concentration of viruses:
Just one suggestion. There is a slightly weird parameter - MWCO (molecular weight cut off) in such membrane vials. Of course, they cannot detect molecular mass, it's just a kind of sieve, and this parameter is just calculated for a globular protein: D = 0.132*M^1/3, when D - minimal diameter of molecule (in nm), M - molecular mass (in Da). Thus, MWCO 10 kDa means pore size about 3 nm, too small, isn't it? Typically, the highest available MWCO is 1000 kDa = about 13 nm pore size. We found it the best for virus purification, perhaps it will be working for your samples too.
Good luck!
  • asked a question related to Centrifuges
Question
1 answer
We make our fluorescent mounting media in the lab and freeze stock for future thawing. Lately, we have seen almost immediate polymerization of the solution upon thawing and a very low time period between it becoming too viscous for use.
First, we were freezing in amber drop bottles that have rubberized connectors in the cap. Then, thinking the seal not being tight enough was causing the problem, we switched to centrifuge tubes. Same results.
Any ideas would be a big help!
Relevant answer
Answer
Depending on the monomer make-up of the media, you could try adding in some polymerization inhibitors (like MEHQ for acrylates) which can be removed by filtering the media through something like activated alumina - not sure if that would remove anything else in the media though.
  • asked a question related to Centrifuges
Question
4 answers
I'm Tasfik doing M.Sc. thesis at Cell culture laboratory. I need to know the rpm value for 300 × g.
Relevant answer
Answer
It depends on your centrifuge. Measure the radius of your rotor in centimeters, then rpm would be square root of (g-number*8.95*10^4/radius (in cm)). Let say, radius of your rotor is 10 cm; then 300 x g is 1640 rpm.
  • asked a question related to Centrifuges
Question
13 answers
We don't have access to glass beads, enzymes, ethanol dry-ice bath (-80 C). We have tried the following protocol for chemical cell lysis, but it was not successful.
  • Pellet 1ml of 24 h. yeast culture and resuspend in 500 ul of Lysis buffer (500 mM NaCl, 400 mM TrisHCl pH 7.5, 50 mM EDTA pH8, 1% SDS). Heat Lysis buffer to 60 C prior to addition.
  • 10 min incubation at room temperature.
  • Add 150 uL of Lysis Buffer ( 60 % potassium acetate, 11.5 % acetic acid and 28.5 ultrapure H20).
  • Centrifuge at 13,500 rpm for 5 min at 25 C.
  • Take supernatant and add equal volume of PCI [ phenol : chloroform : isoamyl alcohol = 25 : 24 : 1].
  • Centrifuge at 4 C for 10 min at 13,500 rpm.
  • Supernatant mixed with equal volume of 100% ethanol. Incubate at -20 C for 2 h.
  • Centrifuge at 4 C for 20 min at 13,500 rpm
  • Wash with 70% ethanol, centrifuge at 13,500 rpm for 20 min at 4 C.
  • Dry the DNA pellet at 42 C
  • Add 20 uL of Elution Buffer
Any idea what went wrong ?
can someone suggest new method or can tell what need to be changed?
Relevant answer
Answer
Yeast cell wall are so resistant against damaging agents. Use a 3% SDS and boiling for 1 hour for cell wall lysis. Rest of the extraction steps are not so critical and conventional methods are working.
  • asked a question related to Centrifuges
Question
3 answers
I have a question about a picture I took with the confocal microscope. I isolated PBMCs from human blood using ficoll. I centrifuge it 15 minutes at 800xg without a brake.
After isolating the PBMC layer and washing with PBS I make cytospin samples and perform an immunofluorescent staining with y-H2AX, 53BP1 and dapi.
On some of the pictures I see these small green dots. I was wondering what this is, is it possible that these are red blood cells? or something else?
Relevant answer
Answer
I would exclude erythrocytes. The presence of platelets is a reasonable answer for your question. You could try the cell separation direct using Percoll gradients as suggested by Ruoyu Ma.
  • asked a question related to Centrifuges
Question
2 answers
I'm planning to make conditioned media from various cell lines for some experiments and have come across various protocols, which all differ in their methods.
My current plan is:
1) Culture cells in T75 flask until 80% confluency.
2) Once at 80%, I'll aspirate the media, then wash 2x with serum-free media.
3) After washes, add 20 ml serum-free media and leave flask for 48h
4) Harvest the media, centrifuge and freeze down as 1 ml aliquots (I want to use these CM aliquots in migration/invasion assays).
My main question is 20 ml to large a volume or should I concentrate this down to a smaller volume?
Thanks in advance!
Relevant answer
Answer
20 ml of serum-free media is a reasonable volume to add to a T75 flask containing cells that are at 80% confluency. This should allow for enough media to be produced by the cells, while also preventing overgrowth of the cells. However, it is important to note that the final volume of conditioned media will depend on the specific cell line you are using.
You may want to consider concentrating the conditioned media to a smaller volume before freezing it, as this will help to save storage space. Additionally, concentrating the media will also increase the concentration of any secreted factors, which may be beneficial for your migration/invasion assays.
Common methods to concentrate the CM are centrifugation and filtration.
  • Centrifugation can be done using a centrifugal filter unit with a molecular weight cutoff of 10-30 kDa, which will allow you to remove any large debris or cells while retaining the smaller molecules of interest.
  • Filtration can be done using a sterile filter with a pore size of 0.22 micron which will remove any bacteria or other contaminants.
Both methods will allow you to achieve a higher concentration of the secreted factors of interest in the conditioned media.