Science method

Confocal Microscopy - Science method

Confocal microscopy is an optical imaging technique used to increase optical resolution and contrast of a micrograph by using point illumination and a spatial pinhole to eliminate out-of-focus light in specimens that are thicker than the focal plane. It enables the reconstruction of three-dimensional structures from the obtained images
Questions related to Confocal Microscopy
  • asked a question related to Confocal Microscopy
Question
5 answers
I am doing PI staining for spinal cord tissue of rats. But the problem is that even at very low concentrations PI is staining the whole cell instead of just the nucleus. I have provided RNase treatment also.
Relevant answer
Answer
May consider using Permai fluorescence dye.
  • asked a question related to Confocal Microscopy
Question
5 answers
I'm trying to do some live/dead staining of s. aureus and e. coli on the confocal microscope using Cyto 9 and Propidium Iodide. The cyto 9 is being taken up and imaging really well but the propidium iodide is not being taken up by the cells as well (these should definitely be dead). Can anybody suggest an optimum concentration/incubation time for the PI please?
Relevant answer
Answer
May consider using Permai fluorescence dye.
  • asked a question related to Confocal Microscopy
Question
4 answers
I'd like to use the confocal to track spermatogenesis in various coral colonies  over the season without having to do too much histology.  But, I'm not sure if this is possible with the confocal/fluorescent dyes.  Side bonus... corals and their symbionts auto-fluoresce green/red respectively.  So, looking for something in the blue (400-450 nm) and yellow (565-600 nm).  If it's possible for one to target spermatogenesis and the other to track oogenesis, then that's just lovely.
Relevant answer
Answer
May consider using Permai fluorescence dye.
  • asked a question related to Confocal Microscopy
Question
2 answers
Hello,
I performed immunocytochemistry and captured images with confocal microscopy. I wanted to measure the fluorescence intensity of different experimental conditions, may i know how many number of cells i can evaluate through imagej from each experimental condition?
TIA
Relevant answer
Answer
Hello, the simple answer is to determine how many cells you need in order to get an assay window of greater than or equal to 2 with the appropriate positive control.
You can also test various numbers of cells for negative and positive control for each end point then use z prime factor to see what gives you the best performance. Zhang et al, 1999, J Biomolecular Screening, 4:67-73
  • asked a question related to Confocal Microscopy
Question
1 answer
Hi! I was staining my tissue with both PI and Calcein AM dyes, but I realize a lot of cells were stained by both PI (red) and Calcein AM (green), for example, the cell at the center of the attached image. I thought PI stained for dying cells while Calcein AM stained for live cells.. so I am curious to know if those cells are actually dying or alive (or both...??).
Also, some of the red signal was really blurry.. could it because those cells at the late stage of dying already (DNA was fragmented and released into interstitial space?)?
Thank you in advance!!
Relevant answer
Answer
Calcein AM is a non-fluorescent cell-permeable derivate of Calcein that is widely used in cell viability measurement. The carboxylic acid groups on Calcein are modified with AM (acetomethoxy) groups, which endows Calcein AM with high hydrophobicity, facilitating its penetration through cell membrane. Once inside the cell, AM groups are hydrolyzed by intracellular esterases. The fluorescent Calcein molecule is restored, which is trapped in the cell and emits strong green fluorescence.
Since dead cells lack esterase activity, only live cells are labeled and detected. The fluorescence intensity will be proportional to esterase activity. Calcein-AM has been proved to be both specific and sensitive for detection and tracking of apoptosis in living cells. The preservation of membrane integrity is one of the most significant features of apoptosis with respect to necrosis. In the presence of membrane defects, Calcein leaks out of the cell and the signal also vanishes in the presence of residual esterase activity.
On the other hand, Propidium iodide (PI) which is a red-fluorescent nuclear stain is not permeant to live cells or cells which are dead but still have an intact membrane (such as the primary apoptotic cells). In late apoptotic and necrotic cells, the integrity of the plasma and nuclear membranes decreases, allowing PI to pass through the membranes, intercalate into nucleic acids, and display red fluorescence.
Calcein generated from esterase in viable cells emits a strong green fluorescence with an excitation and emission maximum at 494nm and 517nm, respectively, while PI once bound to DNA has a maximum emission wavelength at 617nm when excited at 535nm.
There is something that must have gone wrong with your reagent or your process. You may have cells that are either alive or dead, but not both. Cells which are dead but still have an intact membrane (like the primary apoptotic cells), PI is not permeant to these cells.
You may repeat the experiment. Initially, observe the cells in bright field. Then observe the cells in the green fluorescence channel. The live cells will be stained by green, fluorescent Calcein. Follow it by observing the cells in the red fluorescence channel. The dead cells will be stained by the red fluorescent, PI. Then finally you merge the image of green and red channels.
Good Luck!
  • asked a question related to Confocal Microscopy
Question
1 answer
I have primary breast cells embedded in the hydrogel. When stained with EpCAM without the hydrogel, the staining was correctly localized to the membrane as expected! However, after embedding in hydrogel and using the same protocol for immunofluorescent, we are now seeing nuclear staining. Have you encountered this issue? Any recommendations on how to resolve it?
Relevant answer
Answer
Are you sure that this is an artefact? If the antibody epitope is on the intracellular domain it may be genuine signal.
This paper talks about its translocation:
  • asked a question related to Confocal Microscopy
Question
1 answer
Hello dearest EV people!
I want to plate my isolated EVs on coated glass coverslips and image them with confocal microscopy for CDs and other EV markers. I wonder if anyone tried a normal IF staining method on isolated EVs? So far I only saw PEG method published for this...
Thank you in advance!
Relevant answer
Answer
Hey Buse, you could also use dPCR to detect CDs and other markers on the EV surface. I guess that is easier compared to staining, and you will not need too much antibody for it.
You can find the application note for it here: https://www.actome.de/downloads/Actome_EV_Quantification_App_Note.pdf
  • asked a question related to Confocal Microscopy
Question
1 answer
Hello,
Judging by confocal microscopy BLG-nanogels are the most concentrated on interfacial boundary in dispersed system at pH2.5 to 3.5. Therefore BLG-particles are preventing of coalescence and separation of phases. In lower or high higher pH emulsion is separated much quicker. Why in pH 2.5-3.5 they accumulate on IFB ?
Relevant answer
Answer
BLG-nanogels concentrate best at the interfacial boundary of dispersed systems within a pH range of 2.5 to 3.5 because at these pH levels, the BLG proteins carry a positive charge. This positive charge enhances their ability to stick to the negatively charged parts of the interface, helping them effectively prevent the mixture from separating. Outside of this pH range, the charge balance changes, reducing their effectiveness and causing the emulsion to separate more quickly. Essentially, in that ideal pH window, the conditions are just right for BLG to anchor securely at the boundary and stabilize the emulsion.
  • asked a question related to Confocal Microscopy
Question
4 answers
Is there a dye and method that could be effective for staining the mitochondria of isolated insect embryos from eggs for confocal microscopy imaging? We are having trouble finding an appropriate mitochondrial staining method because it is not possible to isolate live embryos from the eggs, and it is also not feasible to perform imaging or measurements while the embryos are inside the egg shell.
Relevant answer
Answer
When staining mitochondria in dead (non-viable) multicellular organisms, traditional dyes like MitoTracker, which depends on membrane potential, are not effective. Here are suitable options for labeling mitochondrial structures in dead cells: MitoTracker Green FM: Stains mitochondria in both live and dead cells, independent of membrane potential. and MitoView Green: Labels mitochondria in both live and fixed cells by binding to mitochondrial proteins.
  • asked a question related to Confocal Microscopy
Question
1 answer
How can confocal microscopy observe cells stained on metal surfaces
Relevant answer
Answer
Epi-fluorescence microscopy (including confocal) will work the same. Just like with cells on glass surface.
Of course the fact that your sample is not transparent will make it harder to find the cells and focus, and the strong reflection from the metal surface might cause issues if the fluorescence filters are of poor quality.
Obviously, you still want to use a coverslip, just like with samples on a glass slide!
  • asked a question related to Confocal Microscopy
Question
5 answers
Dears
I'm going to start working of studying biofilm formation stages in E. coli and Klebsiella pneumoniae and I have to grow and visualise them using Olympus FV1000 CLSM . Can someone suggest me the best stain to use for this purpose, please?
Relevant answer
Answer
May consider using Permai fluorescence dye.
  • asked a question related to Confocal Microscopy
Question
8 answers
Hi
I am new to 3D culture, live cell imaging and confocal microscopy. Is it possible to visualize epithelial and endothelial cells by live cell imaging for a period of time using confocal? In addition, I would like to visualize the bacterial cells and the biofilm produced on the 3D culture over a course of time?
I do have the information on the antibodies to be used for labelling to visualize epithelial & endothelial cells, and biofilm. However, the protocol needs the cells to be fixed. Also, I could get information on the nucleic acid stains which can be used on dead cells to visualize bacteria and eukaryotic cells.
Any thoughts on this?
Thanks in advance
Warm regards
Bindu
Relevant answer
Answer
Thank you Dr. Woo.
  • asked a question related to Confocal Microscopy
Question
3 answers
Hi, I have grown primary nasal cells on semi-permeable trans-well (PET) inserts and would like to prepare a slide (for confocal microscopy). I imagine it has to be fixed and cut out and placed on the glass slide. Does anyone know how to fixate it on the slide without it moving around so its possible to stain it ? 
Your help is much appreciated. 
Thnak you!
Relevant answer
Answer
Hi, I have more to add on this question, how can we prepare such transwell inserts for histology, the ones I am using have an area for 0.3 cm2 ....
  • asked a question related to Confocal Microscopy
Question
7 answers
 I was used 1uM DAPI to stain the nuclear DNA in fixed cells but the efficiency is  very low.
Relevant answer
Answer
May consider using Permai fluorescence dye.
  • asked a question related to Confocal Microscopy
Question
3 answers
I would like to know if it is possible to use antibodies (in this case I would like to use two markers of neutrophils and monocytes such as FITC anti-mouse Ly-6G and FITC anti-mouse Ly-6C) whose application is referred only to cytometry for visualization in confocal microscopy.
Relevant answer
Answer
In general, if antibodies work in tissue (confocal), they should always work in cell suspension (flow cytometry), but not vice versa.
Reasons are below:
1. Epitope availability. Some epitopes on the protein target might be blocked in tissue that cannot be accessed by antibodies using the same epitope. Then you may need to go through the "finding the right clone" process to make it work in tissue.
2. Fluorophore limitation. Many scopes are limited by the choices of laser they have, most don't have UV laser in 350nm, therefore you can't use BUV dye conjugated antibodies. BV dyes can be excited at 405nm but they are large polymer dye that may not be staining complex cell processes and tissue structures as well as small dyes such as Alexa Fluor dyes. Many flow antibodies are designed in these colors and not available in other dyes such as BD antibodies, while many tissue antibodies are limited in colors.
  • asked a question related to Confocal Microscopy
Question
1 answer
Dear all,
I am working with melanoma cryosections from minipigs. My aim is to characterize tumor-infiltrating immune cells between different age groups, mainly by immunohistochemistry. I do multiplex indirect immunofluorescence staining with primary antibody e.g., CD4 and CD8, then incubation with secondary antibody e.g., Alexa 488 and Alexa 555, and of course DNA staining with DAPI. For image acquisition I am using Leica SP5 confocal laser microscopy, then for image analysis I am using imageJ software. My question is how to quantify double positive signals using imageJ? Is there an easy way to do that? I searched in the internet for solutions but they got me very confused. Is there anyone who is experienced and can probably assist me with this please?
Thank you so much in advance!
Relevant answer
Answer
Hello, here's a link to a video that shows how to count co-stained cells with ImageJ https://www.youtube.com/watch?v=Z9-Bb68t6ns If you find it helpful, please subscribe to my channel. Subscribers like you encourage me to grow my YouTube channel. Thanks
  • asked a question related to Confocal Microscopy
Question
3 answers
How do we change the fluorescence intensity of the confocal microscopy to quantitative result by image J?
Relevant answer
Answer
Dear Albrakati,
I would say any image you acquire should store a value in each pixel (eg: from 0-255 for 8-bit images).
In FIJI, you can open these images and select an ROI (region of interest - a rectangle, polygon, or circle where you want to measure).
Then you press M (measure - Menu > Analyze > Measure). This should pop up a Results table in another window.
PS: you can select your desired measurement parameters in Menu > Analyze > Set Measurements...
Cheers from Portugal,
Vítor Yang
  • asked a question related to Confocal Microscopy
Question
10 answers
Greetings all! I am seeking help with a question I recently stuck with.
In the images below, you can see an example of the immunostaining of brain tissue. There is only DAPI and auto-fluorescence from mCherry. I used no green fluorophores. But, surprisingly, I was able to detect weak signals in the green channel that often overlapped with the red ones! I cannot figure out the origin of green signals. The 488 nm laser should not much excite the mCherry according to its spectrum. Even if it does, the bypass filter for the green channel is installed quite far from the emission spectrum of mCherry. According to my knowledge of fluorescent spectra, there should not be any signals in the green channel, especially matched with red signals. But they are. Do anybody have any ideas what's wrong?
I will be very thankful for any help!
There is technical information
Microscope: DragonFly Confocal
EM Gain: 150
Exposure Time/Laser Intensity:
Red-mCherry (40 ms/15%), Green-empty (50 ms/20%), Blue-DAPI (40 ms/15%)
Laser Andor HLE ILE-400 (I am not sure)
Laser for DAPI: 405 nm
Laser for Empty-green: 488 nm
Laser for mCherry: 637 nm
Bandpass Filter Cubes from Nikon with further characteristics
DAPI EX: 361-389 DM: 415 BA: 430-490
FITC EX: 465-495 DM: 505 BA: 512-555
TRITC EX: 540+-25 DM: 565 BA: 605+-5
Links to spectrum
Relevant answer
Answer
The green signal outside the mCherry-expressing cells is most probably from flavins. Its intensity looks a bit higher than I would expect to see considering similar power densities for all fluorescence channels. But we do not know exactly what laser sources were used. If the green laser was an Andor HLE (2 W), then its power density at "50 ms/20%" would be more than 10-fold higher than that from an ILE 637 nm laser.
Concerning the green signal co-localized with the red one from mCherry, I am pretty unsure that it could be attributed to flavins. I'd rather suppose that you observed fluorescence of the mCherry proteins with incomplete chromophore maturation. It is a widespread phenomenon among RFPs (see, for instance, here 10.1007/s43630-021-00060-8) to show some minor green and/or blue populations. Moreover, fixation procedures can likely chemically modify the red chromophore and thus lead to appearance of green fluorescence.
  • asked a question related to Confocal Microscopy
Question
2 answers
Hi all,
I have developed a hydrogel based on silk, and I need to do some fluorescent imaging of the encapsulated cells. When I stain the cells, for example, using CalcinAM, the background is huge and does not allow good-quality images, and I think it is because of the inherent autofluorescence of silk fibroin. Do you have a way to circumvent this?
Thanks
Relevant answer
Answer
Hi Masoud,
I would try with some autofluorescence quencher like True Black or Sudan Black. Applied before doing the staining should reduce the background without affecting the intensity of the target areas.
  • asked a question related to Confocal Microscopy
Question
1 answer
Hello everyone!
I am planning an experiment where I would like to check ubiquitin transfer using confocal microscopy; thus, I intend to add a fluorophore tag to the ubiquitin molecule. I read a few papers and found that people have used mainly GFP tags for in vivo studies or TAMRA and Fluorescein-labeled ubiquitin for in vitro studies.
Since the GFP tag is relatively big there are arguments that it might hamper protein activity.
So, what alternative fluorescent tags are small in size and can be used to study in vivo activity of the protein? Or I would be glad if you could suggest me alternative methods.
Relevant answer
Answer
In your experiment focused on studying ubiquitin transfer using confocal microscopy, it's crucial to choose a fluorescent tag that not only provides reliable visualization but also minimally interferes with the protein's activity. Considering your concern about the size of the GFP tag potentially affecting protein function, here are some alternative small-sized fluorescent tags that you can consider for studying in vivo protein activity:
1. mCherry Tag: The mCherry fluorescent protein is relatively small and has been successfully used in various live-cell imaging studies. Its compact size may help reduce interference with protein activity compared to larger tags.
2. HaloTag: HaloTag is a small protein tag that forms a covalent bond with ligands, allowing for specific and reversible labeling. This tag has been utilized for in vivo protein imaging and tracking.
3. SNAP-Tag: Similar to HaloTag, the SNAP-tag system enables specific and covalent labeling of proteins. Its small size makes it a suitable candidate for minimizing interference with protein function.
4.TurboGFP: TurboGFP is a brighter and more stable variant of GFP. While still larger than some of the other small tags, it offers enhanced fluorescence properties and has been successfully used for protein labeling.
5. mEOS: mEOS is a photoconvertible fluorescent protein that changes color from green to red upon exposure to UV light. This property can help distinguish between newly synthesized and pre-existing protein pools.
6. sfGFP: Superfolder GFP (sfGFP) is a compact GFP variant that folds efficiently even when fused to other proteins. It's small size and robust folding characteristics make it a potentially suitable tag for your in vivo studies.
7. Dendra2: Dendra2 is another photoconvertible fluorescent protein that shifts from green to red fluorescence upon exposure to UV or blue light. This can be useful for tracking protein dynamics over time.
8. Blue Fluorescent Proteins (BFPs): BFPs, such as TagBFP, are smaller than GFP and provide a distinct color for multicolor imaging experiments.
Before finalizing your choice of fluorescent tag, it's recommended to perform pilot experiments to confirm that the tag does not significantly affect the ubiquitin protein's activity and behavior. This can include assessing protein localization, interactions, and function in the presence of the tag.
Alternatively, if you're open to alternative methods, you could consider using techniques like proximity ligation assays (PLA) or Förster resonance energy transfer (FRET) to study protein-protein interactions and ubiquitin transfer without the need for direct fluorescent labeling. These methods can provide valuable insights into protein dynamics while minimizing the impact on protein activity.
  • asked a question related to Confocal Microscopy
Question
6 answers
I want to determine the colocalization of a transmembrane protein along with a membrane marker, could it be possible to obtain the result with fluorescence microscopy alone or it is must to have a confocal microscopy analysis?
Relevant answer
Answer
Dear Manisha,
In my opinion it is not a good idea to check xo-localization with a regular fluorescence microscope.
The staining of your proteins can come from two different focal planes, which means, proteins are not co-localized but look as if they were.
For membrane proteins it should be even more complicated , since you will have fluorescence from the membrane marker all over the cell.
Therefore, use a confocal microscope.
Good luck,
Sebastian
  • asked a question related to Confocal Microscopy
Question
5 answers
Hello,
I'm working on drug delivery in cancer cells. For that, I prepared slides of adherent cancer cells for confocal microscopy. I fixed the cells with 150 ul of 4% paraformaldehyde for 10 min and mounted the cover slip on a glass slide. Then I stored the slides at -20 degC. After one day I did imaging, I found that cells got flattened morphology and some granular structures were seen inside the cells that were totally different from their morphology. Imaging was also not good. I've some doubts regarding this:
1. Whether the incubation time with paraformaldehyde (10 min) was more than required or storage at -20 degC damaged the cells?
2. What should be the optimum time and volume of paraformaldehyde incubation?
3. At what temperature we can store the mounted slides and for how long?
Please guide me regarding this.
Thank you
Relevant answer
Answer
Hello,
I usually incubate my immunofluorescence specimens in a 4% paraformaldehyde solution for 15 minutes and I have never faced any problems following this prodedure (I use about 200 to 250 μl for each slide). I store fixed cells at 4 degrees Celsius in PBS solution containing sodium azide until staining. I've tried keeping them for about 8 weeks and it worked just fine. Just make sure they stay hydrated and sealed to avoid contamination.
Hope this helps,
Best regards,
George
  • asked a question related to Confocal Microscopy
Question
2 answers
Hi.
I have a Lumencor Sola solid state light engine which uses a 5mm liquid light guide (LLG). I would like to use this for a spinning disk confocal setup which uses an FC port for optic fibres. I would like to stay away from lasers for now.
With some primitive calculations based on my objective lenses, I decided to purchase a 400um multimode (MM) optic fibre for UV-VIS (FC-SMA905 plugs). The Sola outputs IR as well, I have decided to either cut out that component with an IR cut filter or disconnect the LED module physically from the board. I do not think heat is good for the fibre.
The problem I am facing now is "squeezing" the output of the Sola into my 400um fibre. Realistically, an efficiency of 20% would be decent.
For the optical scheme, I basically plagiarised Thorlabs' solution for their stabilised light sources, which coincidentally also uses a 400um fibre bundle.
They appear to be using a 40mm best-form lens to collimate the output and an aspherical lens to focus it into the fibre.
I suppose the Lumencor Sola uses a similar method. I will have to open it and check, but I do recall a couple lenses being used, presumably to focus the light into the 5mm LLG. I do not wish to move those lenses and I also do a lot of widefield fluorescence imaging.
Therefore, I suppose I am attempting to collimate the output for a 5mm LLG and then focus it into my 400um MM fibre. I can design and 3D print a bracket for the Sola's output port which will enable a cage system for all the optics.
Another rather unusual method which I am unsure of would be focusing the output light with a microscope objective, straight from the Sola into the MM fibre.
Will my method(s) work? Is there a better method to achieve this with minimal alterations made to the Lumencor Sola?
Thank you for your help and any advice is appreciated!
Relevant answer
Answer
Hello Daniel,
a quick work of advice, squeezing light from a big incoherent source into a small fiber is not only difficult, it is actually physically impossible. This is due to the rule of étendue, which is the optical manifestation of the second principle of thermodynamics.
While if you look up the rule of ètendue you get complex definitions talking about solid angles and apertures, the main, most important consequence of the conservation of étendue is that, given an incoherent source such as an LED or the output of a liquid wave guide, the light intensity per unit of surface can only decrease when passing through a passive optical system. This means that, with your 5mm diameter wave guide, and your 0.4mm diameter multimode fiber, you can at best couple in the fiber a fraction of the light equal to the ratio of the areas, so (0.4^2)/(5^2)=0.6%. You will not be able to couple in anything close to 20%, no matter how complex your optical system is.
Sorry for giving you the bad news, i would suggest looking into multimode lasers for your application, or finding a way to illuminate the disk in your spinning disk without passing through the FC port
  • asked a question related to Confocal Microscopy
Question
2 answers
Hello,
i already have in the lab this live cell stain suitable for flow cytometry (CytoTrack Green 511/525 BIO-RAD) and i was wondering if i could use it to stain live spheroids which will then be analyzed and imaged by confocal microscopy. Has anybody already used this product for applications besides flow cytometry? Thanks in advance for the help
Relevant answer
Answer
Dear Leonardo, I'm also interested in using your tracker! Looking forward to receive news from you
  • asked a question related to Confocal Microscopy
Question
3 answers
May some of you performed staining for immunofluorescence of PBMCs in 96-well plate? As the cells are not going to be cultured, only stained in these plates for confocal microscopy, we are planning to centrifuge (600 g, RT, 6 min) cells in 96 well plate to attach them to the surface of the plate, however I am not sure if this is enough to attach them. I know there are coating plates, but for decreasing costs we are searching alternatives.
Relevant answer
Answer
I think you need to connect with your library or google scholar or approaching the authors will certainly help you
  • asked a question related to Confocal Microscopy
Question
1 answer
Any type of plants will do, but my plant species of intererst is Arabidopsis.
Relevant answer
Answer
Here's a simple protocol you can try:
  1. Harvest your Arabidopsis plants and gently remove them from the soil.
  2. Cut off the above-ground portion of the plant and discard it. You only need the roots for this protocol.
  3. Rinse the roots gently with tap water to remove any loose dirt or debris.
  4. Submerge the roots in 70% ethanol for 30 seconds to sterilize them and kill any surface bacteria or fungi.
  5. Rinse the roots three times in sterile water to remove any residual ethanol.
  6. Place the roots in a 1:1 mixture of 30% hydrogen peroxide and concentrated sulfuric acid for 10-15 minutes. This will remove any remaining tissue from the roots.
  7. Rinse the roots three times in sterile water to remove any residual hydrogen peroxide and sulfuric acid.
  8. Use a fine brush to gently clean any remaining debris from the roots.
  9. Transfer the roots to a glass slide with a small amount of water, and gently press them flat with a coverslip.
  • asked a question related to Confocal Microscopy
Question
1 answer
I am using Glycerol 50% as my mounting media and not getting the required clarity. I have three tagged fluorophores. What can I use as readily available antifade reagent or how can I modify the mounting media to improve the clarity of my images.
Relevant answer
Answer
Probably You should try the Prolong Antifade reagent from Thermo (Link Attached). in my experience I have observed that it gives better signal than the Vectashield, Fisher and Sigma's.
  • asked a question related to Confocal Microscopy
Question
2 answers
I use this chambered culture well plates for immunofluorescent confocal microscopy: https://us.vwr.com/store/product/12361327/culturewelltm-removable-chambered-coverglass-electron-microscopy-sciences
I'm having issues removing the rubber gasket without breaking the attached slide. The slide is very fragile no matter what method I use to remove the gasket. I end up cracking the whole slide and rendering my sample useless.
Does anyone have any suggestions of a technique or a better brand of slides? I need ones that are for small infection volumes (30 ul).
Relevant answer
Answer
I have had some successs treating it like a protein gel asset. I use a small spatula get in between the seal and then slowly twist to crack the well from the slide. You may need to do that in a few spots before fully detaching the wells. This has worked most of the time, but I have had a few break on me if I use too much force. I hope that helps.
  • asked a question related to Confocal Microscopy
Question
1 answer
Recently, I established a mutant strain of Drosophila with an APEX2 tag in its genome. Using this strain, I have successfully performed normal immunoelectron microscopy. However, it is difficult to detect the weak signal (it can be observed by confocal microscopy). Therefore, I would like to try the APEX2-Gold method. what points should I pay attention to when performing the APEX2-Gold method on animals? Also, can I keep the enhancement solution in stock? I would appreciate any tips anyone can give me.
Relevant answer
Answer
Hello,
When performing the APEX2-Gold method on animals, there are a few things to consider:
  1. Make sure that your sample is properly fixed and dehydrated before staining. This will ensure that the APEX2 antibody can bind to the APEX2 tag in your mutant strain.
  2. Use a high-resolution electron microscope, such as a transmission electron microscope (TEM), to visualize the gold particles. This will allow you to see the fine details of the APEX2-tagged proteins in your sample.
  3. Use a high-quality APEX2 antibody and gold conjugate. This will ensure that the signal is strong and specific to the APEX2 tag in your mutant strain.
  4. The enhancement solution is usually not stable for long periods of time, it is best to prepare fresh for each experiment.
  5. Optimize the conditions for the staining by testing different concentrations of the reagents and incubation times.
Overall, the APEX2-Gold method is a powerful tool for detecting and analyzing APEX2-tagged proteins in electron microscopy. With proper technique and high-quality reagents, you should be able to detect a strong signal in your samples.
Regards/
  • asked a question related to Confocal Microscopy
Question
4 answers
Hello,
Could you please recommend DAPI concentration for nuclei staining in mesenchymal stem cells during chondrogenic differentiation?
I have used 1 μg/ml DAPI for 6 min, then as usual PBS washing. but I got fuzzy colour and lots of unspecific binding (but still fuzzy).
If that matters - the brief protocol:
- PBS washing 3x
- permeabilization 0.2% Triton X-100/PBS, 4 min, PBS washing 3x
- blocking 0,2% BSA/PBS 1h, PBS washing 3x
- staining of collagen I, II and X (each with a pair of antibodies, with 3x washing in PBS in between steps).
Thank you!
Relevant answer
Answer
Hi, i think you can use a concentration of 2ug/mL.
  • asked a question related to Confocal Microscopy
Question
2 answers
I am overexpressing an endogeneous protein with c terminal myc tag in entamoeba. Overexpression has been confirmed by real time pcr. But when i am trying to visualize it in confocal microscopy with anti myc antibody i cannot see any staining. Primary antibody added is in ratio 1:100 and secondary antibody ratio is 1:300. Please suggest that what may be the problem.
Relevant answer
Answer
I would try confirming the expression by western blot using both myc antibody and antibody for the protein of your interest. The existence of mRNA does not mean that protein was expressed.
Myc tags are not ideal for microscopy because endogenous c-Myc could also be detected and could interfere with the analysis but it usually doesn't.
  • asked a question related to Confocal Microscopy
Question
3 answers
Hello, fellow researchers! I am trying to develop a project to track the growth and development of embryonic and larval killifish (F. grandis and F. heteroclitus) in varying environmental conditions. Confocal or fluorescent microscopy are methods I am thinking of using, but I am unfamiliar with either of these techniques. I am hoping someone with more experience could help give me the pros/cons of both or provide resources to previous research using either technique so I can familiarize myself with what is possible and the methodology. Thank you for any help you can provide!
  • asked a question related to Confocal Microscopy
Question
5 answers
In my imaging of mouse brains, there are fluorescent dots that appear in the image from confocal microscopy which are not any of the target cells in my experiment. I was wondering if using a secondary antibody from a donkey and a normal donkey serum in the blocking could potentially cause this?
Relevant answer
Answer
You should avoid using blocking serum from the same host species as your primary antibody that is detected by a secondary antibody against the same species (immunoglobulins in the blocking serum will compete for the secondary binding).
I agree with Yulia Panina. Washing is an important step. Washing after each application of antibody or other fluorescent probe eliminates antibodies with lower binding affinity present in the sample and thus reduces non-specific signal, or cross-reactivity. Washing for a few minutes in PBS with at least two buffer exchanges will help to eliminate unbound and loosely bound antibody from the sample.
If you are using donkey secondary, then you should use donkey serum for blocking which you have already been doing. Instead of donkey serum you may also try using using BSA (3-5%) for blocking.
Best.
  • asked a question related to Confocal Microscopy
Question
2 answers
Dear Fellow Researchers,
We are analyzing adaptive immune response in bladder cancer tissues using immunofluorescene stain and confocal microscopy.
Setting pinhole at AU=1 (perfectly confocal) results is very low signal. Increasing it allows to collect more signal but may yield some over-emission.
What pinhole is acceptable for tissue sections?
Relevant answer
Answer
Increasing the size of pinhole will definitely increase the background. I would suggest to keep the pinhole at 1 or may be increasing the pinhole size to 1.5, but in addition, I would definitely suggest to take multiple z-stacks to get a composite image with bigger signal and low background. Hope this helps. Good luck.
  • asked a question related to Confocal Microscopy
Question
3 answers
Hi all,
I am growing some large (1mm) organoids in hydrogel. I am planning to stain the organoids for 3 markers + nuclei and image them by confocal covering all 4 channels of UV, green, red and far red. I would like to use the UV channel for staining the nuclei. I am using a clearing agent (RapiClear) clearing the tissue before the imaging in order to be able to see deep inside the structures. The problem is that when I use DAPI, after clearing, the DAPI seems to fade and is not detectable anymore by confocal. Does anybody have experience with SYTOX blue from Thermofisher for staining the nuclei for confocal? Is it stable after clearing the tissue?
Thanks a lot in advance!
Relevant answer
Answer
Thanks Both Jiasong and Ayse for your answers!
  • asked a question related to Confocal Microscopy
Question
12 answers
Hi everyone.
I am facing a very intriguing phenomenon. My immunofluorescence and confocal microscopy analyses showed a nucleus that is positive for MnSOD (which should be present only within mitochondria). Please, find attached representative images of cells stained for either nucleus or MnSOD positivity, along with the merged image.
Of note, it should be highlighted that the anti-SOD2 primary antiobody we used is extremely specific and that Western immunoblots showed only the specific band.
Did anyone else find MnSOD (SOD2) in the nucleus in some experimental or physiological condition?
Thanks a lot in advance.
Stefano Falone
Relevant answer
  • asked a question related to Confocal Microscopy
Question
2 answers
Hello everyone!
Looking for some useful tips for fluorscence/confocal microscopy of plant root tissue.
I am mostly working with the root tissue of Arabidopsis thaliana where I take confocal/fluorscence images of plant root samples expressing proteins of interest with GFP/mcherry tags also often with Propidium Iodide/DAPI stains. Usually, I don't fix the tissue and take images as such and here are a few problems I face most of the time and I will be glad if you can advise me some suggestions to fix or minimize the following issues-
1.The amount of protein expression in various layers of root tissue is variable ranging from low to very high expression so in a Z-stack often I get highly staurated layers or if I adjust laser power/analog gain to minimize that oversaturation I end up losing signal where protein is less expressing. What should I do in such case?
2. how do you prepare live tissue for imaging, like if you use any specific buffer that does not change the physiological state of the plant tissue but helps in maintianing the GFP fluorescence for a longer duration?
3. Also, no matter how carefully I prepare the slide I never get the root tissue lying uniformly over the slide which means I get regions with good focus and in perfect plane wherease at some regions it gets blurry, out of the focus, how to avoid it?
4. I too have problem with DAPI staining of the root tissue as in most cases barely I see any nuclei but most often I end up with images showing DAPI at the cell borders just like Propidium Iodide.
Thanks a lot in advance for all the incoming beautiful suggestions.
Relevant answer
I work mainly with mammalian cells and have no clue about the plant cells but I feel this could help you with your DAPI problem. DAPI is not great at staining a live nucleus. DAPI most often cannot enter cell membrane/wall and staining requires fixation and permeabilization. I suggest you try Hoechst 33342 which has similar excitation and emission wavelength so you don't have to change your protocol.
Please note that this works for mammalian cells and may or may not work for plant cells but something to think about.
  • asked a question related to Confocal Microscopy
Question
4 answers
Dear all,
I am using confocal microscopy to visualise membrane proteins of oleosomes (in any case proteins surrounding the oleosome) extracted from rapeseed.
The oleosomes have been stained with Nile red to detect the oil, and Fast Green FCF (0.013%) to detect the proteins.
The core oil is easily visualised but I am struggling to clearly visualise the proteins which would surround the oil droplet.
Do you have any advice on how to improve the visualisation? It may depend on the microscope settings I guess.
Thank you very much to anybody who could help me
Filippo
Relevant answer
Answer
Thank you Yulia Panina, I did more tests and the results are better (no tail and green ring around the red droplet), thus it seems to be matter of sample preparation.
Probably if pH is too high, the FCF dye loses its property.
Many thanks for your interest.
Best wishes,
Filippo
  • asked a question related to Confocal Microscopy
Question
4 answers
Hello everybody,
I am doing immunocytochemistry staining for splenic B cells from WT and Knockout mice for one protein. By FACS analysis, it's clear that the signal for the knockout protein is nearly absent in Knockout cells, but when I do confocal microscopy experiment for the same cells, the signal seems to be the same in knockout as in WT. Does anyone have an explanation or can recommend what to do?
P.S: I use the same primary antibody for IF and ICC and No background for the secondary antibody in the control.
Thank you!
Relevant answer
Answer
Thank you Alibek, I will do it and I hope it will work :)
  • asked a question related to Confocal Microscopy
Question
1 answer
Hi Everyone!
I would like to test by immunostaining that a specific membrane protein is present on my bacteria's outer membrane.
I cannot use FACS for this purpose, so I thought of confocal microscopy as an alternative. I would just fix the cells to keep their membrane intact.
I have not found yet any methods for this. Could you recommend one?
If you have any insight please share it with me!
Relevant answer
Answer
Yes. Here is the protocol that we used for detection of proteins on the surface of Lactococcus lactis bacterial cells.
Immunostaining of bacteria for fluorescence microscopy
20 μl of cell culture in stationary phase (OD600=3) was added to 500 μl of Tris-buffered saline (TBS; 50 mM Tris-HCl, 150 mM NaCl, pH 7.5) and centrifuged for 5 min at 5,000 × g and 4°C. The pellet was resuspended in 500 μl of TBS, and 1 μl of FITC-conjugated human IgG antibody (Jackson ImmunoResearch, West Grove, PA) was added. After 2 h of incubation at RT with constant shaking at 550 rpm, cells were washed three times with 200 μl 0.1% TBST and resuspended in 300 μl TBS. Stained cells were fixed to a microscope slide by use of a StatSpine Cytofuge 2 centrifuge (Iris Sample Processing) for 10 min at maximum speed. An LSM 710 confocal microscope (Carl Zeiss, Oberkochen, Germany) was used for observation of prepared specimens. Alexa Fluor 488 was excited with an argon laser (488 nm), and the emission was filtered with a narrow-band 505- to 530-nm filter. All images were taken with the same settings and were analyzed using Carl Zeiss ZEN Lite 2012 software.
  • asked a question related to Confocal Microscopy
Question
2 answers
For my project, I need to find colocalization of my retrograde tracer (tiny red retrobeads) with dopamine neurons in (TH immunolabeling, alexa fluor 488).
My problem is that retrobeads are so tiny and I only have few dots of them inside cell. Also finding very sparse red beads in red autofluorescence background in VTA is really difficult under confocal microscope. Does anyone know how can I solve the autofluorescence issue or potentiate my tiny red dots to be distinguishable in red background?
I fix my tissue in PFA 4% and thickness of my slices is 50um.
Relevant answer
Answer
Hello,
once you do image treatment, you can define a background treshold on the read signal you find as background. You can also do a Z-stack to enhance your bead signal and also have bead signal in another stack that you might be missing. You can merge your stacks to have the maximum signal. If not, you can also do FACS and seek for double tagged populations ^^
  • asked a question related to Confocal Microscopy
Question
4 answers
I'm using JC-1 to measure the mitochondrial membrane potential by laser confocal microscopy. I used 100uM of FCCP as a positive control. Surprisingly, it caused lower green to red ratio and was significantly lower than the untreated cells. My cells are cryopreserved mice oocyte. On the other hand, FCCP did work and caused an elevation in calcium using Fluo 3 AM and caused plasmalemmal membranes depolarization.
Relevant answer
Answer
Dear Omar,
do you got the answer for this question? I've meet the same situation and tried to find the explanation.
Thanks.
  • asked a question related to Confocal Microscopy
Question
3 answers
Hello.
I need to mesuare the change in quantity and size of lysosomes in different experimental condition and I have this fluorescent dye that stains acidic compartments (LysoTracker Green DND-26) in live cells but It´s used for confocal microscopy So I need to know if someone has ever used in flow citometry with good results and if that´s the case: Could someone give me a protocol ?
Thank you
Relevant answer
Hi Diego Suarez have you been using this probe at the end?
  • asked a question related to Confocal Microscopy
Question
1 answer
I was thinking about VGlut2 (presynaptic) and PSD95 (post-synaptic) for the excitatory synapses, and VGAT (presynaptic) and Gephrin (post-synaptic) for the inhibitory ones.
Should I define as a synapse only the areas of colocalization between pre and post synaptic marker? Or should I consider also the isolate VGlut2/VGAT and PSD95/Gephrin ?
Relevant answer
Answer
I think that you can use the Golgi staining to measure the density
  • asked a question related to Confocal Microscopy
Question
6 answers
Dyes for visualizing bacteria
Relevant answer
Answer
Alexa Fluor 647-coupled antibody, Alexa Fluor 488, Alexa Fluor 500, and Alexa Fluor 514 may might be helpful.
Thankyou
  • asked a question related to Confocal Microscopy
Question
4 answers
I am trying to analyse the state of the gut epithelium by analysing by confocal microscopy the state of some adhesion proteins (to note, Claudin and occluding); however, these proteins are not exclusive of epithelial cells, which is why I would like to add a marker specific for this subset. Could you please recommend me any marker/combination of markers that makes easier the identification of epithelial cells? Thank you in advance
Relevant answer
Answer
Hi Maria,
I agree with other people that EPCAM and E-cadherin are good markers. The staining will be found at the lateral membranes of cells (E-cadherin has strong signal at adherens junctions and weak signal at lateral membrane). ZO-1 is also a good marker for tight junctions although it also stains blood vessels and mesothelium (You can distinguish them by morphology).
When you optimize your protocol, I would reccomend to try different fixation method. Most antibodies of tight junction proteins give great staining with ethanol (or methanol) fixation. Acetone treatment following ethanol fixation sometimes improves S/N ratio.
Good luck!
  • asked a question related to Confocal Microscopy
Question
3 answers
I made a script to try to convert 4 colour nd2 files (not stacks) to run in the process> batch function of Fiji (see https://www.youtube.com/watch?v=fU104OU3Kk4 at 3:28) to create single tiffs, of all colours, merged. When I check the results in Fiji I get only the DAPI channel from the script below. However, if I run the macro in Fiji itself without the batch command, just on a single open nd2 file, I am left with the file that I want! So I am not sure why it is only saving the DAPI. Does the batch command save the earliest tiff that it sees and not let the macro finish? I am an inexperienced Fiji user so I am not sure what I am doing wrong.
Code is:
title = getTitle();
run("Split Channels");
one = "C1-" + title;
two = "C2-" + title;
three = "C3-" + title;
four = "C4-" + title;
run("Merge Channels...", "c1=["+one+"] c2=["+two+"] c3=["+three+"] c4=["+four+"] create");
run("RGB Color");
selectWindow(title)
close()
Relevant answer
Answer
I Just checked your macro and it looks okay to me. Maybe you should try and include in the last step
run("Flatten");
I sometimes experience errors with merged images in TIFF format and this seems to help
  • asked a question related to Confocal Microscopy
Question
4 answers
We would like to use an enzyme tagged with eGFP and eGFP-tagged alpha synuclein at the same time to observe them under confocal microscopy. However, this will give wrong readings, so we would like to replace eGFP with mCherry. There is a research used the same way but without a detailed explanation.
I will add the link of this research below:
Dinter E, Saridaki T, Nippold M, Plum S, Diederichs L, Komnig D, Fensky L, May C, Marcus K, Voigt A, Schulz JB, Falkenburger BH. Rab7 induces clearance of α-synuclein aggregates. J Neurochem. 2016 Sep;138(5):758-74. doi: 10.1111/jnc.13712. Epub 2016 Aug 4. PMID: 27333324.
thanks in advance.
Relevant answer
Answer
As has been mentioned, a simple sub-cloning should do the trick. The caveat being that you have some DNA carrying the coding sequence of mCherry.
Cloning options:
1. Restriction enzyme-based cloning
2. Restriction-free cloning - rf-cloning.org (this works great and is often cheaper, especially if you don't want to order restriction enzymes)
3. Gibson cloning
Happy cloning!
  • asked a question related to Confocal Microscopy
Question
8 answers
Hi,
I want to quantify the cell area of bEnd.3 cells (mouse brain endothelial cells) using confocal microscopy by ImageJ. Most of the available protocols need to ignore neighbouring cells where cell-cell boundaries are in touch with each other. However, endothelial cells show continuous morphology. I will be really grateful if someone can help me with the protocol for cell area calculations using ImageJ.
I also want to quantify the distribution of tight junction proteins in cytoplasm vs. plasma membrane of these cells. If there is any way by which this quantification is possible using ImageJ then please help me.
Any help is appreciated.
Thank you in advance.
Relevant answer
If you visually cannot distinguish single&full cells, even with lower magnification, then I am afraid that you need additional staining that evenly stains the membrane to quantify cell surfaces (as I have mentioned above) so the boundaries between cells are clear. Also, nuclei staining would also help to distinguish single cells. PS: General rule: one can only quantify things that are visible.
Regarding the delineation of membrane and cytoplasm compartments - same rule. You can only measure/delineate if you see it, so I am afraid that lower magnification than what you showed above, would not allow that. Also, as already mentioned you have no marker for the membrane, so the delineation will not be very accurate. You would need additional staining of the membrane on top of your protein of interest to be accurate.
Furthermore, is there a way to seed the cells at lower density? This could greatly help your analysis as maybe then you can easier distinguish single cells.
Good luck!
  • asked a question related to Confocal Microscopy
Question
1 answer
Can we use deionized water instead of mounting medium for confocal microscopy to preventing photobleaching? Since all the parts of the tissue does not mix well with thick mounting medium and therefore micro-air bubble develops.
Relevant answer
Answer
Mounting media usually contain anti-fading agents, so they help prevent photobleaching. If it is too thick, you could dilute it with some PBS. Of course you could also use plain PBS, if you are in a hurry and you fo not care about keeping the sample in good condition for later. Ideally you should put the medium on the one side, place the coverlip at a 45 degree angle, then slowly lower it so that the medium goes everywhere on the tissue with as few bubbles as possible.
  • asked a question related to Confocal Microscopy
Question
7 answers
We are going to upgrade our NIKON A1 confocal system with the incubator for long lasting live cell imaging - the incubator should contain atmosfere humidifier, CO2, temperature control etc. Please share your opinions on what system do you consider best and why? What to avoid ? What may be beneficial? The microscope will be used not only for live cell imaging so the system should be reconfigurable . We are looking for something from the top shelf but easy to maintain sterile environment and user friendly.
Relevant answer
Answer
I would say that the question doesn't stand Okolab vs. Tokai Hit. There are many solutions from other companies as well.
Recently I wrote a blog post for FocalPlane about temperature control in light microscopy. I think it could be useful for some people reading this thread.
disclosure: I work for a company that produces a temperature control device. It's called VAHEAT and it could be the device for you, if you do a lot of high resolution microscopy, need high temperature precision, need temperature cycles, not just a constant temperature, use microfluidics and more. interherence.com/vaheat/
  • asked a question related to Confocal Microscopy
Question
1 answer
I am doing a mammalian oocytes maturation, followed by nuclear meiotic evaluation using confocal microscopy, I mount the oocyte over 90% glycerol in a double sided adhesive tape, and I am curious because I don't want to smash the oocytes under the cover slip. is there any recommendation for a good commercial wax cushion to be used in this case. (oocyte diameter is 100 micron).
Thank you,
Relevant answer
Answer
Yes. It is matter of concern and although alternative methods are available in order to prevent oocyte damage during handling in vitro. The alternative methods have some or other type of negative impact on the oocyte. The development of an improved method/materials that can replace glycerol and prevent damage under experimental condition is required. To the best of my knowledge no such appropriate wax cushion is in the market which can be better in handling oocytes in vitro or for the assessment of maturation process in mammalian oocytes for confocal microscope.
  • asked a question related to Confocal Microscopy
Question
2 answers
Hi,
I transfected HEK cells with fluorescent ABCG2 transporter (multidrug resistance transporter) , it has a GFP probe attached, and I am performing a live cell confocal microscopy experiment, where I would like to determine the kinetics of Mitoxantrone (chemotherapeutic, far red fluorescence) accumulation in the plasma membrane of the transfected GFP-positive cells. I am doing it by measuring far red fluorescence over time. For the information, mitoxantrone is the substrate of this transporter and ABCG2 is pumping it out of the cell, but when I apply an inhibitor to it, the accumulation rate increases, proving that the inhibitor works. That is how I am testing some of the compounds designed to be potential ABCG2 inhibitors.
Now that I hopefully explained the idea, I can get to the quantification part. I am confused about how many cells/fields of view to use to have a considerable number of sample for quantification. How many transfected cells should I take picture of? 50, 100? Is it for example 100 cells in 3 different experiments, or it is 100 cells per experiment, 3 times ?
As I have to take pictures in certain time point, I can only have one field of interest, where I usually have around 15 transfected cells. How can I collect 100 of them?
Or should I use regions of interest, few of them per one cell, around the membrane and measure that? I am really lost, so please those with similar experience, help!!!
Kind regards,
Marija
Relevant answer
Answer
Hi Marija,
People tend to do one of two things when determining sample size. You can either refer to a number of replicates that are „conventionally accepted” in your field for a particular experiment, or alternatively use statistical calculations to determine an ideal sample size based on certain assumptions/preliminary data. In the absence of standard protocols, you could go down this second route. For this you’ll need to have some idea of what an „average” set of results from your experiment look like. The number of replicates and fields of view per replicate will depend on:
  • the variability between your cells/regions within an experiment
  • the variability between independent experiments
  • the effect size (ie how big of a difference) you want to detect.
I would recommend you have a brief read (or watch this YouTube video https://www.youtube.com/watch?v=clbg10IwzYA) about power / sample size calculations so you get the theory, and then try an online calculator such as the one found here: https://www.stat.ubc.ca/~rollin/stats/ssize/n2.html
For the first point, the number of fields of views, the main question is whether your 15 cells you currently image accurately represent the entire well. You would need multiple fields if for example you notice that within a single well there are regional differences. If Mitoxantrone intensity is fairly evenly distributed I would not worry too much. If you are concerned, I was wondering if you could extend your field of view. If you quantify average intensity in the whole cytoplasm, you could probably get away with using a lower mag objective to capture a larger field of view and higher number of cells.
Regarding the variability between experiments, if you have already performed it a few times you should have an idea what the standard deviation is and you could plug that into the sample size calculator.
  • asked a question related to Confocal Microscopy
Question
5 answers
Using confocal microscopy imaging technique is it possible to calculate the percent apoptotic index? In some research papers it is mentioned but up to what extent it is reliable and is it necessary to validate using FACS analysis?
Relevant answer
Answer
You have to count manually this way, which you may want to do on a regular fluorescent scope first then use confocal to collect your publication quality images. You will quantify this by calculating the percentage of Annexin-V positive and PI-negative cells divided by the total number of cells. Flow is calculating by sorting and is not necessary. Also, something to think about, are you trying to differentiate between apoptosis or secondary necrosis?
  • asked a question related to Confocal Microscopy
Question
3 answers
Context:
  • Isolated red blood cells (protocol in .docx is attached)
  • Did confocal microscopy (stained RBCs with CellMask Red (photoiodide))
  • Observed red blood cells (circled in blue) and also some cells with spiky surfaces (circled in green) (image as attached)
Questions:
  • What are those spiky cells?
  • Is it normal for my red blood cell extracts to have them?
  • If not, did something go wrong in my sample preparation, and how do I get rid of them?
Thank you!!
Relevant answer
Answer
The simplest explation is that your sample is drying out. As the buffer becomes more hypertonic RBCs undergo crenation. https://www.thoughtco.com/crenation-definition-and-example-609188
  • asked a question related to Confocal Microscopy
Question
2 answers
If there are two different Nps prepared for two different drugs encapsulated in a single polymeric system(co-delivery) then can the uptake of both the NPs detected using confocal microscopy? In this case two types of fluorescent dyes needed ??
In another case, for cellular localization expt which dyes except Lysotracker can be used?
Relevant answer
Answer
Dear Chittaranjan Behera , the simple and general answer to your first question should be clearly negative, the cellular uptake and cellular location of NPs are different experiments. However, they could be the same experiment if all the uptook NP´s are localized at the same organelle or cell region.
With respect to your second question, if you have two different drugs encapsulated in a polymeric delivery system to facilitate the uptake by the cell, they could be detected by confocal microscopy if these two drugs show distinctive fluorescence signals. It means that these two drugs must show auto-fluorescence in the conditions of excitation used and they must show different "colours" to be distinguished. If one or both drugs do not show fluorescence under the working conditions or they show the same or very close colours, then you must use some dye to label each drug. If your delivery system is intended for reaching some cell´s organelle and just there delivering its cargo, may be it could be possible to just follow the whole system (drug A + drug B + polymeric system) and labelling it with the addition of some fluorescent dye, well inside or attached to the polymeric shell. In this way you could see if the whole system reaches the cell and then the target organelle. To use this methodology you need to be sure that both drugs reach the target and that the polymeric shell keeps its cargo till the destination, in other case you would be following an empty- fluorescent- shell.
Hope this helps. Good luck with your research work.
  • asked a question related to Confocal Microscopy
Question
3 answers
how to detect and counted early apoptotic, late apoptotic and necrotic cell population using confocal microscopy?
Is flow cytometry is mandatory to count the population or without using flow cytometry the cell polulations can be counted using confocal microscopy?
For image analysis, confocal microscopy is best of course.
Relevant answer
Answer
In apoptotic cells, the membrane phospholipid phosphatidylserine (PS) is translocated from the inner to the outer leaflet of the plasma membrane, thereby exposing PS to the external cellular environment. Annexin V is a Ca2+ dependent phospholipid-binding protein that has a high affinity for PS. Since annexin V staining precedes the loss of membrane integrity which accompanies the later stages of cell death resulting from either apoptotic or necrotic processes, staining with annexin V-FITC is typically used in conjunction with a dye such as PI allows to identify early apoptotic cells (PI negative, annexin V-FITC positive) from dead cells (PI positive, annexin V-FITC positive). you can see this picture for easy understanding- https://www.creative-bioarray.com/support/annexin-v-apoptosis-assay.htm
If you want to distinguish late apoptotic cells from necrotic ones you have to use flow cytometry. You can also check this for reference
  • asked a question related to Confocal Microscopy
Question
1 answer
I am interested in studying biofilm formation as a function of multiple variables, and would like to go beyond spectrophotometric/colorimetric/microplate readings and to analyze biofilm structures microscopically as well. Unfortunately, fluorescent microscopy (FM), confocal laser scanning microscopy (CLSM) and scanning electron microscope (SEM), tools that are routinely used by well-funded research groups for this purpose, are currently not at my disposal.
To this end, I am considering to utilize traditional staining (e.g., crystal violet) of these structures followed by simple light microscopy. May I ask for recommendations on objectively/quantitatively analyzing the images that can be generated this way (perhaps through a specific software)? What I have found so far are all for FM, CLSM and SEM images. Thank you!
Relevant answer
Answer
Dear Aedrian,
We have faced your problem in the past too - but if you have a good microscope and camera system I think that you can usefully and quantitatively examine differences in biofilm structure using some common dyes for bacterial cells and EPS.
The key questions are whether you have a fluorescent microscope and camera system. If you do, then I'd suggest looking at using live/dead stain mixes, or propidium iodide, and whichever fluorescent DNA and protein dyes you can get. If you only have a light microscope, then you need to be using standard coloured dyes, again for as many cellular components as you can. The second question is whether your camera system can capture single (black and white) or multiple colour images.
No matter what type of microscope and camera system you have, you should be able to take good images of biofilms at a resolution from 10x - 40x and perhaps even 100x (at which point bacterial cells should be visible). You can import these image files into free software such as ImageJ - you can then divide the image into squares and get the total absorbance (e.g. for live/dead stain) and use this in your statistical analysis.
You could look at the variation across the image and correlations between different stains to investigate heterogeneity. You can compare mean values for each image (or measure the absorbance for the entire image) to then compare across images (e.g.different incubation conditions, different strains, etc.).
Regards, Andrew
  • asked a question related to Confocal Microscopy
Question
3 answers
I want to perform cellular uptake and localization study of my siRNA-loaded nanoparticles. For performing both the experiments which fluorophore-labeled siRNA, I should order.
Kindly suggest.
Relevant answer
Answer
Thank you so much.
  • asked a question related to Confocal Microscopy
Question
10 answers
Hi, I am currently working on the colocalisation of 2 proteins and I plan to work it out with confocal microscopy.
In the school I am working we have the leica LasX software with the co-localisation license but it is only available in that one computer connecting with the confocal mic itself. As there are always booking on that confocal mic and the computer is always in-use. I am looking for other softwares for the analysis of co-localisation. Best if I could draw an ROI, shows the pearson's and manders' correlation.
Currently I know ImageJ plug-in JACoP. Any other recommendations? And what's the difference with using the leica LasX co-localization analysis with other softwares such as Image J?
Is there any requirements on the software I use for co-localisation analysis if I am planning to publish my data?
Thanks.
Relevant answer
Answer
Hi there,
The main difference of using freeware vs built-in colocalization tools in expensive software is the price :)
Basically, these software incorporate algorithms to express colocalization and pack them into an user-friendly interface, but they're in no way superior to any other (free) software or plugin.
Additionally, you can gain insight into what these software are doing, so you will be able to know which analysis suits better your experiment by using freeware and reading carefully the guides and the manuals (Thresholds, background subtraction, ROIs, and so on...)
I have always used ICA with pretty good results (it operates in a similar manner to JACoP) and you can define ROIs in it. However there are many others. Check the attached documents and links below for practical working examples and explanations. The two attached documents are for ICA, but they provide good working examples and differences for the coefficients
Overall interesting review:
JACop Plugin original paper:
ICA Pugin original paper:
ImageJ forum about differneces in JACoP and ICA:
Why to choose one or another coefficient?
Manders' Original paper:
Guide to ICA:
Another free plugin:
And, for the second question:
"Is there any requirements on the software I use for co-localisation analysis if I am planning to publish my data?"
No, the requirement is not for the software, but for the explanation/justification and interpretation that you give to the analysis and results. Usually, all the Plugins incorporated in ImageJ have been published if that's what you mean
Why you have chosen this analysis? Have you corrected the background? Are you using ROIs? Is this analysis powerful enough? Why you provide one coefficient and not another?
In this case, understanding what you are doing and using proper controls should be enough to validate the analysis tools that you are using for colocalization.
As positive controls, using two secondary Abs against the same 1ary Ab is usually a really good positive control (if you have time and can spend Ab's doing so). You can also perform randomization to see how accurate your coefficients are, but all these things are explained in the attached documents,
Cheers,
J
  • asked a question related to Confocal Microscopy
Question
6 answers
Does anyone have any experience/reviews of the EVOS XL Core microscope? 
Relevant answer
Answer
Does anyone know how to insert a scale bar on an image using evos xl core? This option is not shown to me on the screen. I’m not sure if it’s not updated or if I have to set this somewhere.
Any ideas?
  • asked a question related to Confocal Microscopy
Question
5 answers
(rather than an imaging plate with black wells). Would this work? Or would there be too much light scattering?
Thanks
Relevant answer
Answer
If you were to image in an inverted cconfocal microscope, you would need a proper holder that can hold a 96 well plate (depending on what magnification you want to image at). However, you can only image one well at a time. Hence, I would recommend (and you would get much better quality images on) using MatTek dishes that have a glass bottom cover slip.
  • asked a question related to Confocal Microscopy
Question
5 answers
I would like to analyze a 1mm in thickness skin sample using confocal microscopy. The sample is too thick to be placed between a slide and coverslip then sealed by nail polish. The idea is to do z-staking and I don't want to cut the sample. Is there is an alternative way to tackle this issue? For example, can I mount the sample using some sort of adhesive tape?
Thanks
Relevant answer
Answer
You can add ‘spacers’ as mounts to raise up the coverslip. Use a diamond scribe glass cutter to cut slices of coverslip. Mount these on your slide using nail polish or glycerol in parallel so your coverslip will rest on top of them with your sample in between in mounting media. Then seal round the edges with glycerol gelatine.
  • asked a question related to Confocal Microscopy
Question
6 answers
I am working on amplifying a signal for confocal imaging using the Labelled Streptavidin/Avidin Biotin (LSAB) technique and ran into a surprising outcome: No staining. This is odd as I've employed this stain previously using the traditional indirect method and see decent specific signaling so I thought by amplifying the signal I'd get better, not worse, staining. Below I'll leave the basic outline as to my design (barring washes in between and counterstains).
  1. Incubate mouse anti-antigen primary antibody at 4 degree overnight (1:100; used at same concentration as in indirect method staining)
  2. Incubate biotinylated goat anti-mouse secondary at RT for 90 mins (1:150)
  3. Incubate Cy3-conjugated streptavidin for 90 mins (1:1000)
Any tips as to how to optimize this protocol and get it to function would be greatly appreciated!
Relevant answer
Answer
I was actually going to ask you if you had double checked your filter/Cy3 combination, but since you were already using Cy3 successfully I figured you were good to go. You might throw some of that SA-Cy3 dilution between a coverslip/slide and just make sure you can see it with your 514 set up.
I wish I was being more helpful!
  • asked a question related to Confocal Microscopy
Question
5 answers
Hi everyone,
As part of my thesis, I'm looking for gfp-tagged bacteria or other type of fluorescent-tagged bacteria already constructed in order to use confocal microscopy. I've already checked on ATTC but I only found a gfp-tagged pseudomonas. Is there another place (other devices or a laboratory) wich provide that type of bacteria ?
Thank you for the consideration.
  • asked a question related to Confocal Microscopy
Question
5 answers
I am working with FFPE sections of mice ileum and am trying to image changes in the mitochondrial ROS production between a treatment and control group. I'm not sure if there are any stains or antibodies that can specifically target mtROS once the tissue is fixed and I have seen another researcher propose using MitoSox Red on tissue prior to fixation to get an assessment of mitochondrial ROS in the live animal. Has anyone else tried this successfully?
We also dabbled using a pimonidazole hypoxyprobe as a measure of total ROS, however the staining in the ileum and colon differ for physiological reasons. If anyone has some ideas as to how to better optimize this stain or an alternative method of detection that works well with ileal tissue, please let me know!
Relevant answer
Answer
Brian,
I would not recommend to use MitoSox with fixed tissue!
1. MitoSox is a membrane permeable weak acid. In live cells it distributes into the mitochondria because , as long as the mitos maintain their membrane potential, the mitochondrial matrix is the most alkaline place in the cell. Once MitoSox arrives in the matrix it gives off a proton. Now electrically charged it is no longer membrane permeable which effectively traps it inside the mitos. Thus, for proper localization it depends on the presence of membrane potentials which are missing in fixed tissue.
2. MitoSox becomes fluorescent after reaction with superoxide. The stain is meant to report superoxide as it is produced by the mitochondria, typically by activity of the electron transport chain (normal or pathologic). Fixed mitochondria are not metabolically active any more and therefore should not be able to produce superoxide any longer. Superoxide is diffusible, reactive (ROS - duh!) and therefore has a very short half-life. Thus, the superoxide that has been present at time of fixation will be long gone at the time you stain your fixed tissue slices.
Anything you find with MitoSox in fixed tissue will probably be caused by non-specific effects, such as autoxidation, and will be more confusing than useful.
I recommend to follow Alexandr's hints and concentrate on immunodetection of permanent ROS-damage to the tissue (protein, nucleic acids).
Good luck with your project!
Bernhard
  • asked a question related to Confocal Microscopy
Question
2 answers
Hi guys. I'm writing my paper for the first time.
. Representative images of glucose uptake were obtained of wavelengths at Excitation/Emission = 488/516 mm by using confocal microscopy
Is this sentence sound weird?
Or how can i correct this sentence ?
Thanks for nice answering .
Relevant answer
Answer
You may use the following sentences:
Confocal microscopy was used to take representative images of glucose uptake. These images were obtained by utilizing excitation/emission wavelengths of 488/516 mm.
  • asked a question related to Confocal Microscopy
Question
7 answers
Hi, I'm doing deconvolution to confocal images using Huygens Professional software, and I'm not sure whether I should decide the right signal-noise ratio (SNR) based on the image stack projected by summing the slices (SUM) or by projecting their maximum intensity (MIP). Despite the stack projection obtained by SUM looks better resolved than the original image, it seems to have more background. On the contrary, the projection obtained by MIP looks cleaner in terms of background, but some of the finest structures disappear compared to the original image (look at the attached images). The same is seen by comparing the intensity plots. Then, I wonder whether I should focus in change SNR to obtain better SUM or MIP projection.
Thanks a lot!
Relevant answer
Answer
There is also a nice online tool to calculate the Nyquist rate:
  • asked a question related to Confocal Microscopy
Question
2 answers
We have a Zeiss LSM 700 attached to an Axio Observer.Z1 microscope equipped with a T-PMT (photomultiplier for transmitted light). When I use the confocal microscope I often want a transmitted light (phase contrast, Ph3) image superimposed on the fluorescence light channel. This gives nice pictures as seen in the first attachment.
However at some point - using the same settings and same protocol - all images from the transmitted light channel has a wave pattern, this is a fault that I do not want in my images. See the second attachment. This problem persists and I have not been able to resolve the problem, do anyone know the cause of this? What I might have to adjust to fix it?
Relevant answer
Answer
Hi,
Is this a spinning disk confocal you are using? If so then try increasing the speed of the disk, because the pattern seems like it probably could be forming due to the spinning disk speed being slow. You can get upgradation on the speed.
  • asked a question related to Confocal Microscopy
Question
6 answers
Hi,
I'm having problems with the Recombinant Anti-MAP2 antibody from Abcam (Recombinant Anti-MAP2 antibody [EPR19691] (ab183830)) antibody on my frozen, 4%PFA perfused, mice brain tissue.
I was wondering if someone could elaborate better the note from the application part of the data sheet; Antigen retrieval: Heated citrate solution (10mM citrate PH 6.0 + 0.05% Tween-20).
To my knowledge AR is not used with frozen tissue, as it is to rough, and can damage the tissue, while also causing tissue detachment from the glass!?
Does someone have any experience with AR and frozen tissue?
I did try using 1/100 dilution, 0,2% triton X-100 and 4%PFA fixation as described in the images part of the datasheet: Immunohistochemical analysis of 4% paraformaldehyde-fixed, 0.2% Triton X-100 permeabilized frozen Mouse Cortex tissue labeling MAP2 with ab183830 at 1/100 dilution.
However I did not observed cytoplasmic staining of my neurons (image as uploaded for reference)!!
My protocol was the following:
4%PFA fixation, 15 min RT
3xPBS, 5min
10 min PBS+0.2% Triton x-100
Blocking solution (3%BSA, Without Triton x-100, 1h)
Anti-MAP2 1:100 dilution in 1%BSA (try both overnight and 2h RT)
3xPBS, 5min
AF-647 1:500, 30-45 min RT
3xPBS, 5 min
DAPI, 5 min
3x PBS, 5 min
dH2O, 5 min
Antifade and coverslip.
The antibody works excellent in IHC-P.
Thanks for the help,